1
|
Pedroza DA, Gao Y, Zhang XHF, Rosen JM. Leveraging preclinical models of metastatic breast cancer. Biochim Biophys Acta Rev Cancer 2024; 1879:189163. [PMID: 39084494 PMCID: PMC11390310 DOI: 10.1016/j.bbcan.2024.189163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/02/2024]
Abstract
Women that present to the clinic with established breast cancer metastases have limited treatment options. Yet, the majority of preclinical studies are actually not directed at developing treatment regimens for established metastatic disease. In this review we will discuss the current state of preclinical macro-metastatic breast cancer models, including, but not limited to syngeneic GEMM, PDX and xenografts. Challenges within these models which are often overlooked include fluorophore-immunogenic neoantigens, differences in experimental vs spontaneous metastasis and tumor heterogeneity. Furthermore, due to cell plasticity in the tumor immune microenvironment (TIME) of the metastatic landscape, the treatment efficacy of newly approved immune checkpoint blockade (ICB) may differ in metastatic sites as compared to primary localized tumors.
Collapse
Affiliation(s)
- Diego A Pedroza
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States of America; Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, United States of America; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States of America
| | - Yang Gao
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States of America; Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, United States of America; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States of America
| | - Xiang H-F Zhang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States of America; Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, United States of America; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States of America
| | - Jeffrey M Rosen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States of America; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States of America.
| |
Collapse
|
2
|
Bogatsa E, Lazaridis G, Stivanaki C, Timotheadou E. Neoadjuvant and Adjuvant Immunotherapy in Resectable NSCLC. Cancers (Basel) 2024; 16:1619. [PMID: 38730571 PMCID: PMC11083960 DOI: 10.3390/cancers16091619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 04/15/2024] [Accepted: 04/18/2024] [Indexed: 05/13/2024] Open
Abstract
Non-small cell lung cancer, even when diagnosed in early stages, has been linked with poor survival rates and distant recurrence patterns. Novel therapeutic approaches harnessing the immune system have been implemented in early stages, following the designated steps of advanced NSCLC treatment strategies. Immune-checkpoint inhibitor (ICI) regimens as monotherapy, combinational, or alongside chemotherapy have been intensely investigated as adjuvant, neoadjuvant, and, more recently, perioperative therapeutic strategies, representing pivotal milestones in the evolution of early lung cancer management while holding great potential for the future. The subject of current ongoing research is optimizing treatment outcomes for patient subsets with different needs and identifying biomarkers that could be predictive of response while translating the trials' endpoints to survival rates. The aim of this review is to discuss all current treatment options with the pros and cons of each, persistent challenges, and future perspectives on immunotherapy as illuminating the path to a new era for resectable NSCLC.
Collapse
Affiliation(s)
| | - George Lazaridis
- Department of Medical Oncology, Aristotle University of Thessaloniki, Papageorgiou Hospital, 56429 Thessaloniki, Greece; (E.B.); (E.T.)
| | | | | |
Collapse
|
3
|
Fiuza-Luces C, Valenzuela PL, Gálvez BG, Ramírez M, López-Soto A, Simpson RJ, Lucia A. The effect of physical exercise on anticancer immunity. Nat Rev Immunol 2024; 24:282-293. [PMID: 37794239 DOI: 10.1038/s41577-023-00943-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2023] [Indexed: 10/06/2023]
Abstract
Regular physical activity is associated with lower cancer incidence and mortality, as well as with a lower rate of tumour recurrence. The epidemiological evidence is supported by preclinical studies in animal models showing that regular exercise delays the progression of cancer, including highly aggressive malignancies. Although the mechanisms underlying the antitumorigenic effects of exercise remain to be defined, an improvement in cancer immunosurveillance is likely important, with different immune cell subtypes stimulated by exercise to infiltrate tumours. There is also evidence that immune cells from blood collected after an exercise bout could be used as adoptive cell therapy for cancer. In this Perspective, we address the importance of muscular activity for maintaining a healthy immune system and discuss the effects of a single bout of exercise (that is, 'acute' exercise) and those of 'regular' exercise (that is, repeated bouts) on anticancer immunity, including tumour infiltrates. We also address the postulated mechanisms and the clinical implications of this emerging area of research.
Collapse
Affiliation(s)
- Carmen Fiuza-Luces
- Physical Activity and Health Research Group ('PaHerg'), Research Institute of the Hospital 12 de Octubre ('imas12'), Madrid, Spain.
| | - Pedro L Valenzuela
- Physical Activity and Health Research Group ('PaHerg'), Research Institute of the Hospital 12 de Octubre ('imas12'), Madrid, Spain
- Systems Biology Department, Universidad de Alcalá, Alcalá de Henares, Spain
| | - Beatriz G Gálvez
- Physical Activity and Health Research Group ('PaHerg'), Research Institute of the Hospital 12 de Octubre ('imas12'), Madrid, Spain
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Universidad Complutense de Madrid, Madrid, Spain
| | - Manuel Ramírez
- Oncohematology Unit, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
- Biomedical Research Foundation, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
- La Princesa Institute of Heah, Madrid, Spain
| | - Alejandro López-Soto
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Oviedo, Oviedo, Spain.
