Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.260
Filter
Add more filters

Publication year range
1.
Cell ; 177(1): 45-57, 2019 03 21.
Article in English | MEDLINE | ID: mdl-30901547

ABSTRACT

In the wake of the Human Genome Project (HGP), strong expectations were set for the timeline and impact of genomics on medicine-an anticipated transformation in the diagnosis, treatment, and prevention of disease. In this Perspective, we take stock of the nascent field of genomic medicine. In what areas, if any, is genomics delivering on this promise, or is the path to success clear? Where are we falling short, and why? What have been the unanticipated developments? Overall, we argue that the optimism surrounding the transformational potential of genomics on medicine remains justified, albeit with a considerably different form and timescale than originally projected. We also argue that the field needs to pivot back to basics, as understanding the entirety of the genotype-to-phenotype equation is a likely prerequisite for delivering on the full potential of the human genome to advance the human condition.


Subject(s)
Genome, Human/genetics , Precision Medicine/methods , Precision Medicine/trends , Genetic Testing , Genomics/methods , Genomics/trends , Human Genome Project , Humans
2.
Cell ; 177(1): 58-69, 2019 03 21.
Article in English | MEDLINE | ID: mdl-30901549

ABSTRACT

Personalized medicine has largely been enabled by the integration of genomic and other data with electronic health records (EHRs) in the United States and elsewhere. Increased EHR adoption across various clinical settings and the establishment of EHR-linked population-based biobanks provide unprecedented opportunities for the types of translational and implementation research that drive personalized medicine. We review advances in the digitization of health information and the proliferation of genomic research in health systems and provide insights into emerging paths for the widespread implementation of personalized medicine.


Subject(s)
Electronic Health Records/trends , Precision Medicine/methods , Precision Medicine/trends , Genetic Testing , Genome, Human/genetics , Genomics/methods , Genomics/trends , Humans , United States
3.
Cell ; 177(1): 38-44, 2019 03 21.
Article in English | MEDLINE | ID: mdl-30901546

ABSTRACT

Innovative analytical frameworks are required to capture the complex gene-environment interactions. We investigate the insufficiency of commonly used models for disease genome analysis and suggest considering genetic interactions in complex diseases. For non-genetic factors, we study the emerging wearable technologies that have enabled quantification of physiological and environmental factors at an unprecedented breadth and depth. We propose a Bayesian framework to hierarchically model personalized gene-environmental interaction to enable precision health and medicine.


Subject(s)
Precision Medicine/methods , Precision Medicine/trends , Wearable Electronic Devices/trends , Bayes Theorem , Epistasis, Genetic/genetics , Gene-Environment Interaction , Genome-Wide Association Study/methods , Humans , Multifactorial Inheritance/genetics
4.
Cell ; 173(3): 535-539, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29677503

ABSTRACT

The integration of proteomics into precision oncology presents opportunities that may transform the molecular analysis of cancer and accelerate basic and clinical cancer research. This Commentary discusses the importance of international collaboration and data sharing inspired by the Cancer Moonshot to accelerate the progress of multi-omic precision medicine-an approach that addresses the global diversity of people and of cancers.


Subject(s)
Genomics/methods , Medical Oncology/trends , Neoplasms/genetics , Neoplasms/therapy , Precision Medicine/trends , Proteomics/methods , Female , Genome, Human , Global Health , Humans , Male , Medical Oncology/methods , Mutation , Precision Medicine/methods , SEER Program , United States
5.
Nature ; 632(8027): 1145-1154, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38862028

