Precision Medicine in Clinical Practice

A special issue of Journal of Personalized Medicine (ISSN 2075-4426).

Deadline for manuscript submissions: closed (10 December 2022) | Viewed by 30915

Special Issue Editors


E-Mail
Guest Editor
Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
Interests: clinical implementation of precision medicine; clinical and economic outcomes related to the use of precision medicine, including the impact of value-based contracts; policy, legal, ethical, economic and societal issues of precision medicine in clinical practice, including disparities in access to precision medicine and outcomes, and ethical implications
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Guest Editor
Associate Professor and Director of Geisinger Genomic Medicine Institute, Danville, PA, USA
Interests: clinical outcomes of genomic screening; interventions to improve risk management for inherited disease risk; access to genomic medicine

Special Issue Information

Dear Colleagues,

Precision medicine is a dynamic area embracing diverse approaches that allow the targeting of new medicines, screening programs or preventive healthcare strategies, including the use of genomic technologies, also potentially taking account of patient preferences. Health insurance plans are increasingly considering different precision medicine interventions for coverage and reimbursement. Precision medicine interventions are also being integrated into clinical care for a range of medical conditions and purposes. The Journal of Personalized Medicine aims to publish a collection of articles that explore challenges and opportunities in the field and fresh insights to ongoing discussions and debate in precision medicine from the US and abroad. We will consider original research, systematic reviews, and well-designed case studies and analyses that report empirical work presenting experiences and perspectives about integrating precision medicine in clinical practice.

Dr. Christine Lu
Prof. Adam Buchanan
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Journal of Personalized Medicine is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2600 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • Access
  • Affordability
  • Sustainability
  • Healthcare system
  • Pharmacogenomics
  • Genomics
  • Polygenic risk scores
  • Genome and exome sequencing
  • Genome screening

Published Papers (8 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

19 pages, 720 KiB  
Article
An Idealized Clinicogenomic Registry to Engage Underrepresented Populations Using Innovative Technology
by Patrick Silva, Deborah Vollmer Dahlke, Matthew Lee Smith, Wendy Charles, Jorge Gomez, Marcia G. Ory and Kenneth S. Ramos
J. Pers. Med. 2022, 12(5), 713; https://doi.org/10.3390/jpm12050713 - 29 Apr 2022
Cited by 7 | Viewed by 3050
Abstract
Current best practices in tumor registries provide a glimpse into a limited time frame over the natural history of disease, usually a narrow window around diagnosis and biopsy. This creates challenges meeting public health and healthcare reimbursement policies that increasingly require robust documentation [...] Read more.
Current best practices in tumor registries provide a glimpse into a limited time frame over the natural history of disease, usually a narrow window around diagnosis and biopsy. This creates challenges meeting public health and healthcare reimbursement policies that increasingly require robust documentation of long-term clinical trajectories, quality of life, and health economics outcomes. These challenges are amplified for underrepresented minority (URM) and other disadvantaged populations, who tend to view the institution of clinical research with skepticism. Participation gaps leave such populations underrepresented in clinical research and, importantly, in policy decisions about treatment choices and reimbursement, thus further augmenting health, social, and economic disparities. Cloud computing, mobile computing, digital ledgers, tokenization, and artificial intelligence technologies are powerful tools that promise to enhance longitudinal patient engagement across the natural history of disease. These tools also promise to enhance engagement by giving participants agency over their data and addressing a major impediment to research participation. This will only occur if these tools are available for use with all patients. Distributed ledger technologies (specifically blockchain) converge these tools and offer a significant element of trust that can be used to engage URM populations more substantively in clinical research. This is a crucial step toward linking composite cohorts for training and optimization of the artificial intelligence tools for enhancing public health in the future. The parameters of an idealized clinical genomic registry are presented. Full article
(This article belongs to the Special Issue Precision Medicine in Clinical Practice)
Show Figures

