A roadmap towards personalized immunology

Sylvie Delhalle, Sebastian F N Bode, Rudi Balling, Markus Ollert, Feng Q He, Sylvie Delhalle, Sebastian F N Bode, Rudi Balling, Markus Ollert, Feng Q He

Abstract

Big data generation and computational processing will enable medicine to evolve from a "one-size-fits-all" approach to precise patient stratification and treatment. Significant achievements using "Omics" data have been made especially in personalized oncology. However, immune cells relative to tumor cells show a much higher degree of complexity in heterogeneity, dynamics, memory-capability, plasticity and "social" interactions. There is still a long way ahead on translating our capability to identify potentially targetable personalized biomarkers into effective personalized therapy in immune-centralized diseases. Here, we discuss the recent advances and successful applications in "Omics" data utilization and network analysis on patients' samples of clinical trials and studies, as well as the major challenges and strategies towards personalized stratification and treatment for infectious or non-communicable inflammatory diseases such as autoimmune diseases or allergies. We provide a roadmap and highlight experimental, clinical, computational analysis, data management, ethical and regulatory issues to accelerate the implementation of personalized immunology.

Conflict of interest statement

The authors declare no competing financial interests.

Figures

Fig. 1
Fig. 1
A roadmap proposed towards personalized immunology. There exist both horizontal and vertical roadmaps towards personalized immunology. Vertically, to translate sample stratification to clinical therapies, we need to utilize the state-of-the-art “Omics” analysis and network integration approaches to stratify patients into subgroups and then implement personalized therapeutic approaches to treat individual patients, which needs to overcome various types of barriers at different steps. Horizontally, we might need to go through at least 7 steps to enable personalized immunotherapies, 1) classic symptom-based approach, 2) deep phenotyping approach, 3) multi-layer “Omics”-based profiling, 4) cell type-specific “Omics”, 5) state-specific “Omics”, 6) single-cell (sc) “Omics” and dynamic response analysis of immune cells, 7) integrated network analysis. FACS, fluorescence activated cell sorting; TCR/BCR, T cell receptor/B-cell receptor; DEG, differential expression gene; PEEP, personalized expression perturbation profile; SSN, sample-specific network; SVM, support vector machine; KNN, K-nearest neighbors; under the first layer (the so-called stratification layer), different colors of patients indicate individual patients with different cellular and/or molecular profiles while brackets represent patient subgroups; under the second layer (the so-called technique layers), different small circles with distinct colors indicate different immune cells while big circles represent patient (sub)groups; under the technique layers, the snapshot of microarray representing either microarray-based or RNA-seq-based transcriptome analysis; under the third layer (the so-called therapeutic layer), the syringes with different colors or tonalities indicate different therapeutic approaches; P1,..., Pn at step 7 designate different patients; G1, G2, G3, G4 represent different genes, the arrows between them representing regulatory relationships. Three images in the second layer of step 1 are used with permissions from Fotolia.com
Fig. 2
Fig. 2
Longitudinal studies and dynamic measurement are critical for discovering various types of biomarkers. a Longitudinal follow-up of individual patients with multilayer “Omics” analysis is essential for identifying different types of biomarkers. The check marker at the given time point indicates the necessary “Omics” measurement and clinical assessment for revealing the given type of biomarker while the cross symbol indicates an unnecessary involvement at the given time point for the given type of biomarker. b Time-series “Omics” analysis of the cultured isolated immune cells from the first visit (T0 at panel a) following certain stimulation or stresses will also be able to help extract various types of biomarkers. c Various types of dynamic patterns of different pathways or modules or subnetworks of the given relevant type of immune cells isolated from PBMC or other tissues of individual patients might be valuable for patient subgroup stratification. Subnetwork activities at the given time can be defined either by the expression levels of the co-expressed genes, or by the expression levels of the effector genes (such as cytokines) or any other readouts which could define the activities or outputs of the given pathway or subnetwork or module