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Asturias, Spain.
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Asturias, Spain.
| | - Richard J Simpson
- School of Nutritional Sciences and Wellness, The University of Arizona, Tucson, AZ, USA
- Department of Paediatrics, The University of Arizona, Tucson, AZ, USA
- Department of Immunobiology, The University of Arizona, Tucson, AZ, USA
| | - Alejandro Lucia
- CIBER of Frailty and Healthy Aging (CIBERFES), Madrid, Spain.
- Faculty of Sport Sciences, Universidad Europea, Madrid, Spain.
| |
Collapse
|
4
|
Houda I, Dickhoff C, Uyl-de Groot CA, Reguart N, Provencio M, Levy A, Dziadziuszko R, Pompili C, Di Maio M, Thomas M, Brunelli A, Popat S, Senan S, Bahce I. New systemic treatment paradigms in resectable non-small cell lung cancer and variations in patient access across Europe. THE LANCET REGIONAL HEALTH. EUROPE 2024; 38:100840. [PMID: 38476748 PMCID: PMC10928304 DOI: 10.1016/j.lanepe.2024.100840] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 01/07/2024] [Accepted: 01/08/2024] [Indexed: 03/14/2024]
Abstract
The treatment landscape of resectable early-stage non-small cell lung cancer (NSCLC) is set to change significantly due to encouraging results from randomized trials evaluating neoadjuvant and adjuvant immunotherapy, as well as adjuvant targeted therapy. As of January 2024, marketing authorization has been granted for four new indications in Europe, and regulatory approvals for other study regimens are expected. Because cost-effectiveness and reimbursement criteria for novel treatments often differ between European countries, access to emerging developments may lead to inequalities due to variations in recommended and available lung cancer care throughout Europe. This Series paper (i) highlights the clinical studies reshaping the treatment landscape in resectable early-stage NSCLC, (ii) compares and contrasts approaches taken by the European Medicines Agency (EMA) for drug approval to that taken by the United States Food and Drug Administration (FDA), and (iii) evaluates the differences in access to emerging treatments from an availability perspective across European countries.
Collapse
Affiliation(s)
- Ilias Houda
- Department of Pulmonary Medicine, Amsterdam UMC, Location VU Medical Center, Cancer Center Amsterdam, de Boelelaan 1117, 1081HV, Amsterdam, the Netherlands
| | - Chris Dickhoff
- Department of Cardiothoracic Surgery, Amsterdam UMC, Location VU Medical Center, Cancer Center Amsterdam, de Boelelaan 1117, 1081HV, Amsterdam, the Netherlands
| | - Carin A. Uyl-de Groot
- Erasmus School of Health Policy & Management/Institute for Medical Technology Assessment, Erasmus University Rotterdam, P.O. Box 1738, 3000 DR, Rotterdam, the Netherlands
| | - Noemi Reguart
- Department of Medical Oncology, Hospital Clínic de Barcelona, C. de Villarroel, 170, 08036, Barcelona, Spain
| | - Mariano Provencio
- Department of Medical Oncology, Hospital Universitario Puerta De Hierro, C. Joaquín Rodrigo, 1, 28222, Majadahonda, Madrid, Spain
| | - Antonin Levy
- Department of Radiation Oncology, International Center for Thoracic Cancers (CICT), Université Paris Saclay, Gustave Roussy, 114 Rue Edouard Vaillant, 94805, Villejuif, France
| | - Rafal Dziadziuszko
- Department of Oncology and Radiotherapy, Faculty of Medicine, Medical University of Gdańsk, 80-210, Gdańsk, Poland
| | - Cecilia Pompili
- Department of Thoracic Surgery, University and Hospital Trust – Ospedale Borgo Trento, P.Le A. Stefani, 1, 37126, Verona, Italy
| | - Massimo Di Maio
- Department of Oncology, University of Turin, Medical Oncology 1U, AOU Città della Salute e della Scienza di Torino, 10126, Turin, Italy
| | - Michael Thomas
- Department of Thoracic Oncology, Thoraxklinik, Heidelberg University Hospital and National Center for Tumor Diseases (NCT), NCT Heidelberg, a Partnership Between DKFZ and Heidelberg University Hospital, Im Neuenheimer Feld 672, 69120, Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research, Heidelberg, Germany
| | - Alessandro Brunelli
- Department of Thoracic Surgery, St. James's University Hospital, Beckett Street, LS9 7TF, Leeds, United Kingdom
| | - Sanjay Popat
- Lung Unit, The Royal Marsden NHS Foundation Trust, Fulham Road, London, SW3 6JJ, UK
| | - Suresh Senan
- Department of Radiation Oncology, Amsterdam UMC, Location VU Medical Center, Cancer Center Amsterdam, de Boelelaan 1117, 1081HV, Amsterdam, the Netherlands
| | - Idris Bahce
- Department of Pulmonary Medicine, Amsterdam UMC, Location VU Medical Center, Cancer Center Amsterdam, de Boelelaan 1117, 1081HV, Amsterdam, the Netherlands
| |
Collapse
|
5
|
Bartos LM, Kirchleitner SV, Kolabas ZI, Quach S, Beck A, Lorenz J, Blobner J, Mueller SA, Ulukaya S, Hoeher L, Horvath I, Wind-Mark K, Holzgreve A, Ruf VC, Gold L, Kunze LH, Kunte ST, Beumers P, Park HE, Antons M, Zatcepin A, Briel N, Hoermann L, Schaefer R, Messerer D, Bartenstein P, Riemenschneider MJ, Lindner S, Ziegler S, Herms J, Lichtenthaler SF, Ertürk A, Tonn JC, von Baumgarten L, Albert NL, Brendel M. Deciphering sources of PET signals in the tumor microenvironment of glioblastoma at cellular resolution. SCIENCE ADVANCES 2023; 9:eadi8986. [PMID: 37889970 PMCID: PMC10610915 DOI: 10.1126/sciadv.adi8986] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023]