ABSTRACT

Spaceflight induces molecular, cellular and physiological shifts in astronauts and poses myriad biomedical challenges to the human body, which are becoming increasingly relevant as more humans venture into space1-6. Yet current frameworks for aerospace medicine are nascent and lag far behind advancements in precision medicine on Earth, underscoring the need for rapid development of space medicine databases, tools and protocols. Here we present the Space Omics and Medical Atlas (SOMA), an integrated data and sample repository for clinical, cellular and multi-omic research profiles from a diverse range of missions, including the NASA Twins Study7, JAXA CFE study8,9, SpaceX Inspiration4 crew10-12, Axiom and Polaris. The SOMA resource represents a more than tenfold increase in publicly available human space omics data, with matched samples available from the Cornell Aerospace Medicine Biobank. The Atlas includes extensive molecular and physiological profiles encompassing genomics, epigenomics, transcriptomics, proteomics, metabolomics and microbiome datasets, which reveal some consistent features across missions, including cytokine shifts, telomere elongation and gene expression changes, as well as mission-specific molecular responses and links to orthologous, tissue-specific mouse datasets. Leveraging the datasets, tools and resources in SOMA can help to accelerate precision aerospace medicine, bringing needed health monitoring, risk mitigation and countermeasure data for upcoming lunar, Mars and exploration-class missions.


Subject(s)
Aerospace Medicine , Astronauts , Biological Specimen Banks , Databases, Factual , Internationality , Space Flight , Animals , Female , Humans , Male , Mice , Aerospace Medicine/methods , Atlases as Topic , Cytokines/metabolism , Datasets as Topic , Epigenomics , Gene Expression Profiling , Genomics , Metabolomics , Microbiota/genetics , Multiomics , Organ Specificity , Precision Medicine/trends , Proteomics , Space Flight/statistics & numerical data , Telomere/metabolism , Twins
6.
Nature ; 632(8027): 995-1008, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38862027

ABSTRACT

The recent acceleration of commercial, private and multi-national spaceflight has created an unprecedented level of activity in low Earth orbit, concomitant with the largest-ever number of crewed missions entering space and preparations for exploration-class (lasting longer than one year) missions. Such rapid advancement into space from many new companies, countries and space-related entities has enabled a 'second space age'. This era is also poised to leverage, for the first time, modern tools and methods of molecular biology and precision medicine, thus enabling precision aerospace medicine for the crews. The applications of these biomedical technologies and algorithms are diverse, and encompass multi-omic, single-cell and spatial biology tools to investigate human and microbial responses to spaceflight. Additionally, they extend to the development of new imaging techniques, real-time cognitive assessments, physiological monitoring and personalized risk profiles tailored for astronauts. Furthermore, these technologies enable advancements in pharmacogenomics, as well as the identification of novel spaceflight biomarkers and the development of corresponding countermeasures. In this Perspective, we highlight some of the recent biomedical research from the National Aeronautics and Space Administration, Japan Aerospace Exploration Agency, European Space Agency and other space agencies, and detail the entrance of the commercial spaceflight sector (including SpaceX, Blue Origin, Axiom and Sierra Space) into aerospace medicine and space biology, the first aerospace medicine biobank, and various upcoming missions that will utilize these tools to ensure a permanent human presence beyond low Earth orbit, venturing out to other planets and moons.


Subject(s)
Aerospace Medicine , Astronauts , Multiomics , Space Flight , Humans , Aerospace Medicine/methods , Aerospace Medicine/trends , Biological Specimen Banks , Biomarkers/metabolism , Biomarkers/analysis , Cognition , Internationality , Monitoring, Physiologic/methods , Monitoring, Physiologic/trends , Multiomics/methods , Multiomics/trends , Pharmacogenetics/methods , Pharmacogenetics/trends , Precision Medicine/methods , Precision Medicine/trends , Space Flight/methods , Space Flight/trends
7.
CA Cancer J Clin ; 71(2): 107-139, 2021 03.
Article in English | MEDLINE | ID: mdl-33326126

ABSTRACT

We are experiencing a revolution in cancer. Advances in screening, targeted and immune therapies, big data, computational methodologies, and significant new knowledge of cancer biology are transforming the ways in which we prevent, detect, diagnose, treat, and survive cancer. These advances are enabling durable progress in the goal to achieve personalized cancer care. Despite these gains, more work is needed to develop better tools and strategies to limit cancer as a major health concern. One persistent gap is the inconsistent coordination among researchers and caregivers to implement evidence-based programs that rely on a fuller understanding of the molecular, cellular, and systems biology mechanisms underpinning different types of cancer. Here, the authors integrate conversations with over 90 leading cancer experts to highlight current challenges, encourage a robust and diverse national research portfolio, and capture timely opportunities to advance evidence-based approaches for all patients with cancer and for all communities.