References

    1. Council, N. R. Toward Precision Medicine: Building a Knowledge Network for Biomedical Research and a New Taxonomy of Disease (The National Academies Press, Washington, DC, 2011).
    1. Ye CJ, et al. Intersection of population variation and autoimmunity genetics in human T cell activation. Science. 2014;345:1254665.
    1. Duffy D, et al. Functional analysis via standardized whole-blood stimulation systems defines the boundaries of a healthy immune response to complex stimuli. Immunity. 2014;40:436–450.
    1. Willis JC, Lord GM. Immune biomarkers: the promises and pitfalls of personalized medicine. Nat. Rev. Immunol. 2015;15:323–329.
    1. Schork NJ. Personalized medicine: time for one-person trials. Nature. 2015;520:609–611.
    1. Cancer Genome Atlas Research Network et al. The cancer genome atlas pan-cancer analysis project. Nat. Genet. 2013;45:1113–1120.
    1. Zhang J, et al. International cancer genome consortium data portal—a one-stop shop for cancer genomics data. Database. 2011;2011:bar026.
    1. Yu KH, Snyder M. Omics profiling in precision oncology. Mol. Cell. Proteom. 2016;15:2525–2536.
    1. Schilsky RL. Implementing personalized cancer care. Nat. Rev. Clin. Oncol. 2014;11:432–438.
    1. Collins FS, Varmus H. A new initiative on precision medicine. N. Engl. J. Med. 2015;372:793–795.
    1. Hood L, Friend SH. Predictive, personalized, preventive, participatory (P4) cancer medicine. Nat. Rev. Clin. Oncol. 2011;8:184–187.
    1. Sahin U, et al. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature. 2017;547:222–226.
    1. Ott PA, et al. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature. 2017 doi: 10.1038/nature22991.
    1. Rieckmann JC, et al. Social network architecture of human immune cells unveiled by quantitative proteomics. Nat. Immunol. 2017;18:583–593.
    1. Davis MM, Tato CM, Furman D. Systems immunology: just getting started. Nat. Immunol. 2017;18:725–732.
    1. Margolis R, et al. The National Institutes of Health’s Big Data to Knowledge (BD2K) initiative: capitalizing on biomedical big data. J. Am. Med. Inform. Assoc. 2014;21:957–958.
    1. McDade TW, et al. Social and physical environments early in development predict DNA methylation of inflammatory genes in young adulthood. Proc. Natl. Acad. Sci. USA. 2017;114:7611–7616.
    1. Kent A, et al. Lymphocyte subpopulations in premature infants: an observational study. Arch. Dis. Child. Fetal Neonatal Ed. 2016 doi: 10.1136/archdischild-2015-309246.
    1. Beral V, Million Women Study Collaborators Breast cancer and hormone-replacement therapy in the Million Women Study. Lancet. 2003;362:419–427.
    1. Shannon CE. Communication in the presence of noise. Proc. IRE. 1949;37:10–21.
    1. Banchereau R, et al. Personalized immunomonitoring uncovers molecular networks that stratify lupus patients. Cell. 2016;165:551–565.
    1. Langfelder P, Horvath S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinforma. 2008;9:559.
    1. Budde P, et al. Multiparametric detection of autoantibodies in systemic lupus erythematosus. Lupus. 2016;25:812–822.
    1. World Allergy Organization. World Allergy Organization (WAO) White Book on Allergy: Update 2013 (World Allergy Organization, Milwaukee, 2013).
    1. Akdis CA, Akdis M. Mechanisms of allergen-specific immunotherapy and immune tolerance to allergens. World Allergy Organ. J. 2015;8:17.
    1. Durham SR, Penagos M. Sublingual or subcutaneous immunotherapy for allergic rhinitis? J. Allergy Clin. Immunol. 2016;137:339–349.
    1. Ryan JF, et al. Successful immunotherapy induces previously unidentified allergen-specific CD4+ T-cell subsets. Proc. Natl. Acad. Sci. USA. 2016 doi: 10.1073/pnas.1520180113.
    1. Syed A, et al. Peanut oral immunotherapy results in increased antigen-induced regulatory T-cell function and hypomethylation of forkhead box protein 3 (FOXP3) J. Allergy Clin. Immunol. 2014;133:500–510.