Abstract
Various cellular sources hamper interpretation of positron emission tomography (PET) biomarkers in the tumor microenvironment (TME). We developed an approach of immunomagnetic cell sorting after in vivo radiotracer injection (scRadiotracing) with three-dimensional (3D) histology to dissect the cellular allocation of PET signals in the TME. In mice with implanted glioblastoma, translocator protein (TSPO) radiotracer uptake per tumor cell was higher compared to tumor-associated microglia/macrophages (TAMs), validated by protein levels. Translation of in vitro scRadiotracing to patients with glioma immediately after tumor resection confirmed higher single-cell TSPO tracer uptake of tumor cells compared to immune cells. Across species, cellular radiotracer uptake explained the heterogeneity of individual TSPO-PET signals. In consideration of cellular tracer uptake and cell type abundance, tumor cells were the main contributor to TSPO enrichment in glioblastoma; however, proteomics identified potential PET targets highly specific for TAMs. Combining cellular tracer uptake measures with 3D histology facilitates precise allocation of PET signals and serves to validate emerging novel TAM-specific radioligands.
Collapse
Affiliation(s)
- Laura M. Bartos
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | | | - Zeynep Ilgin Kolabas
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
- Institute for Stroke and Dementia Research (ISD), University Hospital of Munich, LMU Munich, Munich, Germany
- Graduate School of Systemic Neurosciences (GSN), Munich, Germany
| | - Stefanie Quach
- Department of Neurosurgery, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Alexander Beck
- Center for Neuropathology and Prion Research, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Julia Lorenz
- Department of Neuropathology, Regensburg University Hospital, Regensburg, Germany
| | - Jens Blobner
- Department of Neurosurgery, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Stephan A. Mueller
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- DZNE–German Center for Neurodegenerative Diseases, Munich, Germany
| | - Selin Ulukaya
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
- Faculty of Biology, Master of Science Program in Molecular and Cellular Biology, Ludwig-Maximilians-Universität München, Planegg, Germany
| | - Luciano Hoeher
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
| | - Izabela Horvath
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
- School of Computation, Information and Technology (CIT), TUM, Boltzmannstr. 3, 85748 Garching, Germany
| | - Karin Wind-Mark
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Adrien Holzgreve
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Viktoria C. Ruf
- Center for Neuropathology and Prion Research, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Lukas Gold
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Lea H. Kunze
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Sebastian T. Kunte
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Philipp Beumers
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Ha Eun Park
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Melissa Antons
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Artem Zatcepin
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- DZNE–German Center for Neurodegenerative Diseases, Munich, Germany
| | - Nils Briel
- Center for Neuropathology and Prion Research, Faculty of Medicine, LMU Munich, Munich, Germany
- DZNE–German Center for Neurodegenerative Diseases, Munich, Germany
| | - Leonie Hoermann
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Rebecca Schaefer
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Denise Messerer
- Department of Cardiology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | | | - Simon Lindner
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Sibylle Ziegler
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Jochen Herms
- Center for Neuropathology and Prion Research, Faculty of Medicine, LMU Munich, Munich, Germany
- DZNE–German Center for Neurodegenerative Diseases, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Stefan F. Lichtenthaler
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- DZNE–German Center for Neurodegenerative Diseases, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Ali Ertürk
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
- Institute for Stroke and Dementia Research (ISD), University Hospital of Munich, LMU Munich, Munich, Germany
- Graduate School of Systemic Neurosciences (GSN), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Joerg C. Tonn
- Department of Neurosurgery, University Hospital of Munich, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Louisa von Baumgarten