Subject(s)
Evidence-Based Medicine/organization & administration , Mass Screening/organization & administration , Medical Oncology/organization & administration , Neoplasms/therapy , Professional Practice Gaps , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Cost of Illness , Early Detection of Cancer/methods , Early Detection of Cancer/trends , Evidence-Based Medicine/methods , Evidence-Based Medicine/trends , Humans , Mass Screening/methods , Mass Screening/trends , Medical Oncology/methods , Medical Oncology/trends , Neoplasms/diagnosis , Neoplasms/genetics , Neoplasms/mortality , Precision Medicine/methods , Precision Medicine/trends , United States/epidemiology
8.
Mol Cell ; 78(6): 1002-1018, 2020 06 18.
Article in English | MEDLINE | ID: mdl-32559422

ABSTRACT

We are witnessing several revolutionary technological advances in cancer. These innovations have not only contributed to a growing understanding of the tumor and its microenvironment but also uncovered an increasing array of new therapeutic targets. For most advanced cancers, therapy resistance limits the benefit of single-agent therapies. Therefore, some 5,000 clinical trials are ongoing globally to probe the clinical benefit of new combination treatments. However, the possibilities to combine individual treatments dramatically outnumber the patients available to enroll in clinical trials. This comes at a potential cost of missed opportunities, clinical failure, avoidable toxicity, insufficient patient accrual, and financial loss. A solution may be to design combination therapies more rationally, which are informed by fundamental biological and mechanistic insight. We will discuss some successes and failures of current treatment combinations, as well as interesting emerging preclinical concepts that warrant clinical exploration.


Subject(s)
Drug Therapy, Combination/trends , Neoplasms/therapy , Designer Drugs/therapeutic use , Humans , Neoplasms/metabolism , Precision Medicine/methods , Precision Medicine/trends , Tumor Microenvironment/drug effects
9.
Hum Genomics ; 18(1): 78, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987819

ABSTRACT

Pharmacogenetics investigates sequence of genes that affect drug response, enabling personalized medication. This approach reduces drug-induced adverse reactions and improves clinical effectiveness, making it a crucial consideration for personalized medical care. Numerous guidelines, drawn by global consortia and scientific organizations, codify genotype-driven administration for over 120 active substances. As the scientific community acknowledges the benefits of genotype-tailored therapy over traditionally agnostic drug administration, the push for its implementation into Italian healthcare system is gaining momentum. This evolution is influenced by several factors, including the improved access to patient genotypes, the sequencing costs decrease, the growing of large-scale genetic studies, the rising popularity of direct-to-consumer pharmacogenetic tests, and the continuous improvement of pharmacogenetic guidelines. Since EMA (European Medicines Agency) and AIFA (Italian Medicines Agency) provide genotype information on drug leaflet without clear and explicit clinical indications for gene testing, the regulation of pharmacogenetic testing is a pressing matter in Italy. In this manuscript, we have reviewed how to overcome the obstacles in implementing pharmacogenetic testing in the clinical practice of the Italian healthcare system. Our particular emphasis has been on germline testing, given the absence of well-defined national directives in contrast to somatic pharmacogenetics.


Subject(s)
Pharmacogenetics , Humans , Italy , Pharmacogenetics/methods , Pharmacogenetics/trends , Precision Medicine/trends , Precision Medicine/methods , Pharmacogenomic Testing/methods , Genotype
18.
Cereb Cortex ; 34(13): 8-18, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38696602

ABSTRACT

Noninvasive brain stimulation (NIBS) has been increasingly investigated during the last decade as a treatment option for persons with autism spectrum disorder (ASD). Yet, previous studies did not reach a consensus on a superior treatment protocol or stimulation target. Persons with ASD often suffer from social isolation and high rates of unemployment, arising from difficulties in social interaction. ASD involves multiple neural systems involved in perception, language, and cognition, and the underlying brain networks of these functional domains have been well documented. Aiming to provide an overview of NIBS effects when targeting these neural systems in late adolescent and adult ASD, we conducted a systematic search of the literature starting at 631 non-duplicate publications, leading to six studies corresponding with inclusion and exclusion criteria. We discuss these studies regarding their treatment rationale and the accordingly chosen methodological setup. The results of these studies vary, while methodological advances may allow to explain some of the variability. Based on these insights, we discuss strategies for future clinical trials to personalize the selection of brain stimulation targets taking into account intersubject variability of brain anatomy as well as function.


