1
|
Ma X, Fernández FM. Advances in mass spectrometry imaging for spatial cancer metabolomics. MASS SPECTROMETRY REVIEWS 2024; 43:235-268. [PMID: 36065601 PMCID: PMC9986357 DOI: 10.1002/mas.21804] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/02/2022] [Accepted: 08/02/2022] [Indexed: 05/09/2023]
Abstract
Mass spectrometry (MS) has become a central technique in cancer research. The ability to analyze various types of biomolecules in complex biological matrices makes it well suited for understanding biochemical alterations associated with disease progression. Different biological samples, including serum, urine, saliva, and tissues have been successfully analyzed using mass spectrometry. In particular, spatial metabolomics using MS imaging (MSI) allows the direct visualization of metabolite distributions in tissues, thus enabling in-depth understanding of cancer-associated biochemical changes within specific structures. In recent years, MSI studies have been increasingly used to uncover metabolic reprogramming associated with cancer development, enabling the discovery of key biomarkers with potential for cancer diagnostics. In this review, we aim to cover the basic principles of MSI experiments for the nonspecialists, including fundamentals, the sample preparation process, the evolution of the mass spectrometry techniques used, and data analysis strategies. We also review MSI advances associated with cancer research in the last 5 years, including spatial lipidomics and glycomics, the adoption of three-dimensional and multimodal imaging MSI approaches, and the implementation of artificial intelligence/machine learning in MSI-based cancer studies. The adoption of MSI in clinical research and for single-cell metabolomics is also discussed. Spatially resolved studies on other small molecule metabolites such as amino acids, polyamines, and nucleotides/nucleosides will not be discussed in the context.
Collapse
Affiliation(s)
- Xin Ma
- School of Chemistry and Biochemistry and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Facundo M Fernández
- School of Chemistry and Biochemistry and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
| |
Collapse
|
2
|
Arya R, Jit BP, Kumar V, Kim JJ. Exploring the Potential of Exosomes as Biomarkers in Tuberculosis and Other Diseases. Int J Mol Sci 2024; 25:2885. [PMID: 38474139 DOI: 10.3390/ijms25052885] [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: 01/01/2024] [Revised: 02/23/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
Tuberculosis (TB) is a major cause of morbidity and mortality and remains an important public health issue in developing countries worldwide. The existing methods and techniques available for the diagnosis of TB are based on combinations of laboratory (chemical and biological), radiological, and clinical tests. These methods are sophisticated and laborious and have limitations in terms of sensitivity, specificity, and accuracy. Clinical settings need improved diagnostic biomarkers to accurately detect biological changes due to pathogen invasion and pharmacological responses. Exosomes are membrane-bound vesicles and mediators of intercellular signaling processes that play a significant role in the pathogenesis of various diseases, such as tuberculosis, and can act as promising biomarkers for the monitoring of TB infection. Compared to conventional biomarkers, exosome-derived biomarkers are advantageous because they are easier to detect in different biofluids, are more sensitive and specific, and may be useful in tracking patients' reactions to therapy. This review provides insights into the types of biomarkers, methods of exosome isolation, and roles of the cargo (proteins) present in exosomes isolated from patients through omics studies, such as proteomics. These findings will aid in developing new prognostic and diagnostic biomarkers and could lead to the identification of new therapeutic targets in the clinical setting.
Collapse
Affiliation(s)
- Rakesh Arya
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Bimal Prasad Jit
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Vijay Kumar
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jong Joo Kim
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea
| |
Collapse
|
3
|
Pérez-Gimeno G, Seral-Cortes M, Sabroso-Lasa S, Esteban LM, Lurbe E, Béghin L, Gottrand F, Meirhaeghe A, Muntaner M, Kafatos A, Molnár D, Leclercq C, Widhalm K, Kersting M, Nova E, Salazar-Tortosa DF, Gonzalez-Gross M, Breidenassel C, Sinningen K, De Ruyter T, Labayen I, Rupérez AI, Bueno-Lozano G, Moreno LA. Development of a genetic risk score to predict the risk of hypertension in European adolescents from the HELENA study. Front Cardiovasc Med 2023; 10:1118919. [PMID: 37324619 PMCID: PMC10267871 DOI: 10.3389/fcvm.2023.1118919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 05/11/2023] [Indexed: 06/17/2023] Open
Abstract
Introduction From genome wide association study (GWAS) a large number of single nucleotide polymorphisms (SNPs) have previously been associated with blood pressure (BP) levels. A combination of SNPs, forming a genetic risk score (GRS) could be considered as a useful genetic tool to identify individuals at risk of developing hypertension from early stages in life. Therefore, the aim of our study was to build a GRS being able to predict the genetic predisposition to hypertension (HTN) in European adolescents. Methods Data were extracted from the Healthy Lifestyle in Europe by Nutrition in Adolescence (HELENA) cross-sectional study. A total of 869 adolescents (53% female), aged 12.5-17.5, with complete genetic and BP information were included. The sample was divided into altered (≥130 mmHg for systolic and/or ≥80 mmHg for diastolic) or normal BP. Based on the literature, a total of 1.534 SNPs from 57 candidate genes related with BP were selected from the HELENA GWAS database. Results From 1,534 SNPs available, An initial screening of SNPs univariately associated with HTN (p < 0.10) was established, to finally obtain a number of 16 SNPs significantly associated with HTN (p < 0.05) in the multivariate model. The unweighted GRS (uGRS) and weighted GRS (wGRS) were estimated. To validate the GRSs, the area under the curve (AUC) was explored using ten-fold internal cross-validation for uGRS (0.802) and wGRS (0.777). Further covariates of interest were added to the analyses, obtaining a higher predictive ability (AUC values of uGRS: 0.879; wGRS: 0.881 for BMI z-score). Furthermore, the differences between AUCs obtained with and without the addition of covariates were statistically significant (p < 0.05). Conclusions Both GRSs, the uGRS and wGRS, could be useful to evaluate the predisposition to hypertension in European adolescents.