- Department of Neurosurgery, University Hospital of Munich, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nathalie L. Albert
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- DZNE–German Center for Neurodegenerative Diseases, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
6
|
Mountzios G, Remon J, Hendriks LEL, García-Campelo R, Rolfo C, Van Schil P, Forde PM, Besse B, Subbiah V, Reck M, Soria JC, Peters S. Immune-checkpoint inhibition for resectable non-small-cell lung cancer - opportunities and challenges. Nat Rev Clin Oncol 2023; 20:664-677. [PMID: 37488229 DOI: 10.1038/s41571-023-00794-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/19/2023] [Indexed: 07/26/2023]
Abstract
Therapeutic strategies harnessing the immune system to eliminate tumour cells have been successfully used for several cancer types, including in patients with advanced-stage non-small-cell lung cancer (NSCLC). In these patients, immune-checkpoint inhibitors (ICIs) can provide durable responses and improve overall survival either as monotherapy, or combined with chemotherapy or other immunotherapeutic agents. However, the implementation of ICIs in early stage NSCLC has been hampered by the continuous struggle to develop robust end points to assess their efficacy in this setting, especially those enabling a fast and reproducible evaluation of the clinical activity of neoadjuvant strategies. Several trials are testing ICIs, alone or in combination with chemotherapy, in early stage NSCLC as an adjuvant, neoadjuvant or perioperative approach. As a novelty, most trials in the neoadjuvant setting have adopted pathological response as a primary end point. ICIs have been approved for use in the neoadjuvant and adjuvant settings on the basis of event-free survival and disease-free survival benefit, respectively; however, the correlation of these end points with overall survival remains unclear in these settings. Unresolved challenges for the optimal use of ICIs with curative intent include concerns about their applicability in daily clinical practice and about improving patient selection based on predictive biomarkers or assessment of pathological response and minimal residual disease. In this Review, we discuss the rationale, available strategies and current trial landscape for the implementation of ICIs in patients with resectable NSCLC, and we further elaborate on future approaches to optimize their clinical benefit.
Collapse
Affiliation(s)
- Giannis Mountzios
- Fourth Department of Medical Oncology and Clinical Trials Unit, Henry Dunant Hospital Center, Athens, Greece.
| | - Jordi Remon
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | - Lizza E L Hendriks
- Department of Respiratory Medicine, Maastricht University Medical Centre, GROW School for Oncology and Reproduction, Maastricht, Netherlands
| | | | - Christian Rolfo
- Center for Thoracic Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Paul Van Schil
- Department of Thoracic and Vascular Surgery, University Hospital of Antwerp, Antwerp, Belgium
| | - Patrick M Forde
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Benjamin Besse
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
- Department of Cancer Medicine, Université Paris-Saclay, Orsay, France
| | - Vivek Subbiah
- Department of Cancer Medicine, Sarah Cannon Research Institute, Nashville, TN, USA
| | - Martin Reck
- Department of Thoracic Oncology, Airway Research Center North, German Center of Lung Research, Lung Clinic, Grosshansdorf, Germany
| | | | - Solange Peters
- Oncology Department, CHUV, Lausanne University, Lausanne, Switzerland
| |
Collapse
|
7
|
Berckmans Y, Ceusters J, Vankerckhoven A, Wouters R, Riva M, Coosemans A. Preclinical studies performed in appropriate models could help identify optimal timing of combined chemotherapy and immunotherapy. Front Immunol 2023; 14:1236965. [PMID: 37744323 PMCID: PMC10512939 DOI: 10.3389/fimmu.2023.1236965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/23/2023] [Indexed: 09/26/2023] Open
Abstract
Immune checkpoint inhibitors (ICI) have been revolutionary in the field of cancer therapy. However, their success is limited to specific indications and cancer types. Recently, the combination treatment of ICI and chemotherapy has gained more attention to overcome this limitation. Unfortunately, many clinical trials testing these combinations have provided limited success. This can partly be attributed to an inadequate choice of preclinical models and the lack of scientific rationale to select the most effective immune-oncological combination. In this review, we have analyzed the existing preclinical evidence on this topic, which is only limitedly available. Furthermore, this preclinical data indicates that besides the selection of a specific drug and dose, also the sequence or order of the combination treatment influences the study outcome. Therefore, we conclude that the success of clinical combination trials could be enhanced by improving the preclinical set up, in order to identify the optimal treatment combination and schedule to enhance the anti-tumor immunity.