Figure 1

14 pages, 1499 KiB  
Article
Real-World Impact of a Pharmacogenomics-Enriched Comprehensive Medication Management Program
by Joseph P. Jarvis, Arul Prakasam Peter, Murray Keogh, Vince Baldasare, Gina M. Beanland, Zachary T. Wilkerson, Steven Kradel and Jeffrey A. Shaman
J. Pers. Med. 2022, 12(3), 421; https://doi.org/10.3390/jpm12030421 - 08 Mar 2022
Cited by 18 | Viewed by 11117
Abstract
The availability of clinical decision support systems (CDSS) and other methods for personalizing medicine now allows evaluation of their real-world impact on healthcare delivery. For example, addressing issues associated with polypharmacy in older patients using pharmacogenomics (PGx) and comprehensive medication management (CMM) is [...] Read more.
The availability of clinical decision support systems (CDSS) and other methods for personalizing medicine now allows evaluation of their real-world impact on healthcare delivery. For example, addressing issues associated with polypharmacy in older patients using pharmacogenomics (PGx) and comprehensive medication management (CMM) is thought to hold great promise for meaningful improvements across the goals of the Quadruple Aim. However, few studies testing these tools at scale, using relevant system-wide metrics, and under real-world conditions, have been published to date. Here, we document a reduction of ~$7000 per patient in direct medical charges (a total of $37 million over 5288 enrollees compared to 22,357 non-enrolled) in Medicare Advantage patients (≥65 years) receiving benefits through a state retirement system over the first 32 months of a voluntary PGx-enriched CMM program. We also observe a positive shift in healthcare resource utilization (HRU) away from acute care services and toward more sustainable and cost-effective primary care options. Together with improvements in medication risk assessment, patient/provider communication via pharmacist-mediated medication action plans (MAP), and the sustained positive trends in HRU, we suggest these results validate the use of a CDSS to unify PGx and CMM to optimize care for this and similar patient populations. Full article
(This article belongs to the Special Issue Precision Medicine in Clinical Practice)
Show Figures

Figure 1

11 pages, 1136 KiB  
Article
Education and Training of Non-Genetics Providers on the Return of Genome Sequencing Results in a NICU Setting
by Kelly M. East, Meagan E. Cochran, Whitley V. Kelley, Veronica Greve, Candice R. Finnila, Tanner Coleman, Mikayla Jennings, Latonya Alexander, Elizabeth J. Rahn, Maria I. Danila, Greg Barsh, Bruce Korf and Greg Cooper
J. Pers. Med. 2022, 12(3), 405; https://doi.org/10.3390/jpm12030405 - 05 Mar 2022
Cited by 14 | Viewed by 2016
Abstract
To meet current and expected future demand for genome sequencing in the neonatal intensive care unit (NICU), adjustments to traditional service delivery models are necessary. Effective programs for the training of non-genetics providers (NGPs) may address the known barriers to providing genetic services [...] Read more.
To meet current and expected future demand for genome sequencing in the neonatal intensive care unit (NICU), adjustments to traditional service delivery models are necessary. Effective programs for the training of non-genetics providers (NGPs) may address the known barriers to providing genetic services including limited genetics knowledge and lack of confidence. The SouthSeq project aims to use genome sequencing to make genomic diagnoses in the neonatal period and evaluate a scalable approach to delivering genome sequencing results to populations with limited access to genetics professionals. Thirty-three SouthSeq NGPs participated in a live, interactive training intervention and completed surveys before and after participation. Here, we describe the protocol for the provider training intervention utilized in the SouthSeq study and the associated impact on NGP knowledge and confidence in reviewing, interpreting, and using genome sequencing results. Participation in the live training intervention led to an increased level of confidence in critical skills needed for real-world implementation of genome sequencing. Providers reported a significant increase in confidence level in their ability to review, understand, and use genome sequencing result reports to guide patient care. Reported barriers to implementation of genome sequencing in a NICU setting included test cost, lack of insurance coverage, and turn around time. As implementation of genome sequencing in this setting progresses, effective education of NGPs is critical to provide access to high-quality and timely genomic medicine care. Full article
(This article belongs to the Special Issue Precision Medicine in Clinical Practice)
Show Figures

Figure 1

11 pages, 1652 KiB  
Communication
Clin.iobio: A Collaborative Diagnostic Workflow to Enable Team-Based Precision Genomics
by Alistair Ward, Matt Velinder, Tonya Di Sera, Aditya Ekawade, Sabrina Malone Jenkins, Barry Moore, Rong Mao, Pinar Bayrak-Toydemir and Gabor Marth
J. Pers. Med. 2022, 12(1), 73; https://doi.org/10.3390/jpm12010073 - 08 Jan 2022
Cited by 1 | Viewed by 2540
Abstract
The primary goal of precision genomics is the identification of causative genetic variants in targeted or whole-genome sequencing data. The ultimate clinical hope is that these findings lead to an efficacious change in treatment for the patient. In current clinical practice, these findings [...] Read more.
The primary goal of precision genomics is the identification of causative genetic variants in targeted or whole-genome sequencing data. The ultimate clinical hope is that these findings lead to an efficacious change in treatment for the patient. In current clinical practice, these findings are typically returned by expert analysts as static, text-based reports. Ideally, these reports summarize the quality of the data obtained, integrate known gene–phenotype associations, follow allele segregation and affected status within the sequenced samples, and weigh computational evidence of pathogenicity. These findings are used to prioritize the variant(s) most likely to cause the given patient’s phenotypes. In most diagnostic settings, a team of experts contribute to these reports, including bioinformaticians, clinicians, and genetic counselors, among others. However, these experts often do not have the necessary tools to review genomic findings, test genetic hypotheses, or query specific gene and variant information. Additionally, team members often rely on different tools and methods based on their given expertise, resulting in further difficulties in communicating and discussing genomic findings. Here, we present clin.iobio—a web-based solution to collaborative genomic analysis that enables diagnostic team members to focus on their area of expertise within the diagnostic process, while allowing them to easily review and contribute to all steps of the diagnostic process. Clin.iobio integrates tools from the popular iobio genomic visualization suite into a comprehensive diagnostic workflow, encompassing (1) genomic data quality review, (2) dynamic phenotype-driven gene prioritization, (3) variant prioritization using a comprehensive set of knowledge bases and annotations, (4) and an exportable findings summary. In conclusion, clin.iobio is a comprehensive solution to team-based precision genomics, the findings of which stand to inform genomic considerations in clinical practice. Full article
(This article belongs to the Special Issue Precision Medicine in Clinical Practice)
Show Figures

Figure 1

13 pages, 542 KiB  
Article
Feasibility of a Traceback Approach for Using Pathology Specimens to Facilitate Genetic Testing in the Genetic Risk Analysis in Ovarian Cancer (GRACE) Study Protocol
by Tia L. Kauffman, Yolanda K. Prado, Ana A. Reyes, Jamilyn M. Zepp, Jennifer Sawyer, Larissa Lee White, Jessica Martucci, Suzanne Bianca Salas, Sarah Vertrees, Alan F. Rope, Sheila Weinmann, Nora B. Henrikson, Sandra Soo-Jin Lee, Heather Spencer Feigelson and Jessica Ezzell Hunter
J. Pers. Med. 2021, 11(11), 1194; https://doi.org/10.3390/jpm11111194 - 13 Nov 2021
Cited by 2 | Viewed by 2176
Abstract
Guidelines currently state that genetic testing is clinically indicated for all individuals diagnosed with ovarian cancer. Individuals with a prior diagnosis of ovarian cancer who have not received genetic testing represent missed opportunities to identify individuals with inherited high-risk cancer variants. For deceased [...] Read more.
Guidelines currently state that genetic testing is clinically indicated for all individuals diagnosed with ovarian cancer. Individuals with a prior diagnosis of ovarian cancer who have not received genetic testing represent missed opportunities to identify individuals with inherited high-risk cancer variants. For deceased individuals, post-mortem genetic testing of pathology specimens allows surviving family members to receive important genetic risk information. The Genetic Risk Assessment in Ovarian Cancer (GRACE) study aims to address this significant healthcare gap using a “traceback testing” approach to identify individuals with a prior diagnosis of ovarian cancer and offer genetic risk information to them and their family members. This study will assess the potential ethical and privacy concerns related to an ovarian cancer traceback testing approach in the context of patients who are deceased, followed by implementation and evaluation of the feasibility of an ovarian cancer traceback testing approach using tumor registries and archived pathology tissue. Descriptive and statistical analyses will assess health system and patient characteristics associated with the availability of pathology tissue and compare the ability to contact and uptake of genetic testing between patients who are living and deceased. The results of this study will inform the implementation of future traceback programs. Full article
(This article belongs to the Special Issue Precision Medicine in Clinical Practice)
Show Figures

Figure 1

12 pages, 446 KiB  
Article
Population Genomic Screening for Genetic Etiologies of Neurodevelopmental/Psychiatric Disorders Demonstrates Personal Utility and Positive Participant Responses
by Karen E. Wain, Kasia Tolwinski, Emily Palen, Alexis R. Heidlebaugh, Karahlyn Holdren, Lauren Kasparson Walsh, Matthew T. Oetjens, David H. Ledbetter and Christa Lese Martin
J. Pers. Med. 2021, 11(5), 365; https://doi.org/10.3390/jpm11050365 - 01 May 2021
Cited by 6 | Viewed by 2249
Abstract
Genomic variants that cause neurodevelopmental/psychiatric disorders (NPD) are relatively prevalent and highly penetrant. This study aimed to understand adults’ immediate responses to receiving NPD-related results to inform inclusion in population-based genomic screening programs. Nine recurrent, pathogenic copy number variants (CNVs) were identified from [...] Read more.
Genomic variants that cause neurodevelopmental/psychiatric disorders (NPD) are relatively prevalent and highly penetrant. This study aimed to understand adults’ immediate responses to receiving NPD-related results to inform inclusion in population-based genomic screening programs. Nine recurrent, pathogenic copy number variants (CNVs) were identified from research exome data, clinically confirmed, and disclosed to adult participants of the Geisinger MyCode Community Health Initiative DiscovEHR cohort by experienced genetic counselors. A subset of in-person genetic counseling sessions (n = 27) were audio-recorded, transcribed, and coded using a grounded theory approach. Participant reactions were overwhelmingly positive and indicated that an NPD genetic etiology was highly valuable and personally useful. Participants frequently reported learning disabilities or other NPD that were not documented in their electronic health records and noted difficulties obtaining support for NPD needs. Most intended to share their genetic result with family members and health care providers and were interested in how their result could improve their healthcare. This study indicates that results from population-based NPD genomic screening can provide personal value for adults with NPD, were viewed positively by participants, and could improve clinical outcomes by informing symptom monitoring for NPD and co-morbidities, promoting improved health behaviors, and enhancing psychotherapeutic approaches. Full article
(This article belongs to the Special Issue Precision Medicine in Clinical Practice)
Show Figures

Figure 1

Review

Jump to: Research

15 pages, 297 KiB  
Review
Polygenic Scores in the Direct-to-Consumer Setting: Challenges and Opportunities for a New Era in Consumer Genetic Testing
by Jin K. Park and Christine Y. Lu
J. Pers. Med. 2023, 13(4), 573; https://doi.org/10.3390/jpm13040573 - 23 Mar 2023
Cited by 3 | Viewed by 1742
Abstract
Direct-to-consumer (DTC) genetic tests have generated considerable scholarly attention and public intrigue. Although the current consumer genetic testing regime relies on the reporting of individual variants of interest to consumers, there has recently been interest in the possibility of integrating polygenic scores (PGS), [...] Read more.
Direct-to-consumer (DTC) genetic tests have generated considerable scholarly attention and public intrigue. Although the current consumer genetic testing regime relies on the reporting of individual variants of interest to consumers, there has recently been interest in the possibility of integrating polygenic scores (PGS), which aggregate genetic liability for disease across the entire genome. While PGS have thus far been extensively explored as clinical and public health tools, the use of PGS in consumer genetic testing has not yet received systematic attention, even though they are already in use for some consumer genetic tests. In this narrative review, we highlight the ethical, legal, and social implications of the use of PGS in DTC genetic tests and synthesize existing solutions to these concerns. We organize these concerns into three domains: (1) industry variation; (2) privacy and commercialization; and (3) patient safety and risk. While previously expressed concerns in these domains will remain relevant, the emergence of PGS-based DTC genetic tests raises challenges that will require novel approaches. Full article
(This article belongs to the Special Issue Precision Medicine in Clinical Practice)
18 pages, 2249 KiB  
Review
Patient-Reported Outcomes following Genetic Testing for Familial Hypercholesterolemia, Breast and Ovarian Cancer Syndrome, and Lynch Syndrome: A Systematic Review
by Rachele M. Hendricks-Sturrup, Lucson Joseph and Christine Y. Lu
J. Pers. Med. 2021, 11(9), 850; https://doi.org/10.3390/jpm11090850 - 27 Aug 2021
Cited by 2 | Viewed by 3064
Abstract
Background: Patient-reported outcomes (PROs) and PRO measures (PROMs) are real-world evidence that can help capture patient experiences and perspectives regarding a clinical intervention such as genetic testing. Objective: To identify and capture methods and qualitative PRO themes among studies reporting PROs following genetic [...] Read more.
Background: Patient-reported outcomes (PROs) and PRO measures (PROMs) are real-world evidence that can help capture patient experiences and perspectives regarding a clinical intervention such as genetic testing. Objective: To identify and capture methods and qualitative PRO themes among studies reporting PROs following genetic testing for FH, breast and ovarian cancer syndrome, and Lynch syndrome. Methods: A systematic review was conducted via PubMed/MEDLINE, EMBASE, and Yale University’s TRIP Medical Databases on articles published by April 2021. Results: We identified 24 studies published between 1996 and 2021 representing 4279 participants that reported PROs following genetic testing for FH, breast and ovarian cancer syndrome, and Lynch syndrome. Studies collected and reported PROs from validated PROM instruments (n = 12; 50%), validated surveys (n = 7; 26%), and interviews (n = 10; 42%). PRO themes ranged across all collection methods (e.g., psychological, knowledge, coping and satisfaction, concern about stigma/discrimination, etc.). Conclusions: Important gaps identified include (1) most studies (n = 18; 75%) reported PROs following genetic testing for breast and ovarian cancer, and (2) populations reporting PROs overall were largely of White/Caucasian/Northern European/Anglo-Saxon descent. We offer recommendations and describe real-world implications for the field moving forward. Full article
(This article belongs to the Special Issue Precision Medicine in Clinical Practice)
Show Figures

Figure 1

Back to TopTop