1
|
Chatanaka MK, Avery LM, Diamandis EP. Validation of new, circulating biomarkers for gliomas. Diagnosis (Berl) 2025:dx-2025-0012. [PMID: 40131804 DOI: 10.1515/dx-2025-0012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 02/28/2025] [Indexed: 03/27/2025]
Abstract
OBJECTIVES Biomarkers are useful clinical tools but only a handful of them are used routinely for patient care. Despite intense efforts to discover new, clinically useful biomarkers, very few new circulating biomarkers were implemented in clinical practice in the last 40 years. This is mainly due to rather poor clinical performance. Here, our goal was to validate the performance of a group of newly discovered circulating biomarkers for glioma by comparing our data with data from a paper recently published in Science Advances. METHODS We analyzed our own sets of clinical samples (gliomas (n=30), meningiomas (n=20)) and a different analytical assay (Proximity Extension Assay, OLINK Proteomics) to compare the results of Shen and colleagues. RESULTS Despite the sophistication of the utilized discovery method by the original investigators, we found that the newly proposed biomarkers for glioma (the best one presumably being SERPINA6) did not perform as originally claimed. CONCLUSIONS Scientific irreproducibility has been extensively discussed in the literature. A large proportion of newly discovered candidate biomarkers likely represent "false discovery" and significantly contribute to the propagation of irreproducible results between investigators. One of the best ways to assess the value of any new biomarker is by independent and extensive validation. Based on our previous classification of irreproducible results, we believe that this new work likely represents another example of biomarker false discovery.
Collapse
Affiliation(s)
- Miyo K Chatanaka
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Lisa M Avery
- Biostatistics Division, Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | | |
Collapse
|
2
|
Chatanaka MK, Yousef GM, Diamandis EP. The Unholy Grail of cancer screening: or is it just about the Benjamins? Clin Chem Lab Med 2025; 63:499-506. [PMID: 39301604 DOI: 10.1515/cclm-2024-1013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024]
Abstract
The biotechnology company Grail developed a non-invasive blood test (Galleri test) which is claimed to detect 50 types of cancer at early and potentially curable stages. The initially promising results from prospective studies, and the anticipated financial success of Grail led the sequencing giant Illumina to purchase Grail for $8 billion (2021). Following this event, Grail collaborated with the UK National Health System to further clarify the test's capability, in a 3-year prospective trial, along with the standard of care. The UK-NHS announced that the trial will provide a clearer understanding of the efficacy of the Galleri test within the NHS framework. If the test does not perform as expected, valuable insights will still be gained to guide future research aimed at enhancing cancer screening. We previously expressed concerns about the sensitivity and specificity of the Galleri test. In this opinion paper, we revisit the hyped technology, and we provide new suggestions on the use of this test.
Collapse
Affiliation(s)
- Miyo K Chatanaka
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - George M Yousef
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Laboratory Medicine Program, 7989 University Health Network , Toronto, ON, Canada
| | - Eleftherios P Diamandis
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, Canada
| |
Collapse
|
3
|
Magowan D, Abdulshafea M, Thompson D, Rajamoorthy SI, Owen R, Harris D, Prosser S. Blood-based biomarkers and novel technologies for the diagnosis of colorectal cancer and adenomas: a narrative review. Biomark Med 2024; 18:493-506. [PMID: 38900496 DOI: 10.1080/17520363.2024.2345583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 03/12/2024] [Indexed: 06/21/2024] Open
Abstract
Aim: Blood-based biomarkers have shown promise for diagnosing colorectal cancer (CRC) and adenomas (CRA). This review summarizes recent studies in this area. Methods: A literature search was undertaken for 01/01/2017-01/03/2023. Criteria included CRC, CRA, liquid-biopsy, blood-based tests and diagnosis. Results: 12,378 studies were reduced to 178 for data extraction. Sixty focused on proteomics, 53 on RNA species, 30 on cfDNA methylation, seven on antigens and autoantibodies and 28 on novel techniques. 169 case control and nine cohort studies. Number of participants ranged 100-54,297, mean age 58.26. CRC sensitivity and specificity ranged 9.10-100% and 20.40-100%, respectively. CRA sensitivity and specificity ranged 8.00-95.70% and 4.00-97.00%, respectively. Conclusion: Sensitive and specific blood-based tests exist for CRC and CRA. However, studies demonstrate heterogenous techniques and reporting quality. Further work should concentrate on validation and meta-analyzes.
Collapse
Affiliation(s)
- Drew Magowan
- Swansea University, Singleton Park, SA2 8PP, Swansea, UK
- Swansea Bay University Health Board, Department of General Surgery, Morriston Hospital, SA6 6NL, Swansea, UK
| | - Mansour Abdulshafea
- Swansea Bay University Health Board, Department of General Surgery, Morriston Hospital, SA6 6NL, Swansea, UK
| | - Dominic Thompson
- Swansea Bay University Health Board, Department of General Surgery, Morriston Hospital, SA6 6NL, Swansea, UK
| | - Shri-Ishvarya Rajamoorthy
- Swansea Bay University Health Board, Department of General Surgery, Morriston Hospital, SA6 6NL, Swansea, UK
| | - Rhiannon Owen
- Swansea University, Singleton Park, SA2 8PP, Swansea, UK
| | - Dean Harris
- Swansea University, Singleton Park, SA2 8PP, Swansea, UK
- Swansea Bay University Health Board, Department of General Surgery, Morriston Hospital, SA6 6NL, Swansea, UK
| | - Susan Prosser
- Swansea Bay University Health Board, Department of General Surgery, Morriston Hospital, SA6 6NL, Swansea, UK
| |
Collapse
|
4
|
Okimoto LYS, Mendonca-Neto R, Nakamura FG, Nakamura EF, Fenyö D, Silva CT. Few-shot genes selection: subset of PAM50 genes for breast cancer subtypes classification. BMC Bioinformatics 2024; 25:92. [PMID: 38429657 PMCID: PMC10908178 DOI: 10.1186/s12859-024-05715-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 02/21/2024] [Indexed: 03/03/2024] Open
Abstract
BACKGROUND In recent years, researchers have made significant strides in understanding the heterogeneity of breast cancer and its various subtypes. However, the wealth of genomic and proteomic data available today necessitates efficient frameworks, instruments, and computational tools for meaningful analysis. Despite its success as a prognostic tool, the PAM50 gene signature's reliance on many genes presents challenges in terms of cost and complexity. Consequently, there is a need for more efficient methods to classify breast cancer subtypes using a reduced gene set accurately. RESULTS This study explores the potential of achieving precise breast cancer subtype categorization using a reduced gene set derived from the PAM50 gene signature. By employing a "Few-Shot Genes Selection" method, we randomly select smaller subsets from PAM50 and evaluate their performance using metrics and a linear model, specifically the Support Vector Machine (SVM) classifier. In addition, we aim to assess whether a more compact gene set can maintain performance while simplifying the classification process. Our findings demonstrate that certain reduced gene subsets can perform comparable or superior to the full PAM50 gene signature. CONCLUSIONS The identified gene subsets, with 36 genes, have the potential to contribute to the development of more cost-effective and streamlined diagnostic tools in breast cancer research and clinical settings.
Collapse
Affiliation(s)
- Leandro Y S Okimoto
- Institute of Computing, Universidade Federal do Amazonas, Manaus, BR, Brazil.
| | - Rayol Mendonca-Neto
- Institute of Computing, Universidade Federal do Amazonas, Manaus, BR, Brazil
| | - Fabíola G Nakamura
- Institute of Computing, Universidade Federal do Amazonas, Manaus, BR, Brazil
| | - Eduardo F Nakamura
- Institute of Computing, Universidade Federal do Amazonas, Manaus, BR, Brazil
| | | | | |
Collapse
|
5
|
Pasqualetti F, Barberis A, Zanotti S, Montemurro N, De Salvo GL, Soffietti R, Mazzanti CM, Ius T, Caffo M, Paiar F, Bocci G, Lombardi G, Harris AL, Buffa FM. The impact of survivorship bias in glioblastoma research. Crit Rev Oncol Hematol 2023; 188:104065. [PMID: 37392899 DOI: 10.1016/j.critrevonc.2023.104065] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 06/26/2023] [Accepted: 06/28/2023] [Indexed: 07/03/2023] Open
Abstract
Despite advances in the therapy of Central Nervous System (CNS) malignancies, treatment of glioblastoma (GB) poses significant challenges due to GB resistance and high recurrence rates following post-operative radio-chemotherapy. The majority of prognostic and predictive GB biomarkers are currently developed using tumour samples obtained through surgical interventions. However, the selection criteria adopted by different neurosurgeons to determine which cases are suitable for surgery make operated patients not representative of all GB cases. Particularly, geriatric and frail individuals are excluded from surgical consideration in some cancer centers. Such selection generates a survival (or selection) bias that introduces limitations, rendering the patients or data chosen for downstream analyses not representative of the entire community. In this review, we discuss the implication of survivorship bias on current and novel biomarkers for patient selection, stratification, therapy, and outcome analyses.
Collapse
Affiliation(s)
- Francesco Pasqualetti
- Department of Oncology, University of Oxford, Oxford, UK; Radiation Oncology, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy.
| | | | - Sofia Zanotti
- Anatomic Pathology Unit, IRCCS Humanitas University Research Hospital, Milan, Italy
| | - Nicola Montemurro
- Department of Neurosurgery, Azienda Ospedaliero Universitaria Pisana (AOUP), Pisa, Italy
| | | | - Riccardo Soffietti
- Division of Neuro-Oncology, Department of Neuroscience, University and City of Health and Science University Hospital, Turin, Italy
| | | | - Tamara Ius
- Neurosurgery Unit, Head-Neck and NeuroScience Department University Hospital of Udine, p.le S. Maria della Misericordia 15, 33100 Udine, Italy
| | - Maria Caffo
- Unit of Neurosurgery, Department of Biomorphology and Dental Sciences and Morfophunctional Imaging, University Hospital "G. Martino", Messina, Italy
| | - Fabiola Paiar
- Radiation Oncology, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | - Guido Bocci
- Department of Clinical and Experimental Medicine, University of Pisa, I-56126 Pisa, Italy
| | - Giuseppe Lombardi
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy
| | | | - Francesca M Buffa
- Department of Oncology, University of Oxford, Oxford, UK; Department of Computing Sciences, Bocconi University, Milan, Italy; Institute for Data Science and Analytics, Bocconi University, Milano, Italy
| |
Collapse
|
6
|
Barker AD, Alba MM, Mallick P, Agus DB, Lee JSH. An Inflection Point in Cancer Protein Biomarkers: What Was and What's Next. Mol Cell Proteomics 2023:100569. [PMID: 37196763 PMCID: PMC10388583 DOI: 10.1016/j.mcpro.2023.100569] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 04/26/2023] [Accepted: 05/08/2023] [Indexed: 05/19/2023] Open
Abstract
Biomarkers remain the highest value proposition in cancer medicine today - especially protein biomarkers. Yet despite decades of evolving regulatory frameworks to facilitate the review of emerging technologies, biomarkers have been mostly about promise with very little to show for improvements in human health. Cancer is an emergent property of a complex system and deconvoluting the integrative and dynamic nature of the overall system through biomarkers is a daunting proposition. The last two decades have seen an explosion of multi-omics profiling and a range of advanced technologies for precision medicine, including the emergence of liquid biopsy, exciting advances in single cell analysis, artificial intelligence (machine and deep learning) for data analysis and many other advanced technologies that promise to transform biomarker discovery. Combining multiple omics modalities to acquire a more comprehensive landscape of the disease state, we are increasingly developing biomarkers to support therapy selection and patient monitoring. Furthering precision medicine, especially in oncology, necessitates moving away from the lens of reductionist thinking towards viewing and understanding that complex diseases are, in fact, complex adaptive systems. As such, we believe it is necessary to re-define biomarkers as representations of biological system states at different hierarchical levels of biological order. This definition could include traditional molecular, histologic, radiographic, or physiological characteristics, as well as emerging classes of digital markers and complex algorithms. To succeed in the future, we must move past purely observational individual studies and instead start building a mechanistic framework to enable integrative analysis of new studies within the context of prior studies. Identifying information in complex systems and applying theoretical constructs, such as information theory, to study cancer as a disease of dysregulated communication could prove to be "game changing" for the clinical outcome of cancer patients.
