1
|
Nekhlyudov L, Levit LA, Ganz PA. Delivering High-Quality Cancer Care: Charting a New Course for a System in Crisis: One Decade Later. J Clin Oncol 2024:JCO2401243. [PMID: 39356979 DOI: 10.1200/jco-24-01243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/31/2024] [Accepted: 08/08/2024] [Indexed: 10/04/2024] Open
Abstract
In 2012, the National Academies of Sciences, Engineering, and Medicine convened a committee charged with addressing the quality of cancer care in the United States and providing recommendations to policymakers and the cancer care community on strategies to improve cancer care delivery from the time of diagnosis through end-of-life. The resulting committee report, titled Delivering High-Quality Cancer Care: Charting a New Course for a System in Crisis (2013), presented a conceptual framework that included six interconnected components of care with corresponding recommendations. Over the past decade, the delivery of high-quality of cancer care has become more challenging and increasingly demanding on the workforce. In this manuscript, we review the goals and recommendations made in 2013, describe progress to date, and offer insights into future dedicated efforts and/or new strategies needed to achieve high-quality cancer care.
Collapse
Affiliation(s)
- Larissa Nekhlyudov
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Laura A Levit
- American Society of Clinical Oncology, Alexandria, VA
| | - Patricia A Ganz
- UCLA Fielding School of Public Health, David Geffen School of Medicine at UCLA, and the Jonsson Comprehensive Cancer Center, Los Angeles, CA
| |
Collapse
|
2
|
Kuo ZM, Chen KF, Tseng YJ. MoCab: A framework for the deployment of machine learning models across health information systems. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 255:108336. [PMID: 39079482 DOI: 10.1016/j.cmpb.2024.108336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/13/2024] [Accepted: 07/17/2024] [Indexed: 09/01/2024]
Abstract
BACKGROUND AND OBJECTIVE Machine learning models are vital for enhancing healthcare services. However, integrating them into health information systems (HISs) introduces challenges beyond clinical decision making, such as interoperability and diverse electronic health records (EHR) formats. We proposed Model Cabinet Architecture (MoCab), a framework designed to leverage fast healthcare interoperability resources (FHIR) as the standard for data storage and retrieval when deploying machine learning models across various HISs, addressing the challenges highlighted by platforms such as EPOCH®, ePRISM®, KETOS, and others. METHODS The MoCab architecture is designed to streamline predictive modeling in healthcare through a structured framework incorporating several specialized parts. The Data Service Center manages patient data retrieval from FHIR servers. These data are then processed by the Knowledge Model Center, where they are formatted and fed into predictive models. The Model Retraining Center is crucial in continuously updating these models to maintain accuracy in dynamic clinical environments. The framework further incorporates Clinical Decision Support (CDS) Hooks for issuing clinical alerts. It uses Substitutable Medical Apps Reusable Technologies (SMART) on FHIR to develop applications for displaying alerts, prediction results, and patient records. RESULTS The MoCab framework was demonstrated using three types of predictive models: a scoring model (qCSI), a machine learning model (NSTI), and a deep learning model (SPC), applied to synthetic data that mimic a major EHR system. The implementations showed how MoCab integrates predictive models with health data for clinical decision support, utilizing CDS Hooks and SMART on FHIR for seamless HIS integration. The demonstration confirmed the practical utility of MoCab in supporting clinical decision making, validated by its application in various healthcare settings. CONCLUSIONS We demonstrate MoCab's potential in promoting the interoperability of machine learning models and enhancing its utility across various EHRs. Despite facing challenges like FHIR adoption, MoCab addresses key challenges in adapting machine learning models within healthcare settings, paving the way for further enhancements and broader adoption.
Collapse
Affiliation(s)
- Zhe-Ming Kuo
- Department of Information Management, National Central University, Taoyuan, Taiwan
| | - Kuan-Fu Chen
- College of Intelligent Computing, Chang Gung University, Taoyuan, Taiwan; Medical Statistics Research Center, Chang Gung University, Taoyuan, Taiwan; Department of Emergency Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Yi-Ju Tseng
- Department of Computer Science, National Yang Ming Chiao Tung University, Hsinchu, Taiwan; Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
3
|
Hashemi Karoii D, Bavandi S, Djamali M, Abroudi AS. Exploring the interaction between immune cells in the prostate cancer microenvironment combining weighted correlation gene network analysis and single-cell sequencing: An integrated bioinformatics analysis. Discov Oncol 2024; 15:513. [PMID: 39349877 PMCID: PMC11442730 DOI: 10.1007/s12672-024-01399-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND The rise of treatment resistance and variability across malignant profiles has made precision oncology an imperative in today's medical landscape. Prostate cancer is a prevalent form of cancer in males, characterized by significant diversity in both genomic and clinical characteristics. The tumor microenvironment consists of stroma, tumor cells, and various immune cells. The stromal components and tumor cells engage in mutual communication and facilitate the development of a low-oxygen and pro-cancer milieu by producing cytokines and activating pro-inflammatory signaling pathways. METHODS In order to discover new genes associated with tumor cells that interact and facilitate a hypoxic environment in prostate cancer, we conducted a cutting-edge bioinformatics investigation. This included analyzing high-throughput genomic datasets obtained from the cancer genome atlas (TCGA). RESULTS A combination of weighted gene co-expression network analysis and single-cell sequencing has identified nine dysregulated immune hub genes (AMACR, KCNN3, MME, EGFR, FLT1, GDF15, KDR, IGF1, and KRT7) that are believed to have significant involvement in the biological pathways involved with the advancement of prostate cancer enviriment. In the prostate cancer environment, we observed the overexpression of GDF15 and KRT7 genes, as well as the downregulation of other genes. Additionally, the cBioPortal platform was used to investigate the frequency of alterations in the genes and their effects on the survival of the patients. The Kaplan-Meier survival analysis indicated that the changes in the candidate genes were associated with a reduction in the overall survival of the patients. CONCLUSIONS In summary, the findings indicate that studying the genes and their genomic changes may be used to develop precise treatments for prostate cancer. This approach involves early detection and targeted therapy.
Collapse
Affiliation(s)
- Danial Hashemi Karoii
- Department of Cell and Molecular Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran.
| | - Sobhan Bavandi
- Department of Biology, Qaemshahr Branch, Islamic Azad University, Qaemshahr, Iran
| | - Melika Djamali
- Department of Biology, Faculty of Science, Tehran University, Tehran, Iran
| | - Ali Shakeri Abroudi
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| |
Collapse
|
4
|
Gazzarata R, Almeida J, Lindsköld L, Cangioli G, Gaeta E, Fico G, Chronaki CE. HL7 Fast Healthcare Interoperability Resources (HL7 FHIR) in digital healthcare ecosystems for chronic disease management: Scoping review. Int J Med Inform 2024; 189:105507. [PMID: 38870885 DOI: 10.1016/j.ijmedinf.2024.105507] [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: 01/07/2024] [Revised: 05/14/2024] [Accepted: 