Collapse
Affiliation(s)
- Gloria Pérez-Gimeno
- Growth, Exercise, NUtrition and Development (GENUD), Research Group, Instituto Agroalimentario de Aragón (IA2), Instituto de Investigación Sanitaria Aragón (IIS Aragón), Universidad de Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
| | - Miguel Seral-Cortes
- Growth, Exercise, NUtrition and Development (GENUD), Research Group, Instituto Agroalimentario de Aragón (IA2), Instituto de Investigación Sanitaria Aragón (IIS Aragón), Universidad de Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
| | - Sergio Sabroso-Lasa
- Genetic and Molecular Epidemiology Group (GMEG), Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | | | - Empar Lurbe
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
- INCLIVA Biomedical Research Institute, Pediatric Department, Consorcio Hospital General, University of Valencia, Valencia, Spain
| | - Laurent Béghin
- Université Lille, Inserm, CHU Lille, INFINITE—Institute for Translational Research in Inflammation, Lille, France
| | - Frederic Gottrand
- Université Lille, Inserm, CHU Lille, INFINITE—Institute for Translational Research in Inflammation, Lille, France
| | - Aline Meirhaeghe
- Risk Factors and Molecular Determinants of Aging-Related Diseases (RID-AGE), Centre Hosp. Univ Lille, Institut Pasteur de Lille, Université de Lille, Lille, France
| | - Manon Muntaner
- Risk Factors and Molecular Determinants of Aging-Related Diseases (RID-AGE), Centre Hosp. Univ Lille, Institut Pasteur de Lille, Université de Lille, Lille, France
| | - Anthony Kafatos
- Department of Social Medicine, Preventive Medicine and Nutrition Clinic, University of Crete School of Medicine, Heraklion, Greece
| | - Dénes Molnár
- Department of Pediatrics, University of Pecs, Pecs, Hungary
| | - Catherine Leclercq
- INRAN, National Research Institute for Food and Nutrition, Food and Nutrition Research Centre-Council for Agricultural Research and Economics, Rome, Italy
| | - Kurt Widhalm
- Division of Clinical Nutrition and Prevention, Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Mathilde Kersting
- Departement of Nutrition—Human Nutrition, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Esther Nova
- Department of Metabolism and Nutrition, Institute of Food Science and Technology and Nutrition (ICTAN), CSIC, Madrid, Spain
| | - Diego F. Salazar-Tortosa
- Department of Ecology and Evolutionary Biology, University of Arizona, Tucson, AZ, United States
- PROFITH ‘PROmoting FITness and Health Through Physical Activity’ Research Group, Sport and Health University Research Institute (iMUDS), University of Granada, Granada, Spain
| | - Marcela Gonzalez-Gross
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
- ImFine Research Group, Department of Health and Human Performance, Facultad de Ciencias de la Actividad Física y del Deporte-INEF, Universidad Politécnica de Madrid, Madrid, Spain
| | - Christina Breidenassel
- Departement of Nutrition—Human Nutrition, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
- ImFine Research Group, Department of Health and Human Performance, Facultad de Ciencias de la Actividad Física y del Deporte-INEF, Universidad Politécnica de Madrid, Madrid, Spain
| | - Kathrin Sinningen
- Research Department of Child Nutrition, University Hospital of Pediatrics and Adolescent Medicine, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Thaïs De Ruyter
- Department of Public Health and Primary Care, Ghent University, Ghent, Belgium
| | - Idoia Labayen
- Department of Health Sciences, Institute for Innovation & Sustainable Food Chain Development, Public University of Navarra, Pamplona, Spain
| | - Azahara I. Rupérez
- Growth, Exercise, NUtrition and Development (GENUD), Research Group, Instituto Agroalimentario de Aragón (IA2), Instituto de Investigación Sanitaria Aragón (IIS Aragón), Universidad de Zaragoza, Zaragoza, Spain
| | - Gloria Bueno-Lozano
- Growth, Exercise, NUtrition and Development (GENUD), Research Group, Instituto Agroalimentario de Aragón (IA2), Instituto de Investigación Sanitaria Aragón (IIS Aragón), Universidad de Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
| | - Luis A. Moreno
- Growth, Exercise, NUtrition and Development (GENUD), Research Group, Instituto Agroalimentario de Aragón (IA2), Instituto de Investigación Sanitaria Aragón (IIS Aragón), Universidad de Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
4
|
Huber J, Longaker MT, Quarto N. Circulating and extracellular vesicle-derived microRNAs as biomarkers in bone-related diseases. Front Endocrinol (Lausanne) 2023; 14:1168898. [PMID: 37293498 PMCID: PMC10244776 DOI: 10.3389/fendo.2023.1168898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 03/31/2023] [Indexed: 06/10/2023] Open
Abstract
MicroRNAs (miRNA) are small non-coding RNA molecules that regulate posttranscriptional gene expression by repressing messengerRNA-targets. MiRNAs are abundant in many cell types and are secreted into extracellular fluids, protected from degradation by packaging in extracellular vesicles. These circulating miRNAs are easily accessible, disease-specific and sensitive to small changes, which makes them ideal biomarkers for diagnostic, prognostic, predictive or monitoring purposes. Specific miRNA signatures can be reflective of disease status and development or indicators of poor treatment response. This is especially important in malignant diseases, as the ease of accessibility of circulating miRNAs circumvents the need for invasive tissue biopsy. In osteogenesis, miRNAs can act either osteo-enhancing or osteo-repressing by targeting key transcription factors and signaling pathways. This review highlights the role of circulating and extracellular vesicle-derived miRNAs as biomarkers in bone-related diseases, with a specific focus on osteoporosis and osteosarcoma. To this end, a comprehensive literature search has been performed. The first part of the review discusses the history and biology of miRNAs, followed by a description of different types of biomarkers and an update of the current knowledge of miRNAs as biomarkers in bone related diseases. Finally, limitations of miRNAs biomarker research and future perspectives will be presented.