    1. Blank S, et al. Api m 10, a genuine A. mellifera venom allergen, is clinically relevant but underrepresented in therapeutic extracts. Allergy. 2011;66:1322–1329.
    1. Eberlein B, Krischan L, Darsow U, Ollert M, Ring J. Double positivity to bee and wasp venom: improved diagnostic procedure by recombinant allergen-based IgE testing and basophil activation test including data about cross-reactive carbohydrate determinants. J. Allergy Clin. Immunol. 2012;130:155–161.
    1. Korosec P, et al. High sensitivity of CAP-FEIA rVes v 5 and rVes v 1 for diagnosis of Vespula venom allergy. J. Allergy Clin. Immunol. 2012;129:1406–1408.
    1. Frick M, et al. Predominant Api m 10 sensitization as risk factor for treatment failure in honey bee venom immunotherapy. J. Allergy Clin. Immunol. 2016;138:1663–1671.
    1. Stapel SO, et al. Testing for IgG4 against foods is not recommended as a diagnostic tool: EAACI Task Force Report. Allergy. 2008;63:793–796.
    1. Marth K, Focke-Tejkl M, Lupinek C, Valenta R, Niederberger V. Allergen peptides, recombinant allergens and hypoallergens for allergen-specific immunotherapy. Curr. Treat. Options Allergy. 2014;1:91–106.
    1. Bonifazi F, et al. Prevention and treatment of hymenoptera venom allergy: guidelines for clinical practice. Allergy. 2005;60:1459–1470.
    1. Crawford A, et al. Molecular and transcriptional basis of CD4(+) T cell dysfunction during chronic infection. Immunity. 2014;40:289–302.
    1. Holcomb ZE, Tsalik EL, Woods CW, McClain MT. Host-based peripheral blood gene expression analysis for diagnosis of infectious diseases. J. Clin. Microbiol. 2017;55:360–368.
    1. Nakaya HI, et al. Systems biology of immunity to MF59-adjuvanted versus nonadjuvanted trivalent seasonal influenza vaccines in early childhood. Proc. Natl. Acad. Sci. USA. 2016 doi: 10.1073/pnas.1519690113.
    1. Pulendran B, Li S, Nakaya HI. Systems vaccinology. Immunity. 2010;33:516–529.
    1. Wang IM, Bett AJ, Cristescu R, Loboda A, ter Meulen J. Transcriptional profiling of vaccine-induced immune responses in humans and non-human primates. Microb. Biotechnol. 2012;5:177–187.
    1. Fourati, S. et al. Pre-vaccination inflammation and B-cell signalling predict age-related hyporesponse to hepatitis B vaccination. Nat. Commun.10.1038/ncomms10369 (2016).
    1. Bock C, Farlik M, Sheffield NC. Multi-omics of single cells: strategies and applications. Trends Biotechnol. 2016;34:605–608.
    1. Shen-Orr SS, Gaujoux R. Computational deconvolution: extracting cell type-specific information from heterogeneous samples. Curr. Opin. Immunol. 2013;25:571–578.
    1. Huang AC, et al. T-cell invigoration to tumour burden ratio associated with anti-PD-1 response. Nature. 2017;545:60–65.
    1. McKinney EF, Lee JC, Jayne DR, Lyons PA, Smith KG. T-cell exhaustion, co-stimulation and clinical outcome in autoimmunity and infection. Nature. 2015;523:612–616.
    1. Okhrimenko A, et al. Human memory T cells from the bone marrow are resting and maintain long-lasting systemic memory. Proc. Natl. Acad. Sci. USA. 2014;111:9229–9234.
    1. Saadatpour A, Lai S, Guo G, Yuan GC. Single-cell analysis in cancer genomics. Trends Genet. 2015;31:576–586.
    1. Tang F, Lao K, Surani MA. Development and applications of single-cell transcriptome analysis. Nat. Methods. 2011;8:S6–S11.
    1. Jaitin DA, Keren-Shaul H, Elefant N, Amit I. Each cell counts: hematopoiesis and immunity research in the era of single cell genomics. Semin. Immunol. 2015;27:67–71.
    1. Stegle O, Teichmann SA, Marioni JC. Computational and analytical challenges in single-cell transcriptomics. Nat. Rev. Genet. 2015;16:133–145.
    1. Heath JR, Ribas A, Mischel PS. Single-cell analysis tools for drug discovery and development. Nat. Rev. Drug. Discov. 2016;15:204–216.
    1. Stoeckius M, et al. Simultaneous epitope and transcriptome measurement in single cells. Nat. Methods. 2017 doi: 10.1038/nmeth.4380.
    1. Avraham R, et al. Pathogen cell-to-cell variability drives heterogeneity in host immune responses. Cell. 2015;162:1309–1321.