Collapse
Affiliation(s)
- Yani Berckmans
- Department of Oncology, Laboratory of Tumor Immunology and Immunotherapy, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Jolien Ceusters
- Department of Oncology, Laboratory of Tumor Immunology and Immunotherapy, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Ann Vankerckhoven
- Department of Oncology, Laboratory of Tumor Immunology and Immunotherapy, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Roxanne Wouters
- Department of Oncology, Laboratory of Tumor Immunology and Immunotherapy, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
- Oncoinvent AS, Oslo, Norway
| | - Matteo Riva
- Department of Oncology, Laboratory of Tumor Immunology and Immunotherapy, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
- Department of Neurosurgery, Centre Hospitalier Universitaire (CHU) UCLouvain Namur, University Hospital of Godinne, Yvoir, Belgium
| | - An Coosemans
- Department of Oncology, Laboratory of Tumor Immunology and Immunotherapy, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| |
Collapse
|
8
|
Zhang Z, Wang B, Tan B. Advances in the Mechanism of Action of Neutrophil Extracellular Traps in Gastrointestinal Tumors: A Review. Int J Gen Med 2023; 16:2783-2789. [PMID: 37408845 PMCID: PMC10319275 DOI: 10.2147/ijgm.s419542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 06/23/2023] [Indexed: 07/07/2023] Open
Abstract
Introduction Neutrophils are important immune cells in the body, extremely abundant, phagocytic and bactericidal, and usually involved in the defense against infectious diseases as immune become. However, a new reticulum structure has been discovered: neutrophil extracellular traps (NETs), which consists of various components such as DNA and proteins, etc. Current studies have found that NETs are closely associated with various diseases such as immune diseases, inflammation and tumors, and the study of the development and metastasis of gastrointestinal tumors has become a recent research hotspot. The clinical significance of NETs has been gradually highlighted, especially in the area of immunosuppression. Methods We reviewed a large amount of relevant literature, summarized the latest detection methods of NETs, explored the mechanism of NETs in gastrointestinal tumors and summarized the latest hotspot directions. Results NETs are involved in the development of gastrointestinal tumors, and are closely related to the proliferation and metastasis of gastrointestinal tumors. Higher levels of NETs are associated with poor prognosis of gastrointestinal tumors, promote local growth of tumors through various pathways, participate in tumor-related systemic injury, and promote tumor growth and metastasis by enhancing the mitochondrial function of tumor cells and awakening dormant tumor cells. Discussion NETs are highly expressed in tumors, and tumors and their microenvironment can promote the production of NETs, providing new ideas for the clinical diagnosis and treatment of gastrointestinal tumors. In this paper, we describe the basic information about NETs, explore the research mechanisms related to NETs in gastrointestinal tumors, and prospectively explore the clinical potential of hotspots and inhibitors related to NETs for gastrointestinal tumors, in order to provide new ideas and targets for the diagnosis and treatment of gastrointestinal tumors.
Collapse
Affiliation(s)
- Zaibo Zhang
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Bingyu Wang
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Bibo Tan
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| |
Collapse
|
9
|
Baci D, Cekani E, Imperatori A, Ribatti D, Mortara L. Host-Related Factors as Targetable Drivers of Immunotherapy Response in Non-Small Cell Lung Cancer Patients. Front Immunol 2022; 13:914890. [PMID: 35874749 PMCID: PMC9298844 DOI: 10.3389/fimmu.2022.914890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 05/13/2022] [Indexed: 11/13/2022] Open
Abstract
Despite some significant therapeutic breakthroughs leading to immunotherapy, a high percentage of patients with non-small cell lung cancer (NSCLC) do not respond to treatment on relapse, thus experiencing poor prognosis and survival. The unsatisfying results could be related to the features of the tumor immune microenvironment and the dynamic interactions between a tumor and immune infiltrate. Host-tumor interactions strongly influence the course of disease and response to therapies. Thus, targeting host-associated factors by restoring their physiologic functions altered by the presence of a tumor represents a new therapeutic approach to control tumor development and progression. In NSCLC, the immunogenic tumor balance is shifted negatively toward immunosuppression due to the release of inhibitory factors as well as the presence of immunosuppressive cells. Among these cells, there are myeloid-derived suppressor cells, regulatory T cells that can generate a tumor-permissive milieu by reprogramming the cells of the hosts such as tumor-associated macrophages, tumor-associated neutrophils, natural killer cells, dendritic cells, and mast cells that acquire tumor-supporting phenotypes and functions. This review highlights the current knowledge of the involvement of host-related factors, including innate and adaptive immunity in orchestrating the tumor cell fate and the primary resistance mechanisms to immunotherapy in NSCLC. Finally, we discuss combinational therapeutic strategies targeting different aspects of the tumor immune microenvironment (TIME) to prime the host response. Further research dissecting the characteristics and dynamic interactions within the interface host-tumor is necessary to improve a patient fitness immune response and provide answers regarding the immunotherapy efficacy, with the aim to develop more successful treatments for NSCLC.