Collapse
Affiliation(s)
- Julika Huber
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Department of Plastic Surgery, University Hospital Bergmannsheil Bochum, Bochum, Germany
| | - Michael T. Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Natalina Quarto
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
5
|
Levati L, Bassi C, Mastroeni S, Lupini L, Antonini Cappellini GC, Bonmassar L, Alvino E, Caporali S, Lacal PM, Narducci MG, Molineris I, De Galitiis F, Negrini M, Russo G, D’Atri S. Circulating miR-1246 and miR-485-3p as Promising Biomarkers of Clinical Response and Outcome in Melanoma Patients Treated with Targeted Therapy. Cancers (Basel) 2022; 14:cancers14153706. [PMID: 35954369 PMCID: PMC9367338 DOI: 10.3390/cancers14153706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/16/2022] [Accepted: 07/24/2022] [Indexed: 01/27/2023] Open
Abstract
Despite the significant improvements in advanced melanoma therapy, there is still a pressing need for biomarkers that can predict patient response and prognosis, and therefore support rational treatment decisions. Here, we investigated whether circulating miRNAs could be biomarkers of clinical outcomes in patients treated with targeted therapy. Using next-generation sequencing, we profiled plasma miRNAs at baseline and at progression in patients treated with BRAF inhibitors (BRAFi) or BRAFi + MEKi. Selected miRNAs associated with response to therapy were subjected to validation by real-time quantitative RT-PCR . Receiver Operating Characteristics (ROC), Kaplan–Meier and univariate and multivariate Cox regression analyses were performed on the validated miR-1246 and miR-485-3p baseline levels. The median baseline levels of miR-1246 and miR-485-3p were significantly higher and lower, respectively, in the group of patients not responding to therapy (NRs) as compared with the group of responding patients (Rs). In Rs, a trend toward an increase in miR-1246 and a decrease in miR-485-3p was observed at progression. Baseline miR-1246 level and the miR-1246/miR-485-3p ratio showed a good ability to discriminate between Rs and NRs. Poorer PFS and OS were observed in patients with unfavorable levels of at least one miRNA. In multivariate analysis, a low level of miR-485-3p and a high miR-1246/miR-485-3p ratio remained independent negative prognostic factors for PFS, while a high miR-1246/miR-485-3p ratio was associated with an increased risk of mortality, although statistical significance was not reached. Evaluation of miR-1246 and miR-485-3p baseline plasma levels might help clinicians to identify melanoma patients most likely to be unresponsive to targeted therapy or at higher risk for short-term PFS and mortality, thus improving their management.
Collapse
Affiliation(s)
- Lauretta Levati
- Laboratory of Molecular Oncology, IDI-IRCCS, Via dei Monti di Creta 104, 00167 Rome, Italy; (L.L.); (L.B.); (S.C.); (P.M.L.); (M.G.N.); (G.R.)
| | - Cristian Bassi
- Department of Translational Medicine, University of Ferrara, Via Fossato di Mortara 70, 44121 Ferrara, Italy; (C.B.); (L.L.); (M.N.)
- LTTA Center, University of Ferrara, Via Fossato di Mortara 70, 44121 Ferrara, Italy
| | - Simona Mastroeni
- Clinical Epidemiology Unit, IDI-IRCCS, Via dei Monti di Creta 104, 00167 Rome, Italy;
| | - Laura Lupini
- Department of Translational Medicine, University of Ferrara, Via Fossato di Mortara 70, 44121 Ferrara, Italy; (C.B.); (L.L.); (M.N.)
| | - Gian Carlo Antonini Cappellini
- Department of Oncology and Dermatological Oncology, IDI-IRCCS, Via dei Monti di Creta 104, 00167 Rome, Italy; (G.C.A.C.); (F.D.G.)
| | - Laura Bonmassar
- Laboratory of Molecular Oncology, IDI-IRCCS, Via dei Monti di Creta 104, 00167 Rome, Italy; (L.L.); (L.B.); (S.C.); (P.M.L.); (M.G.N.); (G.R.)
| | - Ester Alvino
- Institute of Translational Pharmacology, National Council of Research, Via Fosso del Cavaliere 100, 00133 Rome, Italy;
| | - Simona Caporali
- Laboratory of Molecular Oncology, IDI-IRCCS, Via dei Monti di Creta 104, 00167 Rome, Italy; (L.L.); (L.B.); (S.C.); (P.M.L.); (M.G.N.); (G.R.)
| | - Pedro Miguel Lacal
- Laboratory of Molecular Oncology, IDI-IRCCS, Via dei Monti di Creta 104, 00167 Rome, Italy; (L.L.); (L.B.); (S.C.); (P.M.L.); (M.G.N.); (G.R.)
| | - Maria Grazia Narducci
- Laboratory of Molecular Oncology, IDI-IRCCS, Via dei Monti di Creta 104, 00167 Rome, Italy; (L.L.); (L.B.); (S.C.); (P.M.L.); (M.G.N.); (G.R.)
| | - Ivan Molineris
- Department of Life Science and System Biology, University of Turin, Via Accademia Albertina 13, 10123 Turin, Italy;
| | - Federica De Galitiis
- Department of Oncology and Dermatological Oncology, IDI-IRCCS, Via dei Monti di Creta 104, 00167 Rome, Italy; (G.C.A.C.); (F.D.G.)
| | - Massimo Negrini
- Department of Translational Medicine, University of Ferrara, Via Fossato di Mortara 70, 44121 Ferrara, Italy; (C.B.); (L.L.); (M.N.)
- LTTA Center, University of Ferrara, Via Fossato di Mortara 70, 44121 Ferrara, Italy
| | - Giandomenico Russo
- Laboratory of Molecular Oncology, IDI-IRCCS, Via dei Monti di Creta 104, 00167 Rome, Italy; (L.L.); (L.B.); (S.C.); (P.M.L.); (M.G.N.); (G.R.)
| | - Stefania D’Atri
- Laboratory of Molecular Oncology, IDI-IRCCS, Via dei Monti di Creta 104, 00167 Rome, Italy; (L.L.); (L.B.); (S.C.); (P.M.L.); (M.G.N.); (G.R.)
- Correspondence:
| |
Collapse
|
6
|
Wang W, Dong D, Chen L, Wang H, Bi B, Liu T. Identification of Crucial Gene Modules Related to the Efficiency of Anti-PD-1/PD-L1 Therapy and Comprehensive Analyses of a Novel Signature Based on These Modules. Front Genet 2022; 13:893380. [PMID: 35937997 PMCID: PMC9354784 DOI: 10.3389/fgene.2022.893380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 06/09/2022] [Indexed: 11/13/2022] Open
Abstract
Biomarker development for clinical checkpoint inhibition is still in its early stages. It is critical to determine the cause of the lack of a long-term response in patients after immune checkpoint blockade (ICB) treatment and to develop composite biomarkers or signatures to improve personalized approaches. Three modules that were significantly correlated with the immunotherapeutic response were identified. Stimulatory pathways of cellular immunity, extracellular matrix formation-related pathways, and ATP metabolism-related pathways were enriched. Two distinct transcriptional subtypes were determined. Tumor microenvironment (TME) characteristics were highly correlated with “hot” and “cold” tumors. The ICB score was significantly correlated with clinical characteristics including age, Breslow depth, Clerk level, AJCC stage, and T stage. Meanwhile, a low ICB score is characterized by increased activation of immunity, a higher level of immune infiltration, and immune molecule expression. The ICB score showed a robust ability to predict melanoma prognosis in the discovery, internal validation, and external validation cohorts. In addition, a low ICB score was linked to a higher CR/PR rate in the immunotherapeutic cohort. The ICB score could reflect the pre-existing immune features and the expression pattern of “Cold” versus “Hot” tumors in melanoma patients. Thus, it has the potential to serve as a reliable predictor of melanoma prognosis and response to ICB therapy.