    1. Villani, A. C. et al. Single-cell RNA-seq reveals new types of human blood dendritic cells, monocytes, and progenitors. Science (2017).
    1. Schwartzman O, Tanay A. Single-cell epigenomics: techniques and emerging applications. Nat. Rev. Genet. 2015;16:716–726.
    1. Macaulay IC, Ponting CP, Voet T. Single-cell multiomics: multiple measurements from single cells. Trends Genet. 2017;33:155–168.
    1. Lorthongpanich C, et al. Single-cell DNA-methylation analysis reveals epigenetic chimerism in preimplantation embryos. Science. 2013;341:1110–1112.
    1. Cheow LF, et al. Single-cell multimodal profiling reveals cellular epigenetic heterogeneity. Nat. Methods. 2016;13:833–836.
    1. Gravina S, Dong X, Yu B, Vijg J. Single-cell genome-wide bisulfite sequencing uncovers extensive heterogeneity in the mouse liver methylome. Genome Biol. 2016;17:150.
    1. Zenobi R. Single-cell metabolomics: analytical and biological perspectives. Science. 2013;342:1243259.
    1. Zheng C, et al. Landscape of Infiltrating T Cells in Liver Cancer Revealed by Single-Cell Sequencing. Cell. 2017;169:1342–1356.
    1. Wang R, et al. Flickering gives early warning signals of a critical transition to a eutrophic lake state. Nature. 2012;492:419–422.
    1. Trefois C, Antony PM, Goncalves J, Skupin A, Balling R. Critical transitions in chronic disease: transferring concepts from ecology to systems medicine. Curr. Opin. Biotechnol. 2015;34:48–55.
    1. McDade E, Bateman RJ. Stop Alzheimer’s before it starts. Nature. 2017;547:153–155.
    1. Fey D, et al. Signaling pathway models as biomarkers: patient-specific simulations of JNK activity predict the survival of neuroblastoma patients. Sci. Signal. 2015;8:ra130.
    1. Kolch W, Halasz M, Granovskaya M, Kholodenko BN. The dynamic control of signal transduction networks in cancer cells. Nat. Rev. Cancer. 2015;15:515–527.
    1. Furman D, et al. Expression of specific inflammasome gene modules stratifies older individuals into two extreme clinical and immunological states. Nat. Med. 2017;23:174–184.
    1. Hernandez-Vargas EA, et al. Effects of aging on influenza virus infection dynamics. J. Virol. 2014;88:4123–4131.
    1. Rangel C, et al. Modeling T-cell activation using gene expression profiling and state-space models. Bioinformatics. 2004;20:1361–1372.
    1. Huang S, Eichler G, Bar-Yam Y, Ingber DE. Cell fates as high-dimensional attractor states of a complex gene regulatory network. Phys. Rev. Lett. 2005;94:128701.
    1. Mar JC, Quackenbush J. Decomposition of gene expression state space trajectories. PLoS. Comput. Biol. 2009;5:e1000626.
    1. Kauffman SA. Metabolic stability and epigenesis in randomly constructed genetic nets. J. Theor. Biol. 1969;22:437–467.
    1. Huang AC, Hu L, Kauffman SA, Zhang W, Shmulevich I. Using cell fate attractors to uncover transcriptional regulation of HL60 neutrophil differentiation. BMC Syst. Biol. 2009;3:20.
    1. Taherian Fard A, Ragan MA. Modeling the attractor landscape of disease progression: a network-based approach. Front. Genet. 2017;8:48.
    1. Huang S, Ernberg I, Kauffman S. Cancer attractors: a systems view of tumors from a gene network dynamics and developmental perspective. Semin. Cell. Dev. Biol. 2009;20:869–876.
    1. del Sol A, Balling R, Hood L, Galas D. Diseases as network perturbations. Curr. Opin. Biotechnol. 2010;21:566–571.
    1. Auger-Methe M, et al. State-space models’ dirty little secrets: even simple linear Gaussian models can have estimation problems. Sci. Rep. 2016;6:26677.
    1. Liitiäinen, E. & Lendasse, A. In Artificial Neural Networks – ICANN 2006: 16th International Conference, Athens, Greece, September 10-14, 2006. Proceedings, Part II (eds Kollias, S., Stafylopatis, A., Duch, W. & Oja, E.) 181–190 (Springer, Berlin, 2006).
    1. Riiser A. The human microbiome, asthma, and allergy. Allergy Asthma Clin. Immunol. 2015;11:35.
    1. Knoll RL, et al. Gut microbiota differs between children with Inflammatory Bowel Disease and healthy siblings in taxonomic and functional composition: a metagenomic analysis. Am. J. Physiol. Gastrointest. Liver Physiol. 2017;312:G327–G339.
    1. Haahtela T, et al. The biodiversity hypothesis and allergic disease: world allergy organization position statement. World Allergy Organ J. 2013;6:3.
    1. Blaser M, Bork P, Fraser C, Knight R, Wang J. The microbiome explored: recent insights and future challenges. Nat. Rev. Microbiol. 2013;11:213–217.
    1. Saulnier DM, et al. Gastrointestinal microbiome signatures of pediatric patients with irritable bowel syndrome. Gastroenterology. 2011;141:1782–1791.
    1. Tedjo DI, et al. The fecal microbiota as a biomarker for disease activity in Crohn’s disease. Sci. Rep. 2016;6:35216.
    1. Cui L, et al. The microbiome and the lung. Ann. Am. Thorac. Soc. 2014;11:S227–S232.
    1. Hilty M, et al. Disordered microbial communities in asthmatic airways. PLoS. ONE. 2010;5:e8578.
    1. Slater M, et al. The impact of azithromycin therapy on the airway microbiota in asthma. Thorax. 2014;69:673–674.
    1. Huang YJ, et al. Airway microbiota and bronchial hyperresponsiveness in patients with suboptimally controlled asthma. J. Allergy Clin. Immunol. 2011;127:372–381.
    1. Hanski I, et al. Environmental biodiversity, human microbiota, and allergy are interrelated. Proc. Natl. Acad. Sci. USA. 2012;109:8334–8339.
    1. Kong HH, et al. Temporal shifts in the skin microbiome associated with disease flares and treatment in children with atopic dermatitis. Genome Res. 2012;22:850–859.
    1. Fahlen A, Engstrand L, Baker BS, Powles A, Fry L. Comparison of bacterial microbiota in skin biopsies from normal and psoriatic skin. Arch. Dermatol. Res. 2012;304:15–22.
    1. Zhu A, Sunagawa S, Mende DR, Bork P. Inter-individual differences in the gene content of human gut bacterial species. Genome Biol. 2015;16:82.
    1. Ashley EA. Towards precision medicine. Nat. Rev. Genet. 2016;17:507–522.
    1. Raphael BJ, Dobson JR, Oesper L, Vandin F. Identifying driver mutations in sequenced cancer genomes: computational approaches to enable precision medicine. Genome Med. 2014;6:5.
    1. Johnson CH, Ivanisevic J, Siuzdak G. Metabolomics: beyond biomarkers and towards mechanisms. Nat. Rev. Mol. Cell. Biol. 2016;17:451–459.
    1. Armitage EG, Barbas C. Metabolomics in cancer biomarker discovery: current trends and future perspectives. J. Pharm. Biomed. Anal. 2014;87:1–11.
    1. Jansson J, et al. Metabolomics reveals metabolic biomarkers of Crohn’s disease. PLoS. ONE. 2009;4:e6386.
    1. Guma M, Tiziani S, Firestein GS. Metabolomics in rheumatic diseases: desperately seeking biomarkers. Nat. Rev. Rheumatol. 2016;12:269–281.
    1. Gaber T, Strehl C, Buttgereit F. Metabolic regulation of inflammation. Nat. Rev. Rheumatol. 2017;13:267–279.
    1. Mak TW, et al. Glutathione Primes T Cell Metabolism for Inflammation. Immunity. 2017;46:675–689.
    1. Menche J, et al. Integrating personalized gene expression profiles into predictive disease-associated gene pools. npj Syst. Biol. Appl. 2017;3:10.
    1. Hewett R, Kijsanayothin P. Tumor classification ranking from microarray data. BMC Genom. 2008;9:S21.
    1. Pusztai L, Hatzis C, Andre F. Reproducibility of research and preclinical validation: problems and solutions. Nat. Rev. Clin. Oncol. 2013;10:720–724.
    1. Vargas AJ, Harris CC. Biomarker development in the precision medicine era: lung cancer as a case study. Nat. Rev. Cancer. 2016;16:525–537.
    1. Bradbury A, Pluckthun A. Reproducibility: Standardize antibodies used in research. Nature. 2015;518:27–29.
    1. Wilkinson MD, et al. The FAIR Guiding Principles for scientific data management and stewardship. Sci. Data. 2016;3:160018.
    1. Houdijk WP, Van Den Besselaar AM. International multicenter international sensitivity index (ISI) calibration of a new human tissue factor thromboplastin reagent derived from cultured human cells. J. Thromb. Haemost. 2004;2:266–270.
    1. Seita J, et al. Gene Expression Commons: an open platform for absolute gene expression profiling. PLoS. ONE. 2012;7:e40321.
    1. Liu X, Wang Y, Ji H, Aihara K, Chen L. Personalized characterization of diseases using sample-specific networks. Nucleic Acids Res. 2016;44:e164.
    1. He FQ, et al. Identification of molecular sub-networks associated with cell survival in a chronically SIVmac-infected human CD4+ T cell line. Virol. J. 2014;11:152.
    1. Archer SN, et al. Mistimed sleep disrupts circadian regulation of the human transcriptome. Proc. Natl. Acad. Sci. USA. 2014;111:E682–E691.
    1. Kulasingam V, Diamandis EP. Strategies for discovering novel cancer biomarkers through utilization of emerging technologies. Nat. Clin. Pract. Oncol. 2008;5:588–599.
    1. Hanash S, Taguchi A. The grand challenge to decipher the cancer proteome. Nat. Rev. Cancer. 2010;10:652–660.
    1. Daugaard I, et al. Identification and validation of candidate epigenetic biomarkers in lung adenocarcinoma. Sci. Rep. 2016;6:35807.
    1. Plant D, Wilson AG, Barton A. Genetic and epigenetic predictors of responsiveness to treatment in RA. Nat. Rev. Rheumatol. 2014;10:329–337.
    1. Meystre, S. M. et al. Clinical data reuse or secondary use: current status and potential future progress. Yearb. Med. Inform. (2017).
    1. Mizuno S, et al. AlzPathway: a comprehensive map of signaling pathways of Alzheimer’s disease. Bmc. Syst. Biol. 2012;6:52.
    1. Fujita KA, et al. Integrating pathways of Parkinson’s disease in a molecular interaction map. Mol. Neurobiol. 2014;49:88–102.
    1. Hasin Y, Seldin M, Lusis A. Multi-omics approaches to disease. Genome Biol. 2017;18:83.
    1. Arai E, et al. Multilayer-omics analysis of renal cell carcinoma, including the whole exome, methylome and transcriptome. Int. J. Cancer. 2014;135:1330–1342.
    1. Stanford NJ, et al. The evolution of standards and data management practices in systems biology. Mol. Syst. Biol. 2015;11:851.
    1. The power of big data must be harnessed for medical progress. Nature539, 467–468, (2016).
    1. Arneric SP, et al. Biometric monitoring devices for assessing end points in clinical trials: developing an ecosystem. Nat. Rev. Drug. Discov. 2017;16:736.
    1. Schadt EE, Linderman MD, Sorenson J, Lee L, Nolan GP. Computational solutions to large-scale data management and analysis. Nat. Rev. Genet. 2010;11:647–657.
    1. Scheufele E, et al. tranSMART: an open source knowledge management and high content data analytics platform. AMIA Jt. Summits Transl. Sci. Proc. 2014;2014:96–101.
    1. Herzinger S, et al. SmartR: an open-source platform for interactive visual analytics for translational research data. Bioinformatics. 2017;33:2229–2231.
    1. Wolstencroft K, et al. FAIRDOMHub: a repository and collaboration environment for sharing systems biology research. Nucleic Acids Res. 2017;45:D404–D407.
    1. Durinx, C. et al. Identifying ELIXIR core data resources. F1000Res, 10.12688/f1000research.9656.2 (2016).
    1. Vincent JL, Creteur J. The hospital of tomorrow in 10 points. Crit. Care. 2017;21:93.
    1. Budin-Ljosne I, et al. Dynamic consent: a potential solution to some of the challenges of modern biomedical research. BMC Med. Ethic-. 2017;18:4.
    1. Ye H, Chen ES. Attribute utility motivated k-anonymization of datasets to support the heterogeneous needs of biomedical researchers. Amia. Annu. Symp. Proc. 2011;2011:1573–1582.
    1. Aggarwal, C. C. & Yu, P. S. (eds). A General Survey of Privacy-Preserving Data Mining Models and Algorithms. In Privacy-Preserving Data Mining: Models and Algorithms 11–52 (Springer, Boston, MA, USA, 2008).
    1. Hayden EC. Privacy protections: the genome hacker. Nature. 2013;497:172–174.
    1. Henderson BE, Lee NH, Seewaldt V, Shen H. The influence of race and ethnicity on the biology of cancer. Nat. Rev. Cancer. 2012;12:648–653.
    1. de la Hera B, et al. Natalizumab-related anaphylactoid reactions in MS patients are associated with HLA class II alleles. Neurol. Neuroimmunol. Neuroinflamm. 2014;1:e47.

Source: PubMed

3
Iratkozz fel