Collapse
Affiliation(s)
- Denisa Baci
- Molecular Cardiology Laboratory, IRCCS-Policlinico San Donato, San Donato Milanese, Milan, Italy.,Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Elona Cekani
- Medical Oncology Clinic, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Andrea Imperatori
- Center for Thoracic Surgery, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Aldo Moro Medical School, Bari, Italy
| | - Lorenzo Mortara
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| |
Collapse
|
10
|
Pathania AS, Prathipati P, Murakonda SP, Murakonda AB, Srivastava A, Avadhesh A, Byrareddy SN, Coulter DW, Gupta SC, Challagundla KB. Immune checkpoint molecules in neuroblastoma: A clinical perspective. Semin Cancer Biol 2022; 86:247-258. [PMID: 35787940 DOI: 10.1016/j.semcancer.2022.06.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 06/21/2022] [Accepted: 06/28/2022] [Indexed: 10/31/2022]
Abstract
High-risk neuroblastoma (NB) is challenging to treat with 5-year long-term survival in patients remaining below 50% and low chances of survival after tumor relapse or recurrence. Different strategies are being tested or under evaluation to destroy resistant tumors and improve survival outcomes in NB patients. Immunotherapy, which uses certain parts of a person's immune system to recognize or kill tumor cells, effectively improves patient outcomes in several types of cancer, including NB. One of the immunotherapy strategies is to block immune checkpoint signaling in tumors to increase tumor immunogenicity and anti-tumor immunity. Immune checkpoint proteins put brakes on immune cell functions to regulate immune activation, but this activity is exploited in tumors to evade immune surveillance and attack. Immune checkpoint proteins play an essential role in NB biology and immune escape mechanisms, which makes these tumors immunologically cold. Therapeutic strategies to block immune checkpoint signaling have shown promising outcomes in NB but only in a subset of patients. However, combining immune checkpoint blockade with other therapies, including conjugated antibody-based immunotherapy, radioimmunotherapy, tumor vaccines, or cellular therapies like modified T or natural killer (NK) cells, has shown encouraging results in enhancing anti-tumor immunity in the preclinical setting. An analysis of publicly available dataset using computational tools has unraveled the complexity of multiple cancer including NB. This review comprehensively summarizes the current information on immune checkpoint molecules, their biology, role in immune suppression and tumor development, and novel therapeutic approaches combining immune checkpoint inhibitors with other therapies to combat high-risk NB.
Collapse
Affiliation(s)
- Anup S Pathania
- Department of Biochemistry and Molecular Biology & The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Philip Prathipati
- Laboratory of Bioinformatics, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki City, Osaka 567-0085, Japan
| | - Swati P Murakonda
- Sri Rajiv Gandhi College of Dental Sciences & Hospital, Bengaluru, Karnataka 560032, India
| | - Ajay B Murakonda
- Sree Sai Dental College & Research Institute, Srikakulam, Andhra Pradesh 532001, India
| | - Ankit Srivastava
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Avadhesh Avadhesh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Siddappa N Byrareddy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Don W Coulter
- Department of Pediatrics, Division of Hematology/Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Subash C Gupta
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India; Department of Biochemistry, All India Institute of Medical Sciences, Guwahati, Assam, India.
| | - Kishore B Challagundla
- Department of Biochemistry and Molecular Biology & The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; The Child Health Research Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
11
|
Cardeña-Gutiérrez A, López Barahona M. Predictive Biomarkers of Severe Immune-Related Adverse Events With Immune Checkpoint Inhibitors: Prevention, Underlying Causes, Intensity, and Consequences. Front Med (Lausanne) 2022; 9:908752. [PMID: 35774996 PMCID: PMC9237384 DOI: 10.3389/fmed.2022.908752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/11/2022] [Indexed: 11/13/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have dramatically transformed oncology by prolonging overall survival and yielding better patient tolerance compared to other chemotherapeutic agents. However, numerous questions remain unanswered about the toxicity profile of ICIs, its relationship with the treatment response, and causes underlying the excellent treatment response in some patients, while recalcitrance in others. Research groups have continued to seek biomarkers that may permit the identification of treatment responders and predict toxicity to facilitate cessation of immunotherapy before the development of severe toxicity. However, some studies have found associations between serious adverse events and longer survivorship. The research question entailed determining whether a biomarker is needed to predict severe immune-related adverse events prior to their development or whether providing early treatment for toxicity would inhibit the immune system from attaining a long-lasting anti-tumor effect. Therefore, this review conducted an in-depth analysis into the molecular basis of these observations.