Collapse
Affiliation(s)
- Anna D Barker
- Lawrence J. Ellison Institute for Transformative Medicine, Los Angeles, CA; Complex Adaptive Systems Initiative and School of Life Sciences, Arizona State University, Tempe, Arizona
| | - Mario M Alba
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA
| | - Parag Mallick
- Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA; Department of Radiology, Stanford University, Stanford, CA
| | - David B Agus
- Lawrence J. Ellison Institute for Transformative Medicine, Los Angeles, CA; Keck School of Medicine, University of Southern California, Los Angeles, CA; Viterbi School of Engineering, University of Southern California, Los Angeles, CA
| | - Jerry S H Lee
- Lawrence J. Ellison Institute for Transformative Medicine, Los Angeles, CA; Keck School of Medicine, University of Southern California, Los Angeles, CA; Viterbi School of Engineering, University of Southern California, Los Angeles, CA
| |
Collapse
|
7
|
Messner CB, Demichev V, Wang Z, Hartl J, Kustatscher G, Mülleder M, Ralser M. Mass spectrometry-based high-throughput proteomics and its role in biomedical studies and systems biology. Proteomics 2023; 23:e2200013. [PMID: 36349817 DOI: 10.1002/pmic.202200013] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/13/2022] [Accepted: 10/13/2022] [Indexed: 11/11/2022]
Abstract
There are multiple reasons why the next generation of biological and medical studies require increasing numbers of samples. Biological systems are dynamic, and the effect of a perturbation depends on the genetic background and environment. As a consequence, many conditions need to be considered to reach generalizable conclusions. Moreover, human population and clinical studies only reach sufficient statistical power if conducted at scale and with precise measurement methods. Finally, many proteins remain without sufficient functional annotations, because they have not been systematically studied under a broad range of conditions. In this review, we discuss the latest technical developments in mass spectrometry (MS)-based proteomics that facilitate large-scale studies by fast and efficient chromatography, fast scanning mass spectrometers, data-independent acquisition (DIA), and new software. We further highlight recent studies which demonstrate how high-throughput (HT) proteomics can be applied to capture biological diversity, to annotate gene functions or to generate predictive and prognostic models for human diseases.
Collapse
Affiliation(s)
- Christoph B Messner
- Precision Proteomics Center, Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Vadim Demichev
- Institute of Biochemistry, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ziyue Wang
- Institute of Biochemistry, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Johannes Hartl
- Institute of Biochemistry, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Georg Kustatscher
- Wellcome Centre for Cell Biology, University of Edinburgh, Max Born Crescent, Edinburgh, Scotland, UK
| | - Michael Mülleder
- Core Facility High Throughput Mass Spectrometry, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Markus Ralser
- Institute of Biochemistry, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
8
|
Wani S, Humaira, Farooq I, Ali S, Rehman MU, Arafah A. Proteomic profiling and its applications in cancer research. Proteomics 2023. [DOI: 10.1016/b978-0-323-95072-5.00015-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
|
9
|
Bliziotis NG, Kluijtmans LAJ, Tinnevelt GH, Reel P, Reel S, Langton K, Robledo M, Pamporaki C, Pecori A, Van Kralingen J, Tetti M, Engelke UFH, Erlic Z, Engel J, Deutschbein T, Nölting S, Prejbisz A, Richter S, Adamski J, Januszewicz A, Ceccato F, Scaroni C, Dennedy MC, Williams TA, Lenzini L, Gimenez-Roqueplo AP, Davies E, Fassnacht M, Remde H, Eisenhofer G, Beuschlein F, Kroiss M, Jefferson E, Zennaro MC, Wevers RA, Jansen JJ, Deinum J, Timmers HJLM. Preanalytical Pitfalls in Untargeted Plasma Nuclear Magnetic Resonance Metabolomics of Endocrine Hypertension. Metabolites 2022; 12:679. [PMID: 35893246 PMCID: PMC9394285 DOI: 10.3390/metabo12080679] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/17/2022] [Accepted: 07/11/2022] [Indexed: 11/24/2022] Open
Abstract
Despite considerable morbidity and mortality, numerous cases of endocrine hypertension (EHT) forms, including primary aldosteronism (PA), pheochromocytoma and functional paraganglioma (PPGL), and Cushing's syndrome (CS), remain undetected. We aimed to establish signatures for the different forms of EHT, investigate potentially confounding effects and establish unbiased disease biomarkers. Plasma samples were obtained from 13 biobanks across seven countries and analyzed using untargeted NMR metabolomics. We compared unstratified samples of 106 PHT patients to 231 EHT patients, including 104 PA, 94 PPGL and 33 CS patients. Spectra were subjected to a multivariate statistical comparison of PHT to EHT forms and the associated signatures were obtained. Three approaches were applied to investigate and correct confounding effects. Though we found signatures that could separate PHT from EHT forms, there were also key similarities with the signatures of sample center of origin and sample age. The study design restricted the applicability of the corrections employed. With the samples that were available, no biomarkers for PHT vs. EHT could be identified. The complexity of the confounding effects, evidenced by their robustness to correction approaches, highlighted the need for a consensus on how to deal with variabilities probably attributed to preanalytical factors in retrospective, multicenter metabolomics studies.
Collapse
Affiliation(s)
- Nikolaos G. Bliziotis
- Department of Laboratory Medicine, Translational Metabolic Laboratory, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
| | - Leo A. J. Kluijtmans
- Department of Laboratory Medicine, Translational Metabolic Laboratory, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
| | - Gerjen H. Tinnevelt
- Department of Analytical Chemistry, Institute for Molecules and Materials, Radboud University, 6500 HB Nijmegen, The Netherlands; (G.H.T.); (J.J.J.)
| | - Parminder Reel
- Division of Population Health and Genomics, School of Medicine, University of Dundee, Dundee DD2 4BF, UK; (P.R.); (S.R.); (E.J.)
| | - Smarti Reel
- Division of Population Health and Genomics, School of Medicine, University of Dundee, Dundee DD2 4BF, UK; (P.R.); (S.R.); (E.J.)
| | - Katharina Langton
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (K.L.); (C.P.); (G.E.)
| | - Mercedes Robledo
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain;
| | - Christina Pamporaki
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (K.L.); (C.P.); (G.E.)
| | - Alessio Pecori
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Torino, 10124 Torino, Italy; (A.P.); (M.T.); (T.A.W.)
| | - Josie Van Kralingen
- British Heart Foundation Glasgow Cardiovascular Research Centre (BHF GCRC), Institute of Cardiovascular & Medical Sciences (ICAMS), University of Glasgow, Glasgow G12 8TA, UK; (J.V.K.); (E.D.)
| | - Martina Tetti
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Torino, 10124 Torino, Italy; (A.P.); (M.T.); (T.A.W.)
| | - Udo F. H. Engelke
- Department of Laboratory Medicine, Translational Metabolic Laboratory, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
| | - Zoran Erlic
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ), University of Zurich (UZH), 8006 Zurich, Switzerland; (Z.E.); (F.B.)
| | - Jasper Engel
- Biometris, Wageningen University & Research, 6708 PB Wageningen, The Netherlands;
| | - Timo Deutschbein
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, 97080 Würzburg, Germany; (T.D.); (M.F.); (H.R.); (M.K.)
- Medicover Oldenburg MVZ, 26122 Oldenburg, Germany
| | - Svenja Nölting
- Department of Medicine IV, University Hospital, LMU Munich, 80336 Munich, Germany;
| | - Aleksander Prejbisz
- Department of Hypertension, Institute of Cardiology, 04-628 Warsaw, Poland; (A.P.); (A.J.)
| | - Susan Richter
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus at the Technische Universität Dresden, 01307 Dresden, Germany;
| | - Jerzy Adamski
- Research Unit Molecular Endocrinology and Metabolism, Genome Analysis Center, Helmholtz Center München, German Research Center for Environmental Health, 85764 Neuherberg, Germany;
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
- Institute of Experimental Genetics, Technical University München, 85350 Freising-Weihenstephan, Germany
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 119077 Singapore, Singapore
| | - Andrzej Januszewicz
- Department of Hypertension, Institute of Cardiology, 04-628 Warsaw, Poland; (A.P.); (A.J.)
| | - Filippo Ceccato
- Endocrinology Unit, Department of Medicine DIMED, University-Hospital of Padova, 35128 Padova, Italy; (F.C.); (C.S.)
| | - Carla Scaroni
- Endocrinology Unit, Department of Medicine DIMED, University-Hospital of Padova, 35128 Padova, Italy; (F.C.); (C.S.)
| | - Michael C. Dennedy
- The Discipline of Pharmacology and Therapeutics, School of Medicine, National University of Ireland, H91 CF50 Galway, Ireland;
| | - Tracy A. Williams
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Torino, 10124 Torino, Italy; (A.P.); (M.T.); (T.A.W.)
| | - Livia Lenzini
- Department of Medicine-DIMED, Emergency and Hypertension Unit, University of Padova, University Hospital, 35126 Padova, Italy;
| | - Anne-Paule Gimenez-Roqueplo
- INSERM, PARCC, Université de Paris, 75015 Paris, France; (A.-P.G.-R.); (M.-C.Z.)
- Service de Genétique, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, 75015 Paris, France
| | - Eleanor Davies
- British Heart Foundation Glasgow Cardiovascular Research Centre (BHF GCRC), Institute of Cardiovascular & Medical Sciences (ICAMS), University of Glasgow, Glasgow G12 8TA, UK; (J.V.K.); (E.D.)
| | - Martin Fassnacht
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, 97080 Würzburg, Germany; (T.D.); (M.F.); (H.R.); (M.K.)
- Core Unit Clinical Mass Spectrometry, University Hospital Würzburg, 97080 Würzburg, Germany
- Comprehensive Cancer Center Mainfranken, Würzburg University, 97070 Würzburg, Germany
| | - Hanna Remde
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, 97080 Würzburg, Germany; (T.D.); (M.F.); (H.R.); (M.K.)
| | - Graeme Eisenhofer
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (K.L.); (C.P.); (G.E.)
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus at the Technische Universität Dresden, 01307 Dresden, Germany;
| | - Felix Beuschlein
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ), University of Zurich (UZH), 8006 Zurich, Switzerland; (Z.E.); (F.B.)
- Department of Medicine IV, University Hospital, LMU Munich, 80336 Munich, Germany;
| | - Matthias Kroiss
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, 97080 Würzburg, Germany; (T.D.); (M.F.); (H.R.); (M.K.)
- Department of Medicine IV, University Hospital, LMU Munich, 80336 Munich, Germany;
- Core Unit Clinical Mass Spectrometry, University Hospital Würzburg, 97080 Würzburg, Germany
- Comprehensive Cancer Center Mainfranken, Würzburg University, 97070 Würzburg, Germany
| | - Emily Jefferson
- Division of Population Health and Genomics, School of Medicine, University of Dundee, Dundee DD2 4BF, UK; (P.R.); (S.R.); (E.J.)
- Institute of Health & Wellbeing, Glasgow University, Glasgow G12 8RZ, UK
| | - Maria-Christina Zennaro
- INSERM, PARCC, Université de Paris, 75015 Paris, France; (A.-P.G.-R.); (M.-C.Z.)
- Service de Genétique, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, 75015 Paris, France
| | - Ron A. Wevers
- Department of Laboratory Medicine, Translational Metabolic Laboratory, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
| | - Jeroen J. Jansen
- Department of Analytical Chemistry, Institute for Molecules and Materials, Radboud University, 6500 HB Nijmegen, The Netherlands; (G.H.T.); (J.J.J.)
| | - Jaap Deinum
- Department of Internal Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
| | - Henri J. L. M. Timmers
- Department of Internal Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
| |
Collapse
|
10
|
Huang HC, Wu Y, Yang Q, Qin LX. PRECISION.array: An R Package for Benchmarking microRNA Array Data Normalization in the Context of Sample Classification. Front Genet 2022; 13:838679. [PMID: 35938023 PMCID: PMC9354575 DOI: 10.3389/fgene.2022.838679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 06/10/2022] [Indexed: 11/13/2022] Open
Abstract
We present a new R package PRECISION.array for assessing the performance of data normalization methods in connection with methods for sample classification. It includes two microRNA microarray datasets for the same set of tumor samples: a re-sampling-based algorithm for simulating additional paired datasets under various designs of sample-to-array assignment and levels of signal-to-noise ratios and a collection of numerical and graphical tools for method performance assessment. The package allows users to specify their own methods for normalization and classification, in addition to implementing three methods for training data normalization, seven methods for test data normalization, seven methods for classifier training, and two methods for classifier validation. It enables an objective and systemic evaluation of the operating characteristics of normalization and classification methods in microRNA microarrays. To our knowledge, this is the first such tool available. The R package can be downloaded freely at https://github.com/LXQin/PRECISION.array.
Collapse
|
11
|
Lamidi S, Williams KM, Hind D, Peckham-Cooper A, Miller AS, Smith AM, Saha A, Macutkiewicz C, Griffiths EA, Catena F, Coccolini F, Toogood G, Tierney GM, Boyd-Carson H, Sartelli M, Blencowe NS, Lockwood S, Coe PO, Lee MJ, Barreto SG, Drake T, Gachabayov M, Hill J, Ioannidis O, Lostoridis E, Mehraj A, Negoi I, Pata F, Steenkamp C, Ahmed S, Alin V, Al-Rashedy M, Atici SD, Bains L, Bandyopadhyay SK, Baraket O, Bates T, Beral D, Brown L, Buonomo L, Burke D, Caravaglios G, Ceresoli M, Chapman SJ, Cillara N, Clarke R, Colak E, Daniels S, Demetrashvili Z, Di Carlo I, Duff S, Dziakova J, Elliott JA, El Zalabany T, Engledow A, Ewnte B, Fraga GP, George R, Giuffrida M, Glasbey J, Isik A, Kechagias A, Kenington C, Kessel B, Khokha V, Kong V, Laloë P, Litvin A, Lostoridis E, Marinis A, Martínez-Pérez A, Menzies D, Mills R, Monzon BI, Morgan R, Neri V, Nita GE, Perra T, Perrone G, Porcu A, Poskus T, Premnath S, Sall I, Sarma DR, Slavchev M, Spence G, Tarasconi A, Tolonen M, Toro A, Venn ML, Vimalachandran D, Wheldon L, Zakaria AD. Defining core patient descriptors for perforated peptic ulcer research: international Delphi. Br J Surg 2022; 109:603-609. [PMID: 35467718 DOI: 10.1093/bjs/znac096] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/09/2022] [Accepted: 03/08/2022] [Indexed: 10/13/2023]
Abstract
BACKGROUND Perforated peptic ulcer (PPU) remains a common condition globally with significant morbidity and mortality. Previous work has demonstrated variation in reporting of patient characteristics in PPU studies, making comparison of studies and outcomes difficult. The aim of this study was to standardize the reporting of patient characteristics, by creating a core descriptor set (CDS) of important descriptors that should be consistently reported in PPU research. METHODS Candidate descriptors were identified through systematic review and stakeholder proposals. An international Delphi exercise involving three survey rounds was undertaken to obtain consensus on key patient characteristics for future research. Participants rated items on a scale of 1-9 with respect to their importance. Items meeting a predetermined threshold (rated 7-9 by over 70 per cent of stakeholders) were included in the final set and ratified at a consensus meeting. Feedback was provided between rounds to allow refinement of ratings. RESULTS Some 116 clinicians were recruited from 29 countries. A total of 63 descriptors were longlisted from the literature, and 27 were proposed by stakeholders. After three survey rounds and a consensus meeting, 27 descriptors were included in the CDS. These covered demographic variables and co-morbidities, risk factors for PPU, presentation and pathway factors, need for organ support, biochemical parameters, prognostic tools, perforation details, and surgical history. CONCLUSION This study defines the core descriptive items for PPU research, which will allow more robust synthesis of studies.
Collapse
|
12
|
Dayon L, Cominetti O, Affolter M. Proteomics of Human Biological Fluids for Biomarker Discoveries: Technical Advances and Recent Applications. Expert Rev Proteomics 2022; 19:131-151. [PMID: 35466824 DOI: 10.1080/14789450.2022.2070477] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Biological fluids are routine samples for diagnostic testing and monitoring. Blood samples are typically measured because of their moderate collection invasiveness and high information content on health and disease. Several body fluids, such as cerebrospinal fluid (CSF), are also studied and suited to specific pathologies. Over the last two decades proteomics has quested to identify protein biomarkers but with limited success. Recent technologies and refined pipelines have accelerated the profiling of human biological fluids. AREAS COVERED We review proteomic technologies for the identification of biomarkers. Those are based on antibodies/aptamers arrays or mass spectrometry (MS), but new ones are emerging. Advances in scalability and throughput have allowed to better design studies and cope with the limited sample size that had until now prevailed due to technological constraints. With these enablers, plasma/serum, CSF, saliva, tears, urine, and milk proteomes have been further profiled; we provide a non-exhaustive picture of some recent highlights (mainly covering literature from last five years in the Scopus database) using MS-based proteomics. EXPERT OPINION While proteomics has been in the shadow of genomics for years, proteomic tools and methodologies have reached a certain maturity. They are better suited to discover innovative and robust biofluid biomarkers.
Collapse
Affiliation(s)
- Loïc Dayon
- Proteomics, Nestlé Institute of Food Safety & Analytical Sciences, Nestlé Research, CH-1015 Lausanne, Switzerland.,Institut des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Ornella Cominetti
- Proteomics, Nestlé Institute of Food Safety & Analytical Sciences, Nestlé Research, CH-1015 Lausanne, Switzerland
| | - Michael Affolter
- Proteomics, Nestlé Institute of Food Safety & Analytical Sciences, Nestlé Research, CH-1015 Lausanne, Switzerland
| |
Collapse
|
13
|
Düren Y, Lederer J, Qin LX. OUP accepted manuscript. Nucleic Acids Res 2022; 50:e56. [PMID: 35188574 PMCID: PMC9177987 DOI: 10.1093/nar/gkac064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 01/03/2022] [Accepted: 02/08/2022] [Indexed: 11/22/2022] Open
Abstract
Deep sequencing has become one of the most popular tools for transcriptome profiling in biomedical studies. While an abundance of computational methods exists for ‘normalizing’ sequencing data to remove unwanted between-sample variations due to experimental handling, there is no consensus on which normalization is the most suitable for a given data set. To address this problem, we developed ‘DANA’—an approach for assessing the performance of normalization methods for microRNA sequencing data based on biology-motivated and data-driven metrics. Our approach takes advantage of well-known biological features of microRNAs for their expression pattern and chromosomal clustering to simultaneously assess (i) how effectively normalization removes handling artifacts and (ii) how aptly normalization preserves biological signals. With DANA, we confirm that the performance of eight commonly used normalization methods vary widely across different data sets and provide guidance for selecting a suitable method for the data at hand. Hence, it should be adopted as a routine preprocessing step (preceding normalization) for microRNA sequencing data analysis. DANA is implemented in R and publicly available at https://github.com/LXQin/DANA.
Collapse
Affiliation(s)
- Yannick Düren
- Department of Mathematical Statistics, Ruhr-University Bochum, Universitätsstraße 150, 44801 Bochum, Germany
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Johannes Lederer
- Department of Mathematical Statistics, Ruhr-University Bochum, Universitätsstraße 150, 44801 Bochum, Germany
| | - Li-Xuan Qin
- To whom correspondence should be addressed. Tel: +1 646 888 8251; Fax: +1 646 888 0010;
| |
Collapse
|
14
|
Mortezaei Z. Computational methods for analyzing RNA-sequencing contaminated samples and its impact on cancer genome studies. INFORMATICS IN MEDICINE UNLOCKED 2022. [DOI: 10.1016/j.imu.2022.101054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
15
|
Guest PC, Popovic D, Steiner J. Challenges of Multiplex Assays for COVID-19 Research: A Machine Learning Perspective. Methods Mol Biol 2022; 2511:37-50. [PMID: 35838950 DOI: 10.1007/978-1-0716-2395-4_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Multiplex assays that provide simultaneous measurement of multiple analytes in biological samples have now developed into widely used technologies in the study of diseases, drug discovery, and other medical areas. These approaches span multiple assay systems and can provide readouts of specific assay components with similar accuracy as the respective single assay measurements. Multiplexing allows the consumption of lower sample volumes, lower costs, and higher throughput compared with carrying out single assays. A number of recent studies have demonstrated the impact of multiplex assays in the study of the SARS-CoV-2 virus, the infectious agent responsible for the current COVID-19 pandemic. In this respect, machine learning techniques have proven to be highly valuable in capturing complex disease phenotypes and converting these insights into models which can be applied in real-world settings. This chapter gives an overview of opportunities and challenges of multiplexed biomarker analysis, with a focus on the use of machine learning aimed at identification of biological signatures for increasing our understanding of COVID-19 disease, and for improved diagnostics and prediction of disease outcomes.
Collapse
Affiliation(s)
- Paul C Guest
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil.
| | - David Popovic
- Section of Forensic Psychiatry, Department of Psychiatry and Psychotherapy, Ludwig-Maximilian-University, Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Munich, Germany
| | - Johann Steiner
- Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- Center for Behavioral Brain Sciences, Magdeburg, Germany
- German Center for Mental Health (DZP), Center for Intervention and Research on adaptive and maladaptive brain Circuits underlying mental health (C-I-R-C), Site Jena-Magdeburg-Halle, Magdeburg, Germany
| |
Collapse
|
16
|
Van Den Bosch T, van Dijk E, Vermeulen L, Miedema DM. Predicting survival of cancer patients by chromosomal copy number heterogeneity. Mol Cell Oncol 2021; 8:1949956. [PMID: 34616875 PMCID: PMC8489936 DOI: 10.1080/23723556.2021.1949956] [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] [Indexed: 10/24/2022]
Abstract
We recently introduced a method to derive intra-tumor heterogeneity (ITH) from a single copy number measurement. This method stratifies patients for survival and could potentially help to identify low and high-risk patients with clinical relevance.
Collapse
Affiliation(s)
- Tom Van Den Bosch
- LEXOR, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam and Amsterdam Gastroenterology & Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Oncode Institute, Amsterdam, The Netherlands
| | - Erik van Dijk
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Louis Vermeulen
- LEXOR, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam and Amsterdam Gastroenterology & Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Oncode Institute, Amsterdam, The Netherlands
| | - Daniël M Miedema
- LEXOR, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam and Amsterdam Gastroenterology & Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Oncode Institute, Amsterdam, The Netherlands
| |
Collapse
|
17
|
Wu Y, Huang HC, Qin LX. Making External Validation Valid for Molecular Classifier Development. JCO Precis Oncol 2021; 5:PO.21.00103. [PMID: 34377885 PMCID: PMC8345919 DOI: 10.1200/po.21.00103] [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: 03/06/2021] [Revised: 05/18/2021] [Accepted: 07/06/2021] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Accurate assessment of a molecular classifier that guides patient care is of paramount importance in precision oncology. Recent years have seen an increasing use of external validation for such assessment. However, little is known about how it is affected by ubiquitous unwanted variations in test data because of disparate experimental handling and by the use of data normalization for alleviating such variations. METHODS In this paper, we studied these issues using two microarray data sets for the same set of tumor samples and additional data simulated by resampling under various levels of signal-to-noise ratio and different designs for array-to-sample allocation. RESULTS We showed that (1) unwanted variations can lead to biased classifier assessment and (2) data normalization mitigates the bias to varying extents depending on the specific method used. In particular, frozen normalization methods for test data outperform their conventional forms in terms of both reducing the bias in accuracy estimation and increasing robustness to handling effects. We make available our benchmarking tool as an R package on GitHub for performing such evaluation on additional methods for normalization and classification. CONCLUSION Our findings thus highlight the importance of proper test-data normalization for valid assessment by external validation and call for caution on the choice of normalization method for molecular classifier development.
Collapse
Affiliation(s)
- Yilin Wu
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Huei-Chung Huang
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Li-Xuan Qin
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
18
|
Kleppe A, Skrede OJ, De Raedt S, Liestøl K, Kerr DJ, Danielsen HE. Designing deep learning studies in cancer diagnostics. Nat Rev Cancer 2021; 21:199-211. [PMID: 33514930 DOI: 10.1038/s41568-020-00327-9] [Citation(s) in RCA: 161] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/09/2020] [Indexed: 12/16/2022]
Abstract
The number of publications on deep learning for cancer diagnostics is rapidly increasing, and systems are frequently claimed to perform comparable with or better than clinicians. However, few systems have yet demonstrated real-world medical utility. In this Perspective, we discuss reasons for the moderate progress and describe remedies designed to facilitate transition to the clinic. Recent, presumably influential, deep learning studies in cancer diagnostics, of which the vast majority used images as input to the system, are evaluated to reveal the status of the field. By manipulating real data, we then exemplify that much and varied training data facilitate the generalizability of neural networks and thus the ability to use them clinically. To reduce the risk of biased performance estimation of deep learning systems, we advocate evaluation in external cohorts and strongly advise that the planned analyses, including a predefined primary analysis, are described in a protocol preferentially stored in an online repository. Recommended protocol items should be established for the field, and we present our suggestions.
Collapse
Affiliation(s)
- Andreas Kleppe
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
- Department of Informatics, University of Oslo, Oslo, Norway
| | - Ole-Johan Skrede
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
- Department of Informatics, University of Oslo, Oslo, Norway
| | - Sepp De Raedt
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
- Department of Informatics, University of Oslo, Oslo, Norway
| | - Knut Liestøl
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
- Department of Informatics, University of Oslo, Oslo, Norway
| | - David J Kerr
- Nuffield Division of Clinical Laboratory Sciences, University of Oxford, Oxford, UK
| | - Håvard E Danielsen
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway.
- Department of Informatics, University of Oslo, Oslo, Norway.
- Nuffield Division of Clinical Laboratory Sciences, University of Oxford, Oxford, UK.
| |
Collapse
|
19
|
Biomarker Discovery and Validation: Statistical Considerations. J Thorac Oncol 2021; 16:537-545. [PMID: 33545385 DOI: 10.1016/j.jtho.2021.01.1616] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 01/04/2021] [Accepted: 01/07/2021] [Indexed: 12/23/2022]
Abstract
Biomarkers have various applications including disease detection, diagnosis, prognosis, prediction of response to intervention, and disease monitoring. In this era of precision medicine, having validated biomarkers to inform clinical decision making is more important than ever. In this article, we discuss best the practices and potential issues in biomarker discovery and validation. We encourage team science partnerships to bring cutting-edge discovery from bench to bedside, leading to improved patient care and outcomes.
Collapse
|
20
|
Plebani M. “Omics” translation: a challenge for laboratory medicine. PRINCIPLES OF TRANSLATIONAL SCIENCE IN MEDICINE 2021:21-32. [DOI: 10.1016/b978-0-12-820493-1.00021-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
21
|
Liu Y, Kaur S, Huang Y, Fahrmann JF, Rinaudo JA, Hanash SM, Batra SK, Singhi AD, Brand RE, Maitra A, Haab BB. Biomarkers and Strategy to Detect Preinvasive and Early Pancreatic Cancer: State of the Field and the Impact of the EDRN. Cancer Epidemiol Biomarkers Prev 2020; 29:2513-2523. [PMID: 32532830 PMCID: PMC7710622 DOI: 10.1158/1055-9965.epi-20-0161] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/20/2020] [Accepted: 06/05/2020] [Indexed: 12/19/2022] Open
Abstract
Patients afflicted with pancreatic ductal adenocarcinoma (PDAC) face a dismal prognosis, but headway could be made if physicians could identify the disease earlier. A compelling strategy to broaden the use of surveillance for PDAC is to incorporate molecular biomarkers in combination with clinical analysis and imaging tools. This article summarizes the components involved in accomplishing biomarker validation and an analysis of the requirements of molecular biomarkers for disease surveillance. We highlight the significance of consortia for this research and highlight resources and infrastructure of the Early Detection Research Network (EDRN). The EDRN brings together the multifaceted expertise and resources needed for biomarker validation, such as study design, clinical care, biospecimen collection and handling, molecular technologies, and biostatistical analysis, and studies coming out of the EDRN have yielded biomarkers that are moving forward in validation. We close the article with an overview of the current investigational biomarkers, an analysis of their performance relative to the established benchmarks, and an outlook on the current needs in the field. The outlook for improving the early detection of PDAC looks promising, and the pace of further research should be quickened through the resources and expertise of the EDRN and other consortia.See all articles in this CEBP Focus section, "NCI Early Detection Research Network: Making Cancer Detection Possible."
Collapse
Affiliation(s)
- Ying Liu
- Van Andel Institute, Grand Rapids, Michigan
| | | | - Ying Huang
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Johannes F Fahrmann
- Sheikh Ahmed Center for Pancreatic Cancer Research, MD Anderson Cancer Center, Houston, Texas
| | - Jo Ann Rinaudo
- Division of Cancer Prevention, National Cancer Institute, Bethesda, Maryland
| | - Samir M Hanash
- Sheikh Ahmed Center for Pancreatic Cancer Research, MD Anderson Cancer Center, Houston, Texas
| | | | - Aatur D Singhi
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Randall E Brand
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Anirban Maitra
- Sheikh Ahmed Center for Pancreatic Cancer Research, MD Anderson Cancer Center, Houston, Texas
| | | |
Collapse
|
22
|
Accelerating protein biomarker discovery and translation from proteomics research for clinical utility. Bioanalysis 2020; 12:1469-1481. [DOI: 10.4155/bio-2020-0198] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Discovery proteomics research has made significant progress in the past several years; however, the number of protein biomarkers deployed in clinical practice remains rather limited. There are several scientific and procedural gaps between discovery proteomics research and clinical implementation, which have contributed to poor biomarker validity and few clinical applications. The complexity and low throughput of proteomics approaches have added additional barriers for biomarker assay translation to clinical applications. Recently, targeted proteomics have become a powerful tool to bridge the biomarker discovery to clinical validation. In this perspective, we discuss the challenges and strategies in proteomics research from a clinical perspective, and propose several recommendations for discovery proteomics research to accelerate protein biomarker discovery and translation for future clinical applications.
Collapse
|
23
|
Challenges and Opportunities in Clinical Applications of Blood-Based Proteomics in Cancer. Cancers (Basel) 2020; 12:cancers12092428. [PMID: 32867043 PMCID: PMC7564506 DOI: 10.3390/cancers12092428] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/23/2020] [Accepted: 08/25/2020] [Indexed: 12/12/2022] Open
Abstract
Simple Summary The traditional approach in identifying cancer related protein biomarkers has focused on evaluation of a single peptide/protein in tissue or circulation. At best, this approach has had limited success for clinical applications, since multiple pathological tumor pathways may be involved during initiation or progression of cancer which diminishes the significance of a single candidate protein/peptide. Emerging sensitive proteomic based technologies like liquid chromatography mass spectrometry (LC-MS)-based quantitative proteomics can provide a platform for evaluating serial serum or plasma samples to interrogate secreted products of tumor–host interactions, thereby revealing a more “complete” repertoire of biological variables encompassing heterogeneous tumor biology. However, several challenges need to be met for successful application of serum/plasma based proteomics. These include uniform pre-analyte processing of specimens, sensitive and specific proteomic analytical platforms and adequate attention to study design during discovery phase followed by validation of discovery-level signatures for prognostic, predictive, and diagnostic cancer biomarker applications. Abstract Blood is a readily accessible biofluid containing a plethora of important proteins, nucleic acids, and metabolites that can be used as clinical diagnostic tools in diseases, including cancer. Like the on-going efforts for cancer biomarker discovery using the liquid biopsy detection of circulating cell-free and cell-based tumor nucleic acids, the circulatory proteome has been underexplored for clinical cancer biomarker applications. A comprehensive proteome analysis of human serum/plasma with high-quality data and compelling interpretation can potentially provide opportunities for understanding disease mechanisms, although several challenges will have to be met. Serum/plasma proteome biomarkers are present in very low abundance, and there is high complexity involved due to the heterogeneity of cancers, for which there is a compelling need to develop sensitive and specific proteomic technologies and analytical platforms. To date, liquid chromatography mass spectrometry (LC-MS)-based quantitative proteomics has been a dominant analytical workflow to discover new potential cancer biomarkers in serum/plasma. This review will summarize the opportunities of serum proteomics for clinical applications; the challenges in the discovery of novel biomarkers in serum/plasma; and current proteomic strategies in cancer research for the application of serum/plasma proteomics for clinical prognostic, predictive, and diagnostic applications, as well as for monitoring minimal residual disease after treatments. We will highlight some of the recent advances in MS-based proteomics technologies with appropriate sample collection, processing uniformity, study design, and data analysis, focusing on how these integrated workflows can identify novel potential cancer biomarkers for clinical applications.
Collapse
|
24
|
Ren AH, Fiala CA, Diamandis EP, Kulasingam V. Pitfalls in Cancer Biomarker Discovery and Validation with Emphasis on Circulating Tumor DNA. Cancer Epidemiol Biomarkers Prev 2020; 29:2568-2574. [PMID: 32277003 DOI: 10.1158/1055-9965.epi-20-0074] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/19/2020] [Accepted: 04/03/2020] [Indexed: 11/16/2022] Open
Abstract
Despite significant investment of funds and resources, few new cancer biomarkers have been introduced to the clinic in the last few decades. Although many candidates produce promising results in the laboratory, deficiencies in sensitivity, specificity, and predictive value make them less than desirable in a patient setting. This review will analyze these challenges in detail as well as discuss false discovery, problems with reproducibility, and tumor heterogeneity. Circulating tumor DNA (ctDNA), an emerging cancer biomarker, is also analyzed, particularly in the contexts of assay specificity, sensitivity, fragmentation, lead time, mutant allele fraction, and clinical relevance. Emerging artificial intelligence technologies will likely be valuable tools in maximizing the clinical utility of ctDNA which is often found in very small quantities in patients with early-stage tumors. Finally, the implications of challenging false discoveries are examined and some insights about improving cancer biomarker discovery are provided.See all articles in this CEBP Focus section, "NCI Early Detection Research Network: Making Cancer Detection Possible."
Collapse
Affiliation(s)
- Annie H Ren
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Clare A Fiala
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Eleftherios P Diamandis
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Department of Clinical Biochemistry, University Health Network, Toronto, Ontario, Canada
| | - Vathany Kulasingam
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada. .,Department of Clinical Biochemistry, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
25
|
Affiliation(s)
- Tri Le
- Department of Mathematics, Science, and InformaticsMercer University Atlanta Georgia
| | - Bertrand Clarke
- Department of StatisticsUniversity of Nebraska‐Lincoln Lincoln Nebraska
| |
Collapse
|
26
|
Abstract
Background Biomarker discovery studies have generated an array of omic data, however few novel biomarkers have reached clinical use. Guidelines for rigorous study designs are needed. Content Biases frequently occur in sample selection, outcome ascertainment, or unblinded sample handling and assaying process. The principles of a prospective-specimen collection and retrospective-blinded-evaluation (PRoBE) design can be adapted to mitigate various sources of biases in discovery. We recommend establishing quality biospecimen repositories using matched two-phase designs to minimize biases and maximize efficiency. We also highlight the importance of taking the clinical context into consideration in both sample selection and power calculation for discovery studies. Summary Biomarker discovery research should follow rigorous design principles in sample se- lection to avoid biases. Consideration of clinical application and the corresponding biomarker performance characteristics in study designs will lead to a more fruitful discovery study. Impact Appropriate study designs will improve the quality and clinical rigor of biomarker discovery studies.
Collapse
Affiliation(s)
- Yingye Zheng
- Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N., M2-B500, Seattle, Washington 98109, ,
| |
Collapse
|
27
|
Čuklina J, Pedrioli PGA, Aebersold R. Review of Batch Effects Prevention, Diagnostics, and Correction Approaches. Methods Mol Biol 2020; 2051:373-387. [PMID: 31552638 DOI: 10.1007/978-1-4939-9744-2_16] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Systematic technical variation in high-throughput studies consisting of the serial measurement of large sample cohorts is known as batch effects. Batch effects reduce the sensitivity of biological signal extraction and can cause significant artifacts. The systematic bias in the data caused by batch effects is more common in studies in which logistical considerations restrict the number of samples that can be prepared or profiled in a single experiment, thus necessitating the arrangement of subsets of study samples in batches. To mitigate the negative impact of batch effects, statistical approaches for batch correction are used at the stage of experimental design and data processing. Whereas in genomics batch effects and possible remedies have been extensively discussed, they are a relatively new challenge in proteomics because methods with sufficient throughput to systematically measure through large sample cohorts have only recently become available. Here we provide general recommendations to mitigate batch effects: we discuss the design of large-scale proteomic studies, review the most commonly used tools for batch effect correction and overview their application in proteomics.
Collapse
Affiliation(s)
- Jelena Čuklina
- Department of Biology, Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland
- Ph.D. Program in Systems Biology, University of Zurich and ETH Zurich, Zürich, Switzerland
| | - Patrick G A Pedrioli
- Department of Biology, Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland
- ETH Zürich, PHRT-MS, Zürich, Switzerland
| | - Ruedi Aebersold
- Department of Biology, Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland.
- Faculty of Science, University of Zürich, Zürich, Switzerland.
| |
Collapse
|
28
|
Grigorieva J, Asmellash S, Oliveira C, Roder H, Net L, Roder J. Application of protein set enrichment analysis to correlation of protein functional sets with mass spectral features and multivariate proteomic tests. CLINICAL MASS SPECTROMETRY 2020. [DOI: 10.1016/j.clinms.2019.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
29
|
Faes L, Wagner SK, Fu DJ, Liu X, Korot E, Ledsam JR, Back T, Chopra R, Pontikos N, Kern C, Moraes G, Schmid MK, Sim D, Balaskas K, Bachmann LM, Denniston AK, Keane PA. Automated deep learning design for medical image classification by health-care professionals with no coding experience: a feasibility study. LANCET DIGITAL HEALTH 2019; 1:e232-e242. [DOI: 10.1016/s2589-7500(19)30108-6] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 08/05/2019] [Accepted: 08/08/2019] [Indexed: 12/13/2022]
|
30
|
Walter FM, Thompson MJ, Wellwood I, Abel GA, Hamilton W, Johnson M, Lyratzopoulos G, Messenger MP, Neal RD, Rubin G, Singh H, Spencer A, Sutton S, Vedsted P, Emery JD. Evaluating diagnostic strategies for early detection of cancer: the CanTest framework. BMC Cancer 2019; 19:586. [PMID: 31200676 PMCID: PMC6570853 DOI: 10.1186/s12885-019-5746-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 05/23/2019] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Novel diagnostic triage and testing strategies to support early detection of cancer could improve clinical outcomes. Most apparently promising diagnostic tests ultimately fail because of inadequate performance in real-world, low prevalence populations such as primary care or general community populations. They should therefore be systematically evaluated before implementation to determine whether they lead to earlier detection, are cost-effective, and improve patient safety and quality of care, while minimising over-investigation and over-diagnosis. METHODS We performed a systematic scoping review of frameworks for the evaluation of tests and diagnostic approaches. RESULTS We identified 16 frameworks: none addressed the entire continuum from test development to impact on diagnosis and patient outcomes in the intended population, nor the way in which tests may be used for triage purposes as part of a wider diagnostic strategy. Informed by these findings, we developed a new framework, the 'CanTest Framework', which proposes five iterative research phases forming a clear translational pathway from new test development to health system implementation and evaluation. CONCLUSION This framework is suitable for testing in low prevalence populations, where tests are often applied for triage testing and incorporated into a wider diagnostic strategy. It has relevance for a wide range of stakeholders including patients, policymakers, purchasers, healthcare providers and industry.
Collapse
Affiliation(s)
- Fiona M. Walter
- The Primary Care Unit, Department of Public Health & Primary Care, University of Cambridge, Cambridge, CB1 8RN UK
| | | | - Ian Wellwood
- The Primary Care Unit, Department of Public Health & Primary Care, University of Cambridge, Cambridge, CB1 8RN UK
| | - Gary A. Abel
- University of Exeter, St Luke’s Campus, Exeter, EX1 2LU UK
| | | | - Margaret Johnson
- The Primary Care Unit, Department of Public Health & Primary Care, University of Cambridge, Cambridge, CB1 8RN UK
| | - Georgios Lyratzopoulos
- Department of Behavioural Science and Health, Epidemiology of Cancer Healthcare and Outcomes (ECHO) Research Group, University College London, London, UK
| | - Michael P. Messenger
- National Institute of Health Research (NIHR) Leeds In Vitro Diagnostic Cooperative (IVDC), Leeds Centre for Personalised Medicine and Health, University of Leeds, Leeds, UK
| | - Richard D. Neal
- Academic Unit of Primary Care, Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
| | - Greg Rubin
- Institute of Health and Society, University of Newcastle, Sir James Spence Institute, Royal Victoria Infirmary, Newcastle, NE1 4LP UK
| | - Hardeep Singh
- Center for Innovations in Quality, Effectiveness and Safety, Michael E. DeBakey Veterans Affairs Medical Center and Baylor College of Medicine, Houston, TX USA
| | - Anne Spencer
- Health Economics Group, University of Exeter, St Luke’s Campus, Exeter, EX1 2LU Devon UK
| | - Stephen Sutton
- The Primary Care Unit, Department of Public Health & Primary Care, University of Cambridge, Cambridge, CB1 8RN UK
| | - Peter Vedsted
- Research Centre for Cancer Diagnosis – CaP, The Research Unit for General Practice and Research Clinic for Innovative Health Care Delivery, Department of Clinical Medicine, Aarhus University, Bartholins Alle 2, 8000 Aarhus, Denmark
| | - Jon D. Emery
- Centre for Cancer Research and Department of General Practice, University of Melbourne, 10th floor, Victorian Comprehensive Cancer Centre, 305 Grattan St, Melbourne, VIC 3010 Australia
| |
Collapse
|
31
|
Sardela PDDO, Sardela VF, da Silva AMDS, Pereira HMG, de Aquino Neto FR. A pilot study of non-targeted screening for stimulant misuse using high-resolution mass spectrometry. Forensic Toxicol 2019. [DOI: 10.1007/s11419-019-00482-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
32
|
Findeisen P, Hemanna S, Maharjan RS, Mindt S, Costina V, Hofheinz R, Neumaier M. Mass spectrometry based analytical quality assessment of serum and plasma specimens with patterns of endo- and exogenous peptides. Clin Chem Lab Med 2019; 57:668-678. [DOI: 10.1515/cclm-2018-0811] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/05/2018] [Indexed: 12/23/2022]
Abstract
Abstract
Background
Inappropriate preanalytical sample handling is a major threat for any biomarker discovery approach. Blood specimens have a genuine proteolytic activity that leads to a time dependent decay of peptidic quality control markers (QCMs). The aim of this study was to identify QCMs for direct assessment of sample quality (DASQ) of serum and plasma specimens.
Methods
Serum and plasma specimens of healthy volunteers and tumor patients were spiked with two synthetic reporter peptides (exogenous QCMs) and aged under controlled conditions for up to 24 h. The proteolytic fragments of endogenous and exogenous QCMs were monitored for each time point by mass spectrometry (MS). The decay pattern of peptides was used for supervised classification of samples according to their respective preanalytical quality.
Results
The classification accuracy for fresh specimens (1 h) was 96% and 99% for serum and plasma specimens, respectively, when endo- and exogenous QCMs were used for the calculations. However, classification of older specimens was more difficult and overall classification accuracy decreased to 79%.
Conclusions
MALDI-TOF MS is a simple and robust method that can be used for DASQ of serum and plasma specimens in a high throughput manner. We propose DASQ as a fast and simple step that can be included in multicentric large-scale projects to ensure the homogeneity of sample quality.
Collapse
|
33
|
Hall PS, Mitchell ED, Smith AF, Cairns DA, Messenger M, Hutchinson M, Wright J, Vinall-Collier K, Corps C, Hamilton P, Meads D, Lewington A. The future for diagnostic tests of acute kidney injury in critical care: evidence synthesis, care pathway analysis and research prioritisation. Health Technol Assess 2019; 22:1-274. [PMID: 29862965 DOI: 10.3310/hta22320] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Acute kidney injury (AKI) is highly prevalent in hospital inpatient populations, leading to significant mortality and morbidity, reduced quality of life and high short- and long-term health-care costs for the NHS. New diagnostic tests may offer an earlier diagnosis or improved care, but evidence of benefit to patients and of value to the NHS is required before national adoption. OBJECTIVES To evaluate the potential for AKI in vitro diagnostic tests to enhance the NHS care of patients admitted to the intensive care unit (ICU) and identify an efficient supporting research strategy. DATA SOURCES We searched ClinicalTrials.gov, The Cochrane Library databases, Embase, Health Management Information Consortium, International Clinical Trials Registry Platform, MEDLINE, metaRegister of Current Controlled Trials, PubMed and Web of Science databases from their inception dates until September 2014 (review 1), November 2015 (review 2) and July 2015 (economic model). Details of databases used for each review and coverage dates are listed in the main report. REVIEW METHODS The AKI-Diagnostics project included horizon scanning, systematic reviewing, meta-analysis of sensitivity and specificity, appraisal of analytical validity, care pathway analysis, model-based lifetime economic evaluation from a UK NHS perspective and value of information (VOI) analysis. RESULTS The horizon-scanning search identified 152 potential tests and biomarkers. Three tests, Nephrocheck® (Astute Medical, Inc., San Diego, CA, USA), NGAL and cystatin C, were subjected to detailed review. The meta-analysis was limited by variable reporting standards, study quality and heterogeneity, but sensitivity was between 0.54 and 0.92 and specificity was between 0.49 and 0.95 depending on the test. A bespoke critical appraisal framework demonstrated that analytical validity was also poorly reported in many instances. In the economic model the incremental cost-effectiveness ratios ranged from £11,476 to £19,324 per quality-adjusted life-year (QALY), with a probability of cost-effectiveness between 48% and 54% when tests were compared with current standard care. LIMITATIONS The major limitation in the evidence on tests was the heterogeneity between studies in the definitions of AKI and the timing of testing. CONCLUSIONS Diagnostic tests for AKI in the ICU offer the potential to improve patient care and add value to the NHS, but cost-effectiveness remains highly uncertain. Further research should focus on the mechanisms by which a new test might change current care processes in the ICU and the subsequent cost and QALY implications. The VOI analysis suggested that further observational research to better define the prevalence of AKI developing in the ICU would be worthwhile. A formal randomised controlled trial of biomarker use linked to a standardised AKI care pathway is necessary to provide definitive evidence on whether or not adoption of tests by the NHS would be of value. STUDY REGISTRATION The systematic review within this study is registered as PROSPERO CRD42014013919. FUNDING The National Institute for Health Research Health Technology Assessment programme.
Collapse
Affiliation(s)
- Peter S Hall
- Edinburgh Cancer Research Centre, University of Edinburgh, Edinburgh, UK
| | | | - Alison F Smith
- Academy of Primary Care, Hull York Medical School, Hull, UK.,National Institute for Health Research (NIHR) Diagnostic Evidence Co-operative Leeds, Leeds, UK
| | - David A Cairns
- Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, UK
| | - Michael Messenger
- National Institute for Health Research (NIHR) Diagnostic Evidence Co-operative Leeds, Leeds, UK
| | | | - Judy Wright
- Academy of Primary Care, Hull York Medical School, Hull, UK
| | | | | | - Patrick Hamilton
- Manchester Institute of Nephrology and Transplantation, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - David Meads
- Academy of Primary Care, Hull York Medical School, Hull, UK
| | | |
Collapse
|
34
|
Shortcomings in the evaluation of biomarkers in ovarian cancer: a systematic review. ACTA ACUST UNITED AC 2019; 58:3-10. [DOI: 10.1515/cclm-2019-0038] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 03/09/2019] [Indexed: 12/22/2022]
Abstract
Abstract
Background
Shortcomings in study design have been hinted at as one of the possible causes of failures in the translation of discovered biomarkers into the care of ovarian cancer patients, but systematic assessments of biomarker studies are scarce. We aimed to document study design features of recently reported evaluations of biomarkers in ovarian cancer.
Methods
We performed a systematic search in PubMed (MEDLINE) for reports of studies evaluating the clinical performance of putative biomarkers in ovarian cancer. We extracted data on study designs and characteristics.
Results
Our search resulted in 1026 studies; 329 (32%) were found eligible after screening, of which we evaluated the first 200. Of these, 93 (47%) were single center studies. Few studies reported eligibility criteria (17%), sampling methods (10%) or a sample size justification or power calculation (3%). Studies often used disjoint groups of patients, sometimes with extreme phenotypic contrasts; 46 studies included healthy controls (23%), but only five (3%) had exclusively included advanced stage cases.
Conclusions
Our findings confirm the presence of suboptimal features in clinical evaluations of ovarian cancer biomarkers. This may lead to premature claims about the clinical value of these markers or, alternatively, the risk of discarding potential biomarkers that are urgently needed.
Collapse
|
35
|
Swami U, Nussenzveig RH, Haaland B, Agarwal N. Revisiting AJCC TNM staging for renal cell carcinoma: quest for improvement. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:S18. [PMID: 31032299 DOI: 10.21037/atm.2019.01.50] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Umang Swami
- Department of Hematology, Oncology and Blood and Marrow Transplantation, The Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Roberto H Nussenzveig
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Benjamin Haaland
- Department of Population Health Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Neeraj Agarwal
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
36
|
Allahyar A, Ubels J, de Ridder J. A data-driven interactome of synergistic genes improves network-based cancer outcome prediction. PLoS Comput Biol 2019; 15:e1006657. [PMID: 30726216 PMCID: PMC6380593 DOI: 10.1371/journal.pcbi.1006657] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 02/19/2019] [Accepted: 11/20/2018] [Indexed: 12/13/2022] Open
Abstract
Robustly predicting outcome for cancer patients from gene expression is an important challenge on the road to better personalized treatment. Network-based outcome predictors (NOPs), which considers the cellular wiring diagram in the classification, hold much promise to improve performance, stability and interpretability of identified marker genes. Problematically, reports on the efficacy of NOPs are conflicting and for instance suggest that utilizing random networks performs on par to networks that describe biologically relevant interactions. In this paper we turn the prediction problem around: instead of using a given biological network in the NOP, we aim to identify the network of genes that truly improves outcome prediction. To this end, we propose SyNet, a gene network constructed ab initio from synergistic gene pairs derived from survival-labelled gene expression data. To obtain SyNet, we evaluate synergy for all 69 million pairwise combinations of genes resulting in a network that is specific to the dataset and phenotype under study and can be used to in a NOP model. We evaluated SyNet and 11 other networks on a compendium dataset of >4000 survival-labelled breast cancer samples. For this purpose, we used cross-study validation which more closely emulates real world application of these outcome predictors. We find that SyNet is the only network that truly improves performance, stability and interpretability in several existing NOPs. We show that SyNet overlaps significantly with existing gene networks, and can be confidently predicted (~85% AUC) from graph-topological descriptions of these networks, in particular the breast tissue-specific network. Due to its data-driven nature, SyNet is not biased to well-studied genes and thus facilitates post-hoc interpretation. We find that SyNet is highly enriched for known breast cancer genes and genes related to e.g. histological grade and tamoxifen resistance, suggestive of a role in determining breast cancer outcome.
Collapse
Affiliation(s)
- Amin Allahyar
- Department of Genetics, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Delft Bioinformatics Lab, Faculty of Electrical Engineering, Mathematics and Computer Science, Delft University of Technology, Delft, The Netherlands
| | - Joske Ubels
- Department of Genetics, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Skyline DX, Rotterdam
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam
| | - Jeroen de Ridder
- Department of Genetics, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
37
|
Klont F, Horvatovich P, Govorukhina N, Bischoff R. Pre- and Post-analytical Factors in Biomarker Discovery. Methods Mol Biol 2019; 1959:1-22. [PMID: 30852812 DOI: 10.1007/978-1-4939-9164-8_1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The translation of promising biomarkers, which were identified in biomarker discovery experiments, to clinical assays is one of the key challenges in present-day proteomics research. Many so-called "biomarker candidates" fail to progress beyond the discovery phase, and much emphasis is placed on pre- and post-analytical variability in an attempt to provide explanations for this bottleneck in the biomarker development pipeline. With respect to such variability, there is a large number of pre- and post-analytical factors which may impact the outcomes of proteomics experiments and thus necessitate tight control. This chapter highlights some of these factors and provides guidance for addressing them on the basis of examples from previously published proteomics studies.
Collapse
Affiliation(s)
- Frank Klont
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Peter Horvatovich
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Natalia Govorukhina
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Rainer Bischoff
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
38
|
Robust prediction of response to immune checkpoint blockade therapy in metastatic melanoma. Nat Med 2018; 24:1545-1549. [PMID: 30127394 DOI: 10.1038/s41591-018-0157-9] [Citation(s) in RCA: 483] [Impact Index Per Article: 69.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 07/18/2018] [Indexed: 01/17/2023]
Abstract
Immune checkpoint blockade (ICB) therapy provides remarkable clinical gains and has been very successful in treatment of melanoma. However, only a subset of patients with advanced tumors currently benefit from ICB therapies, which at times incur considerable side effects and costs. Constructing predictors of patient response has remained a serious challenge because of the complexity of the immune response and the shortage of large cohorts of ICB-treated patients that include both 'omics' and response data. Here we build immuno-predictive score (IMPRES), a predictor of ICB response in melanoma which encompasses 15 pairwise transcriptomics relations between immune checkpoint genes. It is based on two key conjectures: (i) immune mechanisms underlying spontaneous regression in neuroblastoma can predict melanoma response to ICB, and (ii) key immune interactions can be captured via specific pairwise relations of the expression of immune checkpoint genes. IMPRES is validated on nine published datasets1-6 and on a newly generated dataset with 31 patients treated with anti-PD-1 and 10 with anti-CTLA-4, spanning 297 samples in total. It achieves an overall accuracy of AUC = 0.83, outperforming existing predictors and capturing almost all true responders while misclassifying less than half of the nonresponders. Future studies are warranted to determine the value of the approach presented here in other cancer types.
Collapse
|
39
|
Padoan A, Basso D, Zambon CF, Prayer-Galetti T, Arrigoni G, Bozzato D, Moz S, Zattoni F, Bellocco R, Plebani M. MALDI-TOF peptidomic analysis of serum and post-prostatic massage urine specimens to identify prostate cancer biomarkers. Clin Proteomics 2018; 15:23. [PMID: 30065622 PMCID: PMC6060548 DOI: 10.1186/s12014-018-9199-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 07/16/2018] [Indexed: 12/25/2022] Open
Abstract
Background Lower urinary tract symptoms (LUTS) and prostate specific antigen-based parameters seem to have only a limited utility for the differential diagnosis of prostate cancer (PCa). MALDI-TOF/MS peptidomic profiling could be a useful diagnostic tool for biomarker discovery, although reproducibility issues have limited its applicability until now. The current study aimed to evaluate a new MALDI-TOF/MS candidate biomarker. Methods Within- and between-subject variability of MALDI-TOF/MS-based peptidomic urine and serum analyses were evaluated in 20 and 15 healthy donors, respectively. Normalizations and approaches for accounting below limit of detection (LOD) values were utilized to enhance reproducibility, while Monte Carlo experiments were performed to verify whether measurement error can be dealt with LOD data. Post-prostatic massage urine and serum samples from 148 LUTS patients were analysed using MALDI-TOF/MS. Regression-calibration and simulation and extrapolation methods were used to derive the unbiased association between peptidomic features and PCa. Results Although the median normalized peptidomic variability was 24.9%, the within- and between-subject variability showed that median normalization, LOD adjustment, and log2 data transformation were the best combination in terms of reliability; in measurement error conditions, intraclass correlation coefficient was a reliable estimate when the LOD/2 was substituted for below LOD values. In the patients studied, 43 peptides were shared by the urine and serum, and several features were found to be associated with PCa. Only few serum features, however, show statistical significance after the multiple testing procedures were completed. Two serum fragmentation patterns corresponded to the complement C4-A. Conclusions MALDI-TOF/MS serum peptidome profiling was more efficacious with respect to post-prostatic massage urine analysis in discriminating PCa. Electronic supplementary material The online version of this article (10.1186/s12014-018-9199-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Andrea Padoan
- 1Department of Medicine-DIMED, University of Padova, Via Giustiniani 2, 35128 Padua, Italy
| | - Daniela Basso
- 1Department of Medicine-DIMED, University of Padova, Via Giustiniani 2, 35128 Padua, Italy
| | | | - Tommaso Prayer-Galetti
- 3Department of Surgical, Oncological and Gastroenterological Sciences, University of Padova, Padua, Italy
| | - Giorgio Arrigoni
- 2Department of Biomedical Sciences, University of Padova, Padua, Italy.,4Proteomic Center, University of Padova, Padua, Italy
| | - Dania Bozzato
- 1Department of Medicine-DIMED, University of Padova, Via Giustiniani 2, 35128 Padua, Italy
| | - Stefania Moz
- 1Department of Medicine-DIMED, University of Padova, Via Giustiniani 2, 35128 Padua, Italy
| | - Filiberto Zattoni
- 3Department of Surgical, Oncological and Gastroenterological Sciences, University of Padova, Padua, Italy
| | - Rino Bellocco
- 5Department of Statistics and Quantitative Methods, University of Milano-Bicocca, Milan, Italy.,6Department of Medical Epidemiology and Biostatistics (MEB), Karolinska Institute, Stockholm, Sweden
| | - Mario Plebani
- 1Department of Medicine-DIMED, University of Padova, Via Giustiniani 2, 35128 Padua, Italy
| |
Collapse
|
40
|
Huang HC, Qin LX. Empirical evaluation of data normalization methods for molecular classification. PeerJ 2018; 6:e4584. [PMID: 29666754 PMCID: PMC5899419 DOI: 10.7717/peerj.4584] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 03/18/2018] [Indexed: 11/20/2022] Open
Abstract
Background Data artifacts due to variations in experimental handling are ubiquitous in microarray studies, and they can lead to biased and irreproducible findings. A popular approach to correct for such artifacts is through post hoc data adjustment such as data normalization. Statistical methods for data normalization have been developed and evaluated primarily for the discovery of individual molecular biomarkers. Their performance has rarely been studied for the development of multi-marker molecular classifiers-an increasingly important application of microarrays in the era of personalized medicine. Methods In this study, we set out to evaluate the performance of three commonly used methods for data normalization in the context of molecular classification, using extensive simulations based on re-sampling from a unique pair of microRNA microarray datasets for the same set of samples. The data and code for our simulations are freely available as R packages at GitHub. Results In the presence of confounding handling effects, all three normalization methods tended to improve the accuracy of the classifier when evaluated in an independent test data. The level of improvement and the relative performance among the normalization methods depended on the relative level of molecular signal, the distributional pattern of handling effects (e.g., location shift vs scale change), and the statistical method used for building the classifier. In addition, cross-validation was associated with biased estimation of classification accuracy in the over-optimistic direction for all three normalization methods. Conclusion Normalization may improve the accuracy of molecular classification for data with confounding handling effects; however, it cannot circumvent the over-optimistic findings associated with cross-validation for assessing classification accuracy.
Collapse
Affiliation(s)
- Huei-Chung Huang
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Li-Xuan Qin
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
41
|
Qin LX, Huang HC, Begg CB. Cautionary Note on Using Cross-Validation for Molecular Classification. J Clin Oncol 2017; 34:3931-3938. [PMID: 27601553 DOI: 10.1200/jco.2016.68.1031] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose Reproducibility of scientific experimentation has become a major concern because of the perception that many published biomedical studies cannot be replicated. In this article, we draw attention to the connection between inflated overoptimistic findings and the use of cross-validation for error estimation in molecular classification studies. We show that, in the absence of careful design to prevent artifacts caused by systematic differences in the processing of specimens, established tools such as cross-validation can lead to a spurious estimate of the error rate in the overoptimistic direction, regardless of the use of data normalization as an effort to remove these artifacts. Methods We demonstrated this important yet overlooked complication of cross-validation using a unique pair of data sets on the same set of tumor samples. One data set was collected with uniform handling to prevent handling effects; the other was collected without uniform handling and exhibited handling effects. The paired data sets were used to estimate the biologic effects of the samples and the handling effects of the arrays in the latter data set, which were then used to simulate data using virtual rehybridization following various array-to-sample assignment schemes. Results Our study showed that (1) cross-validation tended to underestimate the error rate when the data possessed confounding handling effects; (2) depending on the relative amount of handling effects, normalization may further worsen the underestimation of the error rate; and (3) balanced assignment of arrays to comparison groups allowed cross-validation to provide an unbiased error estimate. Conclusion Our study demonstrates the benefits of balanced array assignment for reproducible molecular classification and calls for caution on the routine use of data normalization and cross-validation in such analysis.
Collapse
Affiliation(s)
- Li-Xuan Qin
- All authors: Memorial Sloan Kettering Cancer Center, New York, NY
| | - Huei-Chung Huang
- All authors: Memorial Sloan Kettering Cancer Center, New York, NY
| | - Colin B Begg
- All authors: Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
42
|
Lu M, Faull KF, Whitelegge JP, He J, Shen D, Saxton RE, Chang HR. Proteomics and Mass Spectrometry for Cancer Biomarker Discovery. Biomark Insights 2017. [DOI: 10.1177/117727190700200005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Proteomics is a rapidly advancing field not only in the field of biology but also in translational cancer research. In recent years, mass spectrometry and associated technologies have been explored to identify proteins or a set of proteins specific to a given disease, for the purpose of disease detection and diagnosis. Such biomarkers are being investigated in samples including cells, tissues, serum/plasma, and other types of body fluids. When sufficiently refined, proteomic technologies may pave the way for early detection of cancer or individualized therapy for cancer. Mass spectrometry approaches coupled with bioinformatic tools are being developed for biomarker discovery and validation. Understanding basic concepts and application of such technology by investigators in the field may accelerate the clinical application of protein biomarkers in disease management.
Collapse
Affiliation(s)
- Ming Lu
- Gonda/UCLA Breast Cancer Research Laboratory, Los Angeles, California
- Revlon/UCLA Breast Center, Department of Surgery/Oncology, David Geffen School of Medicine, Los Angeles, California
| | - Kym F. Faull
- The Pasarow Mass Spectrometry Laboratory, Department of Psychiatry & Biobehavioral and the Neuropsychiatric Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles
| | - Julian P. Whitelegge
- The Pasarow Mass Spectrometry Laboratory, Department of Psychiatry & Biobehavioral and the Neuropsychiatric Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles
| | - Jianbo He
- Gonda/UCLA Breast Cancer Research Laboratory, Los Angeles, California
- Revlon/UCLA Breast Center, Department of Surgery/Oncology, David Geffen School of Medicine, Los Angeles, California
| | - Dejun Shen
- Gonda/UCLA Breast Cancer Research Laboratory, Los Angeles, California
- Revlon/UCLA Breast Center, Department of Surgery/Oncology, David Geffen School of Medicine, Los Angeles, California
| | - Romaine E. Saxton
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine, Los Angeles, California
| | - Helena R. Chang
- Gonda/UCLA Breast Cancer Research Laboratory, Los Angeles, California
- Revlon/UCLA Breast Center, Department of Surgery/Oncology, David Geffen School of Medicine, Los Angeles, California
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine, Los Angeles, California
| |
Collapse
|
43
|
Moore RE, Kirwan J, Doherty MK, Whitfield PD. Biomarker Discovery in Animal Health and Disease: The Application of Post-Genomic Technologies. Biomark Insights 2017. [DOI: 10.1177/117727190700200040] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The causes of many important diseases in animals are complex and multifactorial, which present unique challenges. Biomarkers indicate the presence or extent of a biological process, which is directly linked to the clinical manifestations and outcome of a particular disease. Identifying biomarkers or biomarker profiles will be an important step towards disease characterization and management of disease in animals. The emergence of post-genomic technologies has led to the development of strategies aimed at identifying specific and sensitive biomarkers from the thousands of molecules present in a tissue or biological fluid. This review will summarize the current developments in biomarker discovery and will focus on the role of transcriptomics, proteomics and metabolomics in biomarker discovery for animal health and disease.
Collapse
Affiliation(s)
- Rowan E. Moore
- Proteomics and Functional Genomics Research Group, Faculty of Veterinary Science, University of Liverpool, Liverpool, United Kingdom
| | - Jennifer Kirwan
- Proteomics and Functional Genomics Research Group, Faculty of Veterinary Science, University of Liverpool, Liverpool, United Kingdom
| | - Mary K. Doherty
- Proteomics and Functional Genomics Research Group, Faculty of Veterinary Science, University of Liverpool, Liverpool, United Kingdom
| | - Phillip D. Whitfield
- Proteomics and Functional Genomics Research Group, Faculty of Veterinary Science, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
44
|
Lyons-Weiler J. Standards of Excellence and Open Questions in Cancer Biomarker Research: An Informatics Perspective. Cancer Inform 2017. [DOI: 10.1177/117693510500100105] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
45
|
Abstract
We consider large-scale studies in which thousands of significance tests are performed simultaneously. In some of these studies, the multiple testing procedure can be severely biased by latent confounding factors such as batch effects and unmeasured covariates that correlate with both primary variable(s) of interest (e.g., treatment variable, phenotype) and the outcome. Over the past decade, many statistical methods have been proposed to adjust for the confounders in hypothesis testing. We unify these methods in the same framework, generalize them to include multiple primary variables and multiple nuisance variables, and analyze their statistical properties. In particular, we provide theoretical guarantees for RUV-4 [Gagnon-Bartsch, Jacob and Speed (2013)] and LEAPP [Ann. Appl. Stat. 6 (2012) 1664-1688], which correspond to two different identification conditions in the framework: the first requires a set of "negative controls" that are known a priori to follow the null distribution; the second requires the true nonnulls to be sparse. Two different estimators which are based on RUV-4 and LEAPP are then applied to these two scenarios. We show that if the confounding factors are strong, the resulting estimators can be asymptotically as powerful as the oracle estimator which observes the latent confounding factors. For hypothesis testing, we show the asymptotic z-tests based on the estimators can control the type I error. Numerical experiments show that the false discovery rate is also controlled by the Benjamini-Hochberg procedure when the sample size is reasonably large.
Collapse
Affiliation(s)
- Jingshu Wang
- Department of Statistics, The Wharton School, University of Pennsylvania, 400 Huntsman Hall, 3730 Walnut St, Philadelphia, Pennsylvania 19104, USA
| | - Qingyuan Zhao
- Department of Statistics, The Wharton School, University of Pennsylvania, 400 Huntsman Hall, 3730 Walnut St, Philadelphia, Pennsylvania 19104, USA
| | - Trevor Hastie
- Department of Statistics, Stanford University, 390 Serra Mall, Stanford, California 94305, USA
| | - Art B. Owen
- Department of Statistics, Stanford University, 390 Serra Mall, Stanford, California 94305, USA
| |
Collapse
|
46
|
Mahabir S, Gerlai R. The Importance of Holding Water: Salinity and Chemosensory Cues Affect Zebrafish Behavior. Zebrafish 2017; 14:444-458. [DOI: 10.1089/zeb.2017.1472] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Affiliation(s)
- Samantha Mahabir
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | - Robert Gerlai
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
- Department of Psychology, University of Toronto Mississauga, Mississauga, Canada
| |
Collapse
|
47
|
Posti JP, Dickens AM, Orešič M, Hyötyläinen T, Tenovuo O. Metabolomics Profiling As a Diagnostic Tool in Severe Traumatic Brain Injury. Front Neurol 2017; 8:398. [PMID: 28868043 PMCID: PMC5563327 DOI: 10.3389/fneur.2017.00398] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 07/25/2017] [Indexed: 12/16/2022] Open
Abstract
Traumatic brain injury (TBI) is a complex disease with a multifaceted pathophysiology. Impairment of energy metabolism is a key component of secondary insults. This phenomenon is a consequence of multiple potential mechanisms including diffusion hypoxia, mitochondrial failure, and increased energy needs due to systemic trauma responses, seizures, or spreading depolarization. The degree of disturbance in brain metabolism is affected by treatment interventions and reflected in clinical patient outcome. Hence, monitoring of these secondary events in peripheral blood will provide a window into the pathophysiological course of severe TBI. New methods for assessing perturbation of brain metabolism are needed in order to monitor on-going pathophysiological processes and thus facilitate targeted interventions and predict outcome. Circulating metabolites in peripheral blood may serve as sensitive markers of pathological processes in TBI. The levels of these small molecules in blood are less dependent on the integrity of the blood–brain barrier as compared to protein biomarkers. We have recently characterized a specific metabolic profile in serum that is associated with both initial severity and patient outcome of TBI. We found that two medium-chain fatty acids, octanoic and decanoic acids, as well as several sugar derivatives are significantly associated with the severity of TBI. The top ranking peripheral blood metabolites were also highly correlated with their levels in cerebral microdialyzates. Based on the metabolite profile upon admission, we have been able to develop a model that accurately predicts patient outcome. Moreover, metabolomics profiling improved the performance of the well-established clinical prognostication model. In this review, we discuss metabolomics profiling in patients with severe TBI. We present arguments in support of the need for further development and validation of circulating biomarkers of cerebral metabolism and for their use in assessing patients with severe TBI.
Collapse
Affiliation(s)
- Jussi P Posti
- Division of Clinical Neurosciences, Department of Neurosurgery, Turku University Hospital, Turku, Finland.,Division of Clinical Neurosciences, Department of Rehabilitation and Brain Trauma, Turku University Hospital, Turku, Finland.,Department of Neurology, University of Turku, Turku, Finland
| | - Alex M Dickens
- Turku Centre for Biotechnology, University of Turku, Turku, Finland
| | - Matej Orešič
- Turku Centre for Biotechnology, University of Turku, Turku, Finland
| | | | - Olli Tenovuo
- Division of Clinical Neurosciences, Department of Rehabilitation and Brain Trauma, Turku University Hospital, Turku, Finland.,Department of Neurology, University of Turku, Turku, Finland
| |
Collapse
|
48
|
Hewitt JA, Brown LL, Murphy SJ, Grieder F, Silberberg SD. Accelerating Biomedical Discoveries through Rigor and Transparency. ILAR J 2017; 58:115-128. [PMID: 28575443 PMCID: PMC6279133 DOI: 10.1093/ilar/ilx011] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 03/07/2017] [Accepted: 03/16/2017] [Indexed: 12/13/2022] Open
Abstract
Difficulties in reproducing published research findings have garnered a lot of press in recent years. As a funder of biomedical research, the National Institutes of Health (NIH) has taken measures to address underlying causes of low reproducibility. Extensive deliberations resulted in a policy, released in 2015, to enhance reproducibility through rigor and transparency. We briefly explain what led to the policy, describe its elements, provide examples and resources for the biomedical research community, and discuss the potential impact of the policy on translatability with a focus on research using animal models. Importantly, while increased attention to rigor and transparency may lead to an increase in the number of laboratory animals used in the near term, it will lead to more efficient and productive use of such resources in the long run. The translational value of animal studies will be improved through more rigorous assessment of experimental variables and data, leading to better assessments of the translational potential of animal models, for the benefit of the research community and society.
Collapse
Affiliation(s)
- Judith A. Hewitt
- Judith A. Hewitt, PhD, is the Chief of the Research Resources Section in the Office of Biodefense, Research Resources and Translational Research, in the Division of Microbiology and Infectious Diseases at the National Institute of Allergy and Infectious Diseases in Bethesda, MD. Liliana L. Brown, PhD, is a Program Officer in the Office of Genomics and Advanced Technologies, in the Division of Microbiology and Infectious Diseases at the National Institute of Allergy and Infectious Diseases in Bethesda, MD. Stephanie J. Murphy, VMD, PhD, is the Director of the Division of Comparative Medicine within the Office of Infrastructure Programs in the Division of Program Coordination, Planning, and Strategic Initiatives in the Office of the Director at the National Institutes of Health in Bethesda, MD. Franziska Grieder, DVM, PhD, is the Director of the Office of Infrastructure Programs in the Division of Program Coordination, Planning, and Strategic Initiatives in the Office of the Director at the National Institutes of Health in Bethesda, MD. Shai D. Silberberg, PhD, is the Director of Research Quality at the National Institute of Neurological Disorders and Stroke in Bethesda MD.
| | - Liliana L. Brown
- Judith A. Hewitt, PhD, is the Chief of the Research Resources Section in the Office of Biodefense, Research Resources and Translational Research, in the Division of Microbiology and Infectious Diseases at the National Institute of Allergy and Infectious Diseases in Bethesda, MD. Liliana L. Brown, PhD, is a Program Officer in the Office of Genomics and Advanced Technologies, in the Division of Microbiology and Infectious Diseases at the National Institute of Allergy and Infectious Diseases in Bethesda, MD. Stephanie J. Murphy, VMD, PhD, is the Director of the Division of Comparative Medicine within the Office of Infrastructure Programs in the Division of Program Coordination, Planning, and Strategic Initiatives in the Office of the Director at the National Institutes of Health in Bethesda, MD. Franziska Grieder, DVM, PhD, is the Director of the Office of Infrastructure Programs in the Division of Program Coordination, Planning, and Strategic Initiatives in the Office of the Director at the National Institutes of Health in Bethesda, MD. Shai D. Silberberg, PhD, is the Director of Research Quality at the National Institute of Neurological Disorders and Stroke in Bethesda MD.
| | - Stephanie J. Murphy
- Judith A. Hewitt, PhD, is the Chief of the Research Resources Section in the Office of Biodefense, Research Resources and Translational Research, in the Division of Microbiology and Infectious Diseases at the National Institute of Allergy and Infectious Diseases in Bethesda, MD. Liliana L. Brown, PhD, is a Program Officer in the Office of Genomics and Advanced Technologies, in the Division of Microbiology and Infectious Diseases at the National Institute of Allergy and Infectious Diseases in Bethesda, MD. Stephanie J. Murphy, VMD, PhD, is the Director of the Division of Comparative Medicine within the Office of Infrastructure Programs in the Division of Program Coordination, Planning, and Strategic Initiatives in the Office of the Director at the National Institutes of Health in Bethesda, MD. Franziska Grieder, DVM, PhD, is the Director of the Office of Infrastructure Programs in the Division of Program Coordination, Planning, and Strategic Initiatives in the Office of the Director at the National Institutes of Health in Bethesda, MD. Shai D. Silberberg, PhD, is the Director of Research Quality at the National Institute of Neurological Disorders and Stroke in Bethesda MD.
| | - Franziska Grieder
- Judith A. Hewitt, PhD, is the Chief of the Research Resources Section in the Office of Biodefense, Research Resources and Translational Research, in the Division of Microbiology and Infectious Diseases at the National Institute of Allergy and Infectious Diseases in Bethesda, MD. Liliana L. Brown, PhD, is a Program Officer in the Office of Genomics and Advanced Technologies, in the Division of Microbiology and Infectious Diseases at the National Institute of Allergy and Infectious Diseases in Bethesda, MD. Stephanie J. Murphy, VMD, PhD, is the Director of the Division of Comparative Medicine within the Office of Infrastructure Programs in the Division of Program Coordination, Planning, and Strategic Initiatives in the Office of the Director at the National Institutes of Health in Bethesda, MD. Franziska Grieder, DVM, PhD, is the Director of the Office of Infrastructure Programs in the Division of Program Coordination, Planning, and Strategic Initiatives in the Office of the Director at the National Institutes of Health in Bethesda, MD. Shai D. Silberberg, PhD, is the Director of Research Quality at the National Institute of Neurological Disorders and Stroke in Bethesda MD.
| | - Shai D. Silberberg
- Judith A. Hewitt, PhD, is the Chief of the Research Resources Section in the Office of Biodefense, Research Resources and Translational Research, in the Division of Microbiology and Infectious Diseases at the National Institute of Allergy and Infectious Diseases in Bethesda, MD. Liliana L. Brown, PhD, is a Program Officer in the Office of Genomics and Advanced Technologies, in the Division of Microbiology and Infectious Diseases at the National Institute of Allergy and Infectious Diseases in Bethesda, MD. Stephanie J. Murphy, VMD, PhD, is the Director of the Division of Comparative Medicine within the Office of Infrastructure Programs in the Division of Program Coordination, Planning, and Strategic Initiatives in the Office of the Director at the National Institutes of Health in Bethesda, MD. Franziska Grieder, DVM, PhD, is the Director of the Office of Infrastructure Programs in the Division of Program Coordination, Planning, and Strategic Initiatives in the Office of the Director at the National Institutes of Health in Bethesda, MD. Shai D. Silberberg, PhD, is the Director of Research Quality at the National Institute of Neurological Disorders and Stroke in Bethesda MD.
| |
Collapse
|
49
|
Coyle KM, Boudreau JE, Marcato P. Genetic Mutations and Epigenetic Modifications: Driving Cancer and Informing Precision Medicine. BIOMED RESEARCH INTERNATIONAL 2017; 2017:9620870. [PMID: 28685150 PMCID: PMC5480027 DOI: 10.1155/2017/9620870] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 04/06/2017] [Accepted: 05/10/2017] [Indexed: 12/21/2022]
Abstract
Cancer treatment is undergoing a significant revolution from "one-size-fits-all" cytotoxic therapies to tailored approaches that precisely target molecular alterations. Precision strategies for drug development and patient stratification, based on the molecular features of tumors, are the next logical step in a long history of approaches to cancer therapy. In this review, we discuss the history of cancer treatment from generic natural extracts and radical surgical procedures to site-specific and combinatorial treatment regimens, which have incrementally improved patient outcomes. We discuss the related contributions of genetics and epigenetics to cancer progression and the response to targeted therapies and identify challenges and opportunities for the success of precision medicine. The identification of patients who will benefit from targeted therapies is more complex than simply identifying patients whose tumors harbour the targeted aberration, and intratumoral heterogeneity makes it difficult to determine if a precision therapy is successful during treatment. This heterogeneity enables tumors to develop resistance to targeted approaches; therefore, the rational combination of therapeutic agents will limit the threat of acquired resistance to therapeutic success. By incorporating the view of malignant transformation modulated by networks of genetic and epigenetic interactions, molecular strategies will enable precision medicine for effective treatment across cancer subtypes.
Collapse
Affiliation(s)
| | - Jeanette E. Boudreau
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada
| | - Paola Marcato
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
50
|
Ioannidis JPA, Bossuyt PMM. Waste, Leaks, and Failures in the Biomarker Pipeline. Clin Chem 2017; 63:963-972. [DOI: 10.1373/clinchem.2016.254649] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 11/30/2016] [Indexed: 01/05/2023]
Abstract
Abstract
BACKGROUND
The large, expanding literature on biomarkers is characterized by almost ubiquitous significant results, with claims about the potential importance, but few of these discovered biomarkers are used in routine clinical care.
CONTENT
The pipeline of biomarker development includes several specific stages: discovery, validation, clinical translation, evaluation, implementation (and, in the case of nonutility, deimplementation). Each of these stages can be plagued by problems that cause failures of the overall pipeline. Some problems are nonspecific challenges for all biomedical investigation, while others are specific to the peculiarities of biomarker research. Discovery suffers from poor methods and incomplete and selective reporting. External independent validation is limited. Selection for clinical translation is often shaped by nonrational choices. Evaluation is sparse and the clinical utility of many biomarkers remains unknown. The regulatory environment for biomarkers remains weak and guidelines can reach biased or divergent recommendations. Removing inefficient or even harmful biomarkers that have been entrenched in clinical care can meet with major resistance.
SUMMARY
The current biomarker pipeline is too prone to failures. Consideration of clinical needs should become a starting point for the development of biomarkers. Improvements can include the use of more stringent methodology, better reporting, larger collaborative studies, careful external independent validation, preregistration, rigorous systematic reviews and umbrella reviews, pivotal randomized trials, and implementation and deimplementation studies. Incentives should be aligned toward delivering useful biomarkers.
Collapse
Affiliation(s)
- John P A Ioannidis
- Departments of Medicine, Health Research and Policy, and Statistics, and the Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, CA
| | - Patrick M M Bossuyt
- Department of Clinical Epidemiology, Biostatistics & Bioinformatics, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|