Collapse
Affiliation(s)
- Wei Wang
- Department of Plastic and Aesthetic Surgery, Huadong Hospital, Fudan University, Shanghai, China
- Shanghai Medical College of Fudan University, Shanghai, China
| | - Dong Dong
- Department of Plastic and Aesthetic Surgery, Huadong Hospital, Fudan University, Shanghai, China
- Shanghai Medical College of Fudan University, Shanghai, China
| | - Liang Chen
- Department of Plastic and Aesthetic Surgery, Huadong Hospital, Fudan University, Shanghai, China
| | - Heng Wang
- Department of Plastic and Aesthetic Surgery, Huadong Hospital, Fudan University, Shanghai, China
- Shanghai Medical College of Fudan University, Shanghai, China
| | - Bo Bi
- Department of Cosmetic Surgery, Shanghai Changning Maternity and Infant Health Hospital, Shanghai, China
- *Correspondence: Tianyi Liu, ; Bo Bi,
| | - Tianyi Liu
- Department of Plastic and Aesthetic Surgery, Huadong Hospital, Fudan University, Shanghai, China
- *Correspondence: Tianyi Liu, ; Bo Bi,
| |
Collapse
|
7
|
Loth E, Ahmad J, Chatham C, López B, Carter B, Crawley D, Oakley B, Hayward H, Cooke J, San José Cáceres A, Bzdok D, Jones E, Charman T, Beckmann C, Bourgeron T, Toro R, Buitelaar J, Murphy D, Dumas G. The meaning of significant mean group differences for biomarker discovery. PLoS Comput Biol 2021; 17:e1009477. [PMID: 34793435 PMCID: PMC8601419 DOI: 10.1371/journal.pcbi.1009477] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Over the past decade, biomarker discovery has become a key goal in psychiatry to aid in the more reliable diagnosis and prognosis of heterogeneous psychiatric conditions and the development of tailored therapies. Nevertheless, the prevailing statistical approach is still the mean group comparison between "cases" and "controls," which tends to ignore within-group variability. In this educational article, we used empirical data simulations to investigate how effect size, sample size, and the shape of distributions impact the interpretation of mean group differences for biomarker discovery. We then applied these statistical criteria to evaluate biomarker discovery in one area of psychiatric research-autism research. Across the most influential areas of autism research, effect size estimates ranged from small (d = 0.21, anatomical structure) to medium (d = 0.36 electrophysiology, d = 0.5, eye-tracking) to large (d = 1.1 theory of mind). We show that in normal distributions, this translates to approximately 45% to 63% of cases performing within 1 standard deviation (SD) of the typical range, i.e., they do not have a deficit/atypicality in a statistical sense. For a measure to have diagnostic utility as defined by 80% sensitivity and 80% specificity, Cohen's d of 1.66 is required, with still 40% of cases falling within 1 SD. However, in both normal and nonnormal distributions, 1 (skewness) or 2 (platykurtic, bimodal) biologically plausible subgroups may exist despite small or even nonsignificant mean group differences. This conclusion drastically contrasts the way mean group differences are frequently reported. Over 95% of studies omitted the "on average" when summarising their findings in their abstracts ("autistic people have deficits in X"), which can be misleading as it implies that the group-level difference applies to all individuals in that group. We outline practical approaches and steps for researchers to explore mean group comparisons for the discovery of stratification biomarkers.
Collapse
Affiliation(s)
- Eva Loth
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- Sackler Institute for Translational Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Jumana Ahmad
- Department of Psychology, Social Work and Counselling, Faculty of Education and Health, University of Greenwich, London, United Kingdom
| | - Chris Chatham
- Neuroscience & Rare Diseases, Pharma Research & Early Development, Roche Innovation Center New York, New York, United States of America
| | - Beatriz López
- Department of Psychology, Portsmouth University, Portsmouth, United Kingdom
| | - Ben Carter
- Department of Biostatistics, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Daisy Crawley
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Bethany Oakley
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Hannah Hayward
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Jennifer Cooke
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Antonia San José Cáceres
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- Instituto de Investigación Sanitaria Gregorio Marañón, Departamento de Psiquiatría del Niño y del Adolescente, Hospital General Universitario Gregorio Marañón and Centro Investigación Biomédica en Red Salud Mental (CIBERSAM), Madrid, Spain
| | - Danilo Bzdok
- Department of Biomedical Engineering, McConnell Brain-Imaging Centre (BIC), Montreal Neurological Institute (MNI), Faculty of Medicine, McGill University, Montreal, Canada
- Canadian Institute for Advanced Research (CIFAR), Canada
- Mila–Quebec Artificial Intelligence Institute, Montreal, Canada
| | - Emily Jones
- Centre for Brain and Cognitive Development, Birkbeck, University of London, London, United Kingdom
| | - Tony Charman
- Department of Psychology, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Christian Beckmann
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Thomas Bourgeron
- Human Genetics and Cognitive Functions, Institut Pasteur, UMR3571 CNRS, Université de Paris, Paris, France
| | - Roberto Toro
- Human Genetics and Cognitive Functions, Institut Pasteur, UMR3571 CNRS, Université de Paris, Paris, France
| | - Jan Buitelaar
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Declan Murphy
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- Sackler Institute for Translational Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Guillaume Dumas
- Mila–Quebec Artificial Intelligence Institute, Montreal, Canada
- Human Genetics and Cognitive Functions, Institut Pasteur, UMR3571 CNRS, Université de Paris, Paris, France
- Precision Psychiatry and Social Physiology laboratory, CHU Sainte-Justine Research Center, Department of Psychiatry, University of Montreal, Quebec, Canada
| |
Collapse
|
8
|
Drljevic-Nielsen A, Rasmussen F, Mains JR, Thorup K, Donskov F. Blood Volume as a new functional image-based biomarker of progression in metastatic renal cell carcinoma. Sci Rep 2021; 11:19659. [PMID: 34608226 PMCID: PMC8490379 DOI: 10.1038/s41598-021-99122-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 09/21/2021] [Indexed: 11/09/2022] Open
Abstract
RECIST v1.1 has limitations in evaluating progression. We assessed Dynamic Constrast Enhanced Computed Tomography (DCE-CT) identified Blood Volume (BV) for the evaluation of progressive disease (PD) in patients with metastatic renal cell carcinoma (mRCC). BV was quantified prospectively at baseline, after one month, then every three months until PD. Relative changes (ΔBV) were assessed at each timepoint compared with baseline values. The primary endpoint was Time to PD (TTP), the secondary endpoint was Time to the scan prior to PD (PDminus1). Cox proportional hazard models adjusted ΔBV for treatments and International mRCC Database Consortium factors. A total of 62 patients had analyzable scans at the PD timepoint. Median BV was 23.92 mL × 100 g-1 (range 4.40-399.04) at PD and 26.39 mL × 100 g-1 (range 8.70-77.44) at PDminus1. In the final multivariate analysis higher ΔBV was statistically significantly associated with shorter Time to PD, HR 1.11 (95% CI 1.07-1.15, P < 0.001). Also assessed at PDminus1, higher ΔBV was significantly associated with shorter time to PD, HR 1.14 (95% CI 1.01-1.28, P = 0.031). In conclusion, DCE-CT identified BV is a new image-based biomarker of therapy progression in patients with mRCC.
Collapse
Affiliation(s)
- Aska Drljevic-Nielsen
- Department of Radiology, Aarhus University Hospital, Aarhus N, Denmark.,Department of Oncology, Aarhus University Hospital, Aarhus N, Denmark
| | - Finn Rasmussen
- Department of Radiology, Aarhus University Hospital, Aarhus N, Denmark
| | - Jill Rachel Mains
- Department of Radiology, Aarhus University Hospital, Aarhus N, Denmark
| | - Kennet Thorup
- Department of Radiology, Aarhus University Hospital, Aarhus N, Denmark
| | - Frede Donskov
- Department of Oncology, Aarhus University Hospital, Aarhus N, Denmark.
| |
Collapse
|
9
|
Ohnuma K, Uchida T, Leung GNW, Ueda T, Obara T, Ishii H. Establishment of a post-race biomarkers database and application of pathway analysis to identify potential biomarkers in post-race equine plasma. Drug Test Anal 2021; 14:915-928. [PMID: 33835667 DOI: 10.1002/dta.3041] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/30/2021] [Accepted: 04/02/2021] [Indexed: 12/30/2022]
Abstract
In the context of doping control, conventional direct chemical testing detects only a limited scope of target substances in equine biological samples. To expand the ability to detect doping agents and their detection windows, metabolomics has recently become a common approach for monitoring alteration of biomarkers caused by doping agents in relevant metabolic pathways. In horse racing, remarkable changes in metabolic profiles between the rest state and racing are likely to affect the identification of doping biomarkers. Previously, we reported a limited number of significantly upregulated metabolites after racing, based on a non-targeted metabolomics approach using out-of-competition and post-race equine plasma samples. In this study, we performed a more thorough analysis of the data set, using pathway analysis to establish a post-race biomarkers database (PBD) that includes upregulated and downregulated metabolites, their fold changes, and relevant pathways, with the main objective of improving our understanding of changes in physiological status related to horse racing. Statistical analysis of the PBD revealed that two peak combinations of pentadecanoyl carnitine/galactosylglycerol (P/G) and heptadecanoyl carnitine/galactosylglycerol (H/G) could be used as potential biomarkers for the discrimination of the rest and post-race groups. To demonstrate the applicability of the PBD, we validated the post-race biomarkers P/G and H/G (highly involved in lipid metabolism) by a single-blind test. This strategy, which combines establishment of a biomarker database with pathway analysis, represents a powerful tool for discovering potential doping biomarkers in the future.
Collapse
Affiliation(s)
- Kohei Ohnuma
- Drug Analysis Department, Laboratory of Racing Chemistry, Utsunomiya, Japan
| | - Taiga Uchida
- Drug Analysis Department, Laboratory of Racing Chemistry, Utsunomiya, Japan
| | - Gary Ngai-Wa Leung
- Drug Analysis Department, Laboratory of Racing Chemistry, Utsunomiya, Japan
| | - Toshiki Ueda
- Drug Analysis Department, Laboratory of Racing Chemistry, Utsunomiya, Japan.,Bioinformatics Team, Research Laboratory, H. U. Group Research Institute G.K., Hachioji, Japan
| | - Taku Obara
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan
| | - Hideaki Ishii
- Drug Analysis Department, Laboratory of Racing Chemistry, Utsunomiya, Japan.,Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Japan
| |
Collapse
|
10
|
Development of a Genetic Risk Score to predict the risk of overweight and obesity in European adolescents from the HELENA study. Sci Rep 2021; 11:3067. [PMID: 33542408 PMCID: PMC7862459 DOI: 10.1038/s41598-021-82712-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 01/22/2021] [Indexed: 11/08/2022] Open
Abstract
Obesity is the result of interactions between genes and environmental factors. Since monogenic etiology is only known in some obesity-related genes, a genetic risk score (GRS) could be useful to determine the genetic predisposition to obesity. Therefore, the aim of our study was to build a GRS able to predict genetic predisposition to overweight and obesity in European adolescents. A total of 1069 adolescents (51.3% female), aged 11-19 years participating in the Healthy Lifestyle in Europe by Nutrition in Adolescence (HELENA) cross-sectional study were genotyped. The sample was divided in non-overweight (non-OW) and overweight/obesity (OW/OB). From 611 single nucleotide polymorphisms (SNP) available, a first screening of 104 SNPs univariately associated with obesity (p < 0.20) was established selecting 21 significant SNPs (p < 0.05) in the multivariate model. Unweighted GRS (uGRS) was calculated by summing the number of risk alleles and weighted GRS (wGRS) by multiplying the risk alleles to each estimated coefficient. The area under curve (AUC) was calculated in uGRS (0.723) and wGRS (0.734) using tenfold internal cross-validation. Both uGRS and wGRS were significantly associated with body mass index (BMI) (p < .001). Both GRSs could potentially be considered as useful genetic tools to evaluate individual's predisposition to overweight/obesity in European adolescents.
Collapse
|
11
|
Kim H, Kim Y, Bang S, Park S, Jee S, Sim J, Shin SJ, Paik SS, Jang K. Low Expression of Single-stranded DNA Binding Protein 2 (SSBP2) Predicts Unfavourable Postoperative Outcomes in Patients With Clear Cell Renal Cell Carcinoma. In Vivo 2020; 34:101-107. [PMID: 31882468 DOI: 10.21873/invivo.11750] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 10/31/2019] [Accepted: 11/05/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Single-stranded DNA binding protein 2 (SSBP2) is a subunit of a single-stranded DNA binding complex, which is involved in the maintenance of hematopoietic stem cells and stress responses. Numerous studies have suggested that SSBP2 functions as a tumor suppressor and is silenced through a pathway mediated by promoter hypermethylation. However, the role of SSBP2 in human renal cell carcinoma has not been reported, to date. Herein, we investigated the clinicopathological significance of SSBP2 expression in clear cell renal cell carcinoma (ccRCC). MATERIALS AND METHODS We constructed tissue micro arrays consisting of 173 ccRCC tissues, and SSBP2 expression was evaluated semi-quantitatively based on the staining intensity and the proportion of stained cells. Regarding statistical analysis, the tissues were divided into two groups according to SSBP2 expression, and correlation of SSBP2 expression with various clinicopathological characteristics and patient outcomes was evaluated. RESULTS Low SSBP2 expression was observed in 114 of 175 (65.9%) of ccRCC cases, and low SSBP2 expression was significantly correlated with larger tumor size (p=0.005, Chi-square test), higher WHO/ISUP histological grade (p<0.001, Chi-square test), tumor necrosis (p=0.008, Chi-square test), sarcomatoid change (p=0.021, Chi-square test), and higher pT AJCC stage (p=0.002, Chi-square test). Kaplan-Meier survival curves revealed that patients with low SSBP2 expression had worse recurrence-free survival (p=0.041, log-rank test). CONCLUSION ccRCC with low SSBP2 expression was associated with adverse clinicopathological characteristics and poor patient outcomes.
Collapse
Affiliation(s)
- Hyunsung Kim
- Department of Pathology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Yeseul Kim
- Department of Pathology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Seongsik Bang
- Department of Pathology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Seongeon Park
- Department of Pathology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Seungyun Jee
- Department of Pathology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Jongmin Sim
- Department of Pathology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Su-Jin Shin
- Department of Pathology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Seung Sam Paik
- Department of Pathology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Kiseok Jang
- Department of Pathology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| |
Collapse
|
12
|
Potential Salivary mRNA Biomarkers for Early Detection of Oral Cancer. J Clin Med 2020; 9:jcm9010243. [PMID: 31963366 PMCID: PMC7019677 DOI: 10.3390/jcm9010243] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/07/2020] [Accepted: 01/12/2020] [Indexed: 12/11/2022] Open
Abstract
We evaluated potential biomarkers in human whole saliva for the early diagnosis of oral squamous cell carcinoma (OSCC). We selected 30 candidate genes with relevance to cancer from recent reports in PubMed. Saliva samples were obtained from 34 non-tumor control and 33 OSCC patients. Real-time PCR was performed, and mRNA levels were compared. Normalized mRNA levels of six genes (NGFI-A binding protein 2 (NAB2), cytochrome P450, family 27, subfamily A, polypeptide 1 (CYP27A1), nuclear pore complex interacting protein family, member B4 (NPIPB4), monoamine oxidase B (MAOB), sialic acid acetyltransferase (SIAE), and collagen, type III, alpha 1 (COL3A1)) were significantly lower in saliva of OSCC patients. Receiver operating characteristics (ROC) analysis was used to individually evaluate the predictive power of the potential biomarkers for OSCC diagnosis. The area under the curve (AUC) values were evaluated for the OSCC vs. non-tumor groups via univariate ROC analyses, as well as multivariate ROC analyses of combinations of multiple potential biomarkers. The combination of CYP27A1 + SIAE showed a favorable AUC value of 0.84. When we divided saliva samples into two groups according to age using a 60-year cut-off, with OSCC patients and controls evaluated together, the AUC of MAOB-NAB2 was more predictive of OSCC in the under-60 group (AUC, 0.91; sensitivity, 0.92; and specificity, 0.86) than any other gene combination. These results are expected to aid the early diagnosis of OSCC, especially in patients under 60 years of age. While more studies with larger numbers of patients are necessary, our result suggest that salivary mRNA would be a potent biomarker for early OSCC diagnosis.
Collapse
|
13
|
Computational Prediction of Subjective Human Immunodeficiency Virus Status in Malawi Using a Random Forest Approach. AIDS Res Treat 2019; 2019:5849183. [PMID: 31637055 PMCID: PMC6766123 DOI: 10.1155/2019/5849183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 07/17/2019] [Indexed: 01/21/2023] Open
Abstract
An individual's subjective judgment about his or her Human Immunodeficiency Virus status depends on certain factors, behavioral, health, and sociodemographic alike. This paper aims to develop a model with good accuracy for predicting subjective HIV infection status using the random forest approach. A total of 12,796 responses of Malawians over a 12-year period were assessed. Fourteen risk factors including behavioral, health, and sociodemographic information were analysed as potential predictors of subjective Human Immunodeficiency Virus infection status in the general population and thirteen behavioral, health, and sociodemographic information were analysed among males and females. The random forest approach was adopted to build a comprehensive model comprising 14 risk factors in Malawi. It was revealed that age, worries about infection, and health rate were the most significant predictors as compared to use of condoms, marital status, and education which were the least important predictors of subjective Human Immunodeficiency Virus status in Malawi. However, the importance of infidelity on the part of a spouse and marital status as predictors of subjective Human Immunodeficiency Virus status alternated among males and females. The importance of infidelity and marital status was relatively high among females than among males. The model achieved a prediction accuracy of about 97%-99% measured by c-statistic with jack-knife cross validation and verified by Mathews correlation coefficient. As a result, RF based model has great potential to be an effective approach for analysing subjective health status.
Collapse
|
14
|
Lu S, Stein JE, Rimm DL, Wang DW, Bell JM, Johnson DB, Sosman JA, Schalper KA, Anders RA, Wang H, Hoyt C, Pardoll DM, Danilova L, Taube JM. Comparison of Biomarker Modalities for Predicting Response to PD-1/PD-L1 Checkpoint Blockade: A Systematic Review and Meta-analysis. JAMA Oncol 2019; 5:1195-1204. [PMID: 31318407 PMCID: PMC6646995 DOI: 10.1001/jamaoncol.2019.1549] [Citation(s) in RCA: 416] [Impact Index Per Article: 83.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 03/15/2019] [Indexed: 12/30/2022]
Abstract
IMPORTANCE PD-L1 (programmed cell death ligand 1) immunohistochemistry (IHC), tumor mutational burden (TMB), gene expression profiling (GEP), and multiplex immunohistochemistry/immunofluorescence (mIHC/IF) assays have been used to assess pretreatment tumor tissue to predict response to anti-PD-1/PD-L1 therapies. However, the relative diagnostic performance of these modalities has yet to be established. OBJECTIVE To compare studies that assessed the diagnostic accuracy of PD-L1 IHC, TMB, GEP, and mIHC/IF in predicting response to anti-PD-1/PD-L1 therapy. EVIDENCE REVIEW A search of PubMed (from inception to June 2018) and 2013 to 2018 annual meeting abstracts from the American Association for Cancer Research, American Society of Clinical Oncology, European Society for Medical Oncology, and Society for Immunotherapy of Cancer was conducted to identify studies that examined the use of PD-L1 IHC, TMB, GEP, and mIHC/IF assays to determine objective response to anti-PD-1/PD-L1 therapy. For PD-L1 IHC, only clinical trials that resulted in US Food and Drug Administration approval of indications for anti-PD-1/PD-L1 were included. Studies combining more than 1 modality were also included. Preferred Reporting Items for Systematic Reviews and Meta-analysis guidelines were followed. Two reviewers independently extracted the clinical outcomes and test results for each individual study. MAIN OUTCOMES AND MEASURES Summary receiver operating characteristic (sROC) curves; their associated area under the curve (AUC); and pooled sensitivity, specificity, positive and negative predictive values (PPV, NPV), and positive and negative likelihood ratios (LR+ and LR-) for each assay modality. RESULTS Tumor specimens representing over 10 different solid tumor types in 8135 patients were assayed, and the results were correlated with anti-PD-1/PD-L1 response. When each modality was evaluated with sROC curves, mIHC/IF had a significantly higher AUC (0.79) compared with PD-L1 IHC (AUC, 0.65, P < .001), GEP (AUC, 0.65, P = .003), and TMB (AUC, 0.69, P = .049). When multiple different modalities were combined such as PD-L1 IHC and/or GEP + TMB, the AUC drew nearer to that of mIHC/IF (0.74). All modalities demonstrated comparable NPV and LR-, whereas mIHC/IF demonstrated higher PPV (0.63) and LR+ (2.86) than the other approaches. CONCLUSIONS AND RELEVANCE In this meta-analysis, tumor mutational burden, PD-L1 IHC, and GEP demonstrated comparable AUCs in predicting response to anti-PD-1/PD-L1 treatment. Multiplex immunohistochemistry/IF and multimodality biomarker strategies appear to be associated with improved performance over PD-L1 IHC, TMB, or GEP alone. Further studies with mIHC/IF and composite approaches with a larger number of patients will be required to confirm these findings. Additional study is also required to determine the most predictive analyte combinations and to determine whether biomarker modality performance varies by tumor type.
Collapse
Affiliation(s)
- Steve Lu
- Department of Dermatology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Julie E. Stein
- Department of Dermatology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - David L. Rimm
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Daphne W. Wang
- Department of Dermatology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - J. Michael Bell
- Department of Dermatology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Douglas B. Johnson
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jeffrey A. Sosman
- Department of Medicine, Division of Hematology-Oncology, Northwestern University Medical Center, and Robert H. Lurie Cancer Center, Chicago, Illinois
| | - Kurt A. Schalper
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Robert A. Anders
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Hao Wang
- Division of Biostatistics & Bioinformatics at the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins Medical Institutions, Baltimore, Maryland
| | | | - Drew M. Pardoll
- Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Ludmila Danilova
- Division of Biostatistics & Bioinformatics at the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins Medical Institutions, Baltimore, Maryland
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Janis M. Taube
- Department of Dermatology, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins Medical Institutions, Baltimore, Maryland
| |
Collapse
|
15
|
Caporali S, Amaro A, Levati L, Alvino E, Lacal PM, Mastroeni S, Ruffini F, Bonmassar L, Antonini Cappellini GC, Felli N, Carè A, Pfeffer U, D'Atri S. miR-126-3p down-regulation contributes to dabrafenib acquired resistance in melanoma by up-regulating ADAM9 and VEGF-A. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:272. [PMID: 31227006 PMCID: PMC6588909 DOI: 10.1186/s13046-019-1238-4] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 05/21/2019] [Indexed: 02/06/2023]
Abstract
Background Development of resistance to inhibitors of BRAF (BRAFi) and MEK (MEKi) remains a great challenge for targeted therapy in patients with BRAF-mutant melanoma. Here, we explored the role of miRNAs in melanoma acquired resistance to BRAFi. Methods miRNA expression in two BRAF-mutant melanoma cell lines and their dabrafenib-resistant sublines was determined using Affymetrix GeneChip® miRNA 3.1 microarrays and/or qRT-PCR. The effects of miR-126-3p re-expression on proliferation, apoptosis, cell cycle, ERK1/2 and AKT phosphorylation, dabrafenib sensitivity, invasiveness and VEGF-A secretion were evaluated in the dabrafenib-resistant sublines using MTT assays, flow cytometry, immunoblotting, invasion assays in Boyden chambers and ELISA. ADAM9, PIK3R2, MMP7 and CXCR4 expression in the sensitive and dabrafenib-resistant cells was determined by immunoblotting. Small RNA interference was performed to investigate the consequence of VEGFA or ADAM9 silencing on proliferation, invasiveness or dabrafenib sensitivity of the resistant sublines. Long-term proliferation assays were carried out in dabrafenib-sensitive cells to assess the effects of enforced miR-126-3p expression or ADAM9 silencing on resistance development. VEGF-A serum levels in melanoma patients treated with BRAFi or BRAFi+MEKi were evaluated at baseline (T0), after two months of treatment (T2) and at progression (TP) by ELISA. Results miR-126-3p was significantly down-regulated in the dabrafenib-resistant sublines as compared with their parental counterparts. miR-126-3p replacement in the drug-resistant cells inhibited proliferation, cell cycle progression, phosphorylation of ERK1/2 and/or AKT, invasiveness, VEGF-A and ADAM9 expression, and increased dabrafenib sensitivity. VEGFA or ADAM9 silencing impaired proliferation and invasiveness of the drug-resistant sublines. ADAM9 knock-down in the resistant cells increased dabrafenib sensitivity, whereas miR-126-3p enforced expression or ADAM9 silencing in the drug-sensitive cells delayed the development of resistance. At T0 and T2, statistically significant differences were observed in VEGF-A serum levels between patients who responded to therapy and patients who did not. In responder patients, a significant increase of VEGF-A levels was observed at TP versus T2. Conclusions Strategies restoring miR-126-3p expression or targeting VEGF-A or ADAM9 could restrain growth and metastasis of dabrafenib-resistant melanomas and increase their drug sensitivity. Circulating VEGF-A is a promising biomarker for predicting patients’ response to BRAFi or BRAFi+MEKi and for monitoring the onset of resistance. Electronic supplementary material The online version of this article (10.1186/s13046-019-1238-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Simona Caporali
- Laboratory of Molecular Oncology, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Via dei Monti di Creta 104, 00167, Rome, Italy
| | - Adriana Amaro
- Molecular Pathology, IRCCS-Ospedale Policlinico San Martino, Genoa, Italy
| | - Lauretta Levati
- Laboratory of Molecular Oncology, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Via dei Monti di Creta 104, 00167, Rome, Italy
| | - Ester Alvino
- Institute of Translational Pharmacology, National Council of Research, Rome, Italy
| | - Pedro Miguel Lacal
- Laboratory of Molecular Oncology, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Via dei Monti di Creta 104, 00167, Rome, Italy
| | | | - Federica Ruffini
- Laboratory of Molecular Oncology, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Via dei Monti di Creta 104, 00167, Rome, Italy
| | - Laura Bonmassar
- Laboratory of Molecular Oncology, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Via dei Monti di Creta 104, 00167, Rome, Italy
| | | | - Nadia Felli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Alessandra Carè
- Center of Gender Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Ulrich Pfeffer
- Molecular Pathology, IRCCS-Ospedale Policlinico San Martino, Genoa, Italy
| | - Stefania D'Atri
- Laboratory of Molecular Oncology, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Via dei Monti di Creta 104, 00167, Rome, Italy.
| |
Collapse
|
16
|
Sato T, Kawasaki Y, Maekawa M, Takasaki S, Saigusa D, Ota H, Shimada S, Yamashita S, Mitsuzuka K, Yamaguchi H, Ito A, Kinoshita K, Koshiba S, Mano N, Arai Y. Value of global metabolomics in association with diagnosis and clinicopathological factors of renal cell carcinoma. Int J Cancer 2019; 145:484-493. [PMID: 30628065 DOI: 10.1002/ijc.32115] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 12/26/2018] [Accepted: 01/03/2019] [Indexed: 01/28/2023]
Abstract
Renal cell carcinoma (RCC) is a malignant tumor that currently lacks clinically useful biomarkers indicative of early diagnosis or disease status. RCC has commonly been diagnosed based on imaging results. Metabolomics offers a potential technology for discovering biomarkers and therapeutic targets by comprehensive screening of metabolites from patients with various cancers. We aimed to identify metabolites associated with early diagnosis and clinicopathological factors in RCC using global metabolomics (G-Met). Tumor and nontumor tissues were sampled from 20 cases of surgically resected clear cell RCC. G-Met was performed by liquid chromatography mass spectrometry and important metabolites specific to RCC were analyzed by multivariate statistical analysis for cancer diagnostic ability based on area under the curve (AUC) and clinicopathological factors (tumor volume, pathological T stage, Fuhrman grade, presence of coagulation necrosis and distant metastasis). We identified 58 metabolites showing significantly increased levels in tumor tissues, 34 of which showed potential early diagnostic ability (AUC >0.8), but 24 did not discriminate between tumor and nontumor tissues (AUC ≤0.8). We recognized 6 pathways from 9 metabolites with AUC >0.8 and 7 pathways from 10 metabolites with AUC ≤0.8 about malignant status. Clinicopathological factors involving malignant status correlated significantly with metabolites showing AUC ≤0.8 (p = 0.0279). The tricarboxylic acid cycle (TCA) cycle, TCA cycle intermediates, nucleotide sugar pathway and inositol pathway were characteristic pathways for the malignant status of RCC. In conclusion, our study found that metabolites and their pathways allowed discrimination between early diagnosis and malignant status in RCC according to our G-Met protocol.
Collapse
Affiliation(s)
- Tomonori Sato
- Department of Urology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Yoshihide Kawasaki
- Department of Urology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Masamitsu Maekawa
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Shinya Takasaki
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Daisuke Saigusa
- Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan.,Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan.,LEAP, Japan Agency for Medical Research and Development (AMED), Chiyoda, Tokyo, Japan
| | - Hideki Ota
- Diagnostic Radiology, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Shuichi Shimada
- Department of Urology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Shinichi Yamashita
- Department of Urology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Koji Mitsuzuka
- Department of Urology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Hiroaki Yamaguchi
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Akihiro Ito
- Department of Urology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Kengo Kinoshita
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Seizo Koshiba
- Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan.,Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Nariyasu Mano
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Yoichi Arai
- Department of Urology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| |
Collapse
|
17
|
Motzer RJ, Escudier B, Gannon A, Figlin RA. Sunitinib: Ten Years of Successful Clinical Use and Study in Advanced Renal Cell Carcinoma. Oncologist 2016; 22:41-52. [PMID: 27807302 DOI: 10.1634/theoncologist.2016-0197] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 08/03/2016] [Indexed: 01/07/2023] Open
Abstract
The oral multikinase inhibitor sunitinib malate was approved by the U.S. Food and Drug Administration in January 2006 for use in patients with advanced renal cell carcinoma (RCC). Since then, it has been approved globally for this indication and for patients with imatinib-resistant or -intolerant gastrointestinal stromal tumors and advanced pancreatic neuroendocrine tumors. As we mark the 10-year anniversary of the beginning of the era of targeted therapy, and specifically the approval of sunitinib, it is worthwhile to highlight the progress that has been made in advanced RCC as it relates to the study of sunitinib. We present the key trials and data for sunitinib that established it as a reference standard of care for first-line advanced RCC therapy and, along with other targeted agents, significantly altered the treatment landscape in RCC. Moreover, we discuss the research with sunitinib that has sought to refine its role via patient selection and prognostic markers, improve dosing and adverse event management, and identify predictive efficacy biomarkers, plus the extent to which this research has contributed to the overall understanding and management of RCC. We also explore the key learnings regarding study design and data interpretation from the sunitinib studies and how these findings and the sunitinib development program, in general, can be a model for successful development of other agents. Finally, ongoing research into the continued and future role of sunitinib in RCC management is discussed. THE ONCOLOGIST 2017;22:41-52 IMPLICATIONS FOR PRACTICE: Approved globally, sunitinib is established as a standard of care for first-line advanced renal cell carcinoma (RCC) therapy and, along with other targeted agents, has significantly altered the treatment landscape in RCC. Research with sunitinib that has sought to refine its role via patient selection and prognostic markers, improve dosing and adverse event management, and identify predictive efficacy biomarkers has contributed to the overall understanding and management of RCC. Key learnings regarding study design and data interpretation from the sunitinib studies and the sunitinib development program, in general, can be a model for the successful development of other agents.
Collapse
Affiliation(s)
- Robert J Motzer
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | | | - Robert A Figlin
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|