Collapse
Affiliation(s)
- Ana Cardeña-Gutiérrez
- MedicalOncologyDepartment, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | | |
Collapse
|
12
|
Patient-Derived Organoids of Colorectal Cancer: A Useful Tool for Personalized Medicine. J Pers Med 2022; 12:jpm12050695. [PMID: 35629118 PMCID: PMC9147270 DOI: 10.3390/jpm12050695] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/22/2022] [Accepted: 04/24/2022] [Indexed: 11/18/2022] Open
Abstract
Colorectal cancer is one of the most important malignancies worldwide, with high incidence and mortality rates. Several studies have been conducted using two-dimensional cultured cell lines; however, these cells do not represent a study model of patient tumors very well. In recent years, advancements in three-dimensional culture methods have facilitated the establishment of patient-derived organoids, which have become indispensable for molecular biology-related studies of colorectal cancer. Patient-derived organoids are useful in both basic science and clinical practice; they can help predict the sensitivity of patients with cancer to chemotherapy and radiotherapy and provide the right treatment to the right patient. Regarding precision medicine, combining gene panel testing and organoid-based screening can increase the effectiveness of medical care. In this study, we review the development of three-dimensional culture methods and present the most recent information on the clinical application of patient-derived organoids. Moreover, we discuss the problems and future prospects of organoid-based personalized medicine.
Collapse
|
13
|
Hendriks LE, Remon J, Reck M. Chemotherapy + PD-1/PD-L1 Blockade Should Not Be the Preferred Option in the Neoadjuvant Therapy of NSCLC. J Thorac Oncol 2022; 17:499-502. [DOI: 10.1016/j.jtho.2021.11.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 11/19/2021] [Accepted: 11/21/2021] [Indexed: 10/18/2022]
|
14
|
Cheng KH, Contreras GP, Yeh TY. Potential Role of Neutrophil Extracellular Traps in Cardio-Oncology. Int J Mol Sci 2022; 23:ijms23073573. [PMID: 35408933 PMCID: PMC8998890 DOI: 10.3390/ijms23073573] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/14/2022] [Accepted: 03/23/2022] [Indexed: 12/18/2022] Open
Abstract
Cardiovascular toxicity has emerged as the leading cause of death in patients undergoing cancer treatment. Thus, cardio-oncology (CO) care must also focus on the prevention and management of related cardiovascular (CV) complications caused by cancer therapy. Neutrophil extracellular traps (NETs)—entities with released DNA, proteases, proinflammatory and prooxidative substances from blasted neutrophils—play an important role in cancer proliferation, propagation metastasis, and incident CV events (acute coronary syndrome, thromboembolic events, and heart failure). Although NETs have been shown to be involved in cancer progression and incident CV events, little is known about their relationship with cardio-oncology, especially on cancer treatment-related cardiovascular toxicity (CTRCT). This review aims to explore the evidence of the impact of NETs on cancer, CV events, and CTRCT, and the possible solutions based on the mechanism of NETs activation and NETs released toxic substances.
Collapse
Affiliation(s)
- Kai-Hung Cheng
- Division of Cardiology, Department of Internal Medicine, E-Da Cancer Hospital, Kaohsiung 82445, Taiwan;
- College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan
| | - Gregory P. Contreras
- Auxergen Inc., Columbus Center, 701 East Pratt Street, Baltimore, MD 21202, USA;
| | - Ting-Yu Yeh
- Auxergen Inc., Columbus Center, 701 East Pratt Street, Baltimore, MD 21202, USA;
- Correspondence:
| |
Collapse
|
15
|
Mayca Pozo F, Geng X, Tamagno I, Jackson MW, Heimsath EG, Hammer JA, Cheney RE, Zhang Y. MYO10 drives genomic instability and inflammation in cancer. SCIENCE ADVANCES 2021; 7:eabg6908. [PMID: 34524844 PMCID: PMC8443186 DOI: 10.1126/sciadv.abg6908] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 07/26/2021] [Indexed: 05/29/2023]
Abstract
Genomic instability is a hallmark of human cancer; yet the underlying mechanisms remain poorly understood. Here, we report that the cytoplasmic unconventional Myosin X (MYO10) regulates genome stability, through which it mediates inflammation in cancer. MYO10 is an unstable protein that undergoes ubiquitin-conjugating enzyme H7 (UbcH7)/β-transducin repeat containing protein 1 (β-TrCP1)–dependent degradation. MYO10 is upregulated in both human and mouse tumors and its expression level predisposes tumor progression and response to immune therapy. Overexpressing MYO10 increased genomic instability, elevated the cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes (STING)–dependent inflammatory response, and accelerated tumor growth in mice. Conversely, depletion of MYO10 ameliorated genomic instability and reduced the inflammation signaling. Further, inhibiting inflammation or disrupting Myo10 significantly suppressed the growth of both human and mouse breast tumors in mice. Our data suggest that MYO10 promotes tumor progression through inducing genomic instability, which, in turn, creates an immunogenic environment for immune checkpoint blockades.
Collapse
Affiliation(s)
- Franklin Mayca Pozo
- Department of Pharmacology, Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Xinran Geng
- Department of Pharmacology, Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Ilaria Tamagno
- Department of Pathology, Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Mark W. Jackson
- Department of Pathology, Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Ernest G. Heimsath
- Department of Cell Biology and Physiology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - John A. Hammer
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, Bethesda, MD 20892, USA
| | - Richard E. Cheney
- Department of Cell Biology and Physiology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Youwei Zhang
- Department of Pharmacology, Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
16
|
Hong HK, Yun NH, Jeong YL, Park J, Doh J, Lee WY, Cho YB. Establishment of patient-derived organotypic tumor spheroid models for tumor microenvironment modeling. Cancer Med 2021; 10:5589-5598. [PMID: 34240815 PMCID: PMC8366099 DOI: 10.1002/cam4.4114] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/30/2021] [Accepted: 06/10/2021] [Indexed: 12/26/2022] Open
Abstract
Patient‐derived cancer models that reconstitute the characteristics of the tumor microenvironment may facilitate efforts in precision immune‐oncology and the discovery of effective anticancer therapies. Organoids that have recently emerged as robust preclinical models typically contain tumor epithelial cells and lack the native tumor immune microenvironment. A patient‐derived organotypic tumor spheroid (PDOTS) is a novel and innovative ex vivo system that retains key features of the native tumor immune microenvironment. Here, we established and characterized a series of colorectal cancer PDOTS models for use as a preclinical platform for testing effective immunotherapy and its combinations with other drugs. Partially dissociated (> 100 μm in diameter) tumor tissues were embedded in Matrigel‐containing organoid media and subsequently formed into organoid structures within 3 to 7 days of culture. The success rate of growing PDOTS from fresh tissues was ~86%. Morphological analysis showed that the PDOTSs varied in size and structure. Immunofluorescence and flow cytometry analysis revealed that the PDOTSs retained autologous tumor‐infiltrating lymphoid cells and tumor‐infiltrating lymphoid cells were continually decreased through serial passages. Notably, PDOTSs from tumors from a high‐level microsatellite instability‐harboring patient were sensitive to anti‐PD‐1 or anti‐PD‐L1 antibodies. Our results demonstrate that the PDOTS model in which the tumor immune microenvironment is preserved may represent an advantageous ex vivo system to develop effective immune therapeutics.
Collapse
Affiliation(s)
- Hye Kyung Hong
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
| | - Nak Hyeon Yun
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
| | - Ye-Lin Jeong
- Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
| | - Jeehun Park
- Research Institute of Advanced Materials, Seoul National University, Seoul, Korea
| | - Junsang Doh
- Research Institute of Advanced Materials, Seoul National University, Seoul, Korea.,Department of Materials Science and Engineering, Seoul National University, Seoul, Korea
| | - Woo Yong Lee
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
| | - Yong Beom Cho
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea.,Department of Biopharmaceutical Convergence, Sunkyunkwan University, Seoul, Korea
| |
Collapse
|
17
|
Anderson R, Blidner AG, Rapoport BL. Frontiers in Pharmacology: Review Manuscript Targeting of the Neutrophil as an Adjunctive Strategy in Non-Small Cell Lung Cancer. Front Pharmacol 2021; 12:676399. [PMID: 34168563 PMCID: PMC8218630 DOI: 10.3389/fphar.2021.676399] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/17/2021] [Indexed: 01/14/2023] Open
Abstract
Lung cancer remains the leading cause of cancer mortality in the United States, with non-small cell lung cancer (NSCLC) accounting for around 85% of cases. Of particular concern is the poor responsiveness of this malignancy to therapy, resulting in a very low 5-year survival rate (17.4%) and a prominent tendency to progress to metastatic disease. A number of very recent studies, both pre-clinical and clinical, have implicated the neutrophil in both the pathogenesis and unsatisfactory response to therapy of NSCLC. In this context, movement of neutrophils into the tumor microenvironment (TME) is a common feature of NSCLC. Indeed neutrophils are the dominant type of immune cell in the NSCLC TME, creating a highly immunosuppressive milieu that is not only conducive to tumor growth and spread, but also represents a significant obstacle to the success of anti-tumor therapy, especially novel immunotherapies. The clinically relevant adverse impact of a neutrophil predominance both systemically and in the TME of patients with NSCLC is underscored by the negative prognostic value of both a persistent neutrophilia and, in particular, a high (≥5) neutrophil:lymphocyte ratio. On a more positive note, however, recognition of the involvement of the neutrophil in both the pathophysiology of NSCLC and treatment failure has enabled identification of neutrophil-targeted strategies that have the potential to serve as adjuncts to standard anti-cancer therapies, including immunotherapy. These strategies together with a consideration of the immunosuppressive, pro-tumorigenic properties of the neutrophil represent the major thrusts of this review.
Collapse
Affiliation(s)
- Ronald Anderson
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Ada Gabriela Blidner
- Laboratory of Immunopathology, Institute of Biology and Experimental Medicine CONICET, Buenos Aires, Argentina
| | - Bernardo Leon Rapoport
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa.,The Medical Oncology Centre of Rosebank, Johannesburg, South Africa
| |
Collapse
|