Subject(s)
Brain , Humans , Adult , Autism Spectrum Disorder/therapy , Precision Medicine/methods , Precision Medicine/trends , Transcranial Magnetic Stimulation/methods , Autistic Disorder/therapy , Autistic Disorder/physiopathology , Autistic Disorder/psychology , Transcranial Direct Current Stimulation/methods
19.
Eur Heart J ; 45(35): 3204-3218, 2024 Sep 14.
Article in English | MEDLINE | ID: mdl-38976371

ABSTRACT

The advent of digital health and artificial intelligence (AI) has promised to revolutionize clinical care, but real-world patient evaluation has yet to witness transformative changes. As history taking and physical examination continue to rely on long-established practices, a growing pipeline of AI-enhanced digital tools may soon augment the traditional clinical encounter into a data-driven process. This article presents an evidence-backed vision of how promising AI applications may enhance traditional practices, streamlining tedious tasks while elevating diverse data sources, including AI-enabled stethoscopes, cameras, and wearable sensors, to platforms for personalized medicine and efficient care delivery. Through the lens of traditional patient evaluation, we illustrate how digital technologies may soon be interwoven into routine clinical workflows, introducing a novel paradigm of longitudinal monitoring. Finally, we provide a skeptic's view on the practical, ethical, and regulatory challenges that limit the uptake of such technologies.


Subject(s)
Artificial Intelligence , Digital Health , Humans , Artificial Intelligence/trends , Digital Health/trends , Physical Examination/instrumentation , Physical Examination/methods , Physical Examination/trends , Precision Medicine/instrumentation , Precision Medicine/methods , Precision Medicine/trends
20.
J Hepatol ; 81(4): 756-762, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38871125

ABSTRACT

Primary liver tumours, including benign liver tumours, hepatocellular carcinoma and cholangiocarcinoma, present a multifaceted challenge, necessitating a collaborative approach, as evidenced by the role of the multidisciplinary tumour board (MDTB). The approach to managing primary liver tumours involves specialised teams, including surgeons, radiologists, oncologists, pathologists, hepatologists, and radiation oncologists, coming together to propose individualised treatment plans. The evolving landscape of primary liver cancer treatment introduces complexities, particularly with the expanding array of systemic and locoregional therapies, alongside the potential integration of molecular biology and artificial intelligence (AI) into MDTBs in the future. Precision medicine demands collaboration across disciplines, challenging traditional frameworks. In the next decade, we anticipate the convergence of AI, molecular biology, pathology, and advanced imaging, requiring adaptability in MDTB structure to incorporate these cutting-edge technologies. Navigating this evolution also requires a focus on enhancing basic, translational, and clinical research, as well as boosting clinical trials through an upgraded use of MDTBs as hubs for scientific collaboration and raising literacy about AI and new technologies. In this review, we will delineate the current unmet needs in the clinical management of primary liver cancers, discuss our perspective on the future role of MDTBs in primary liver cancers ("next generation" MDTBs), and unravel the potential power and limitations of novel technologies that may shape the multidisciplinary care landscape for primary liver cancers in the coming decade.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Patient Care Team , Precision Medicine , Humans , Liver Neoplasms/therapy , Liver Neoplasms/pathology , Patient Care Team/trends , Precision Medicine/methods , Precision Medicine/trends , Carcinoma, Hepatocellular/therapy , Artificial Intelligence/trends , Cholangiocarcinoma/therapy
SELECTION OF CITATIONS
SEARCH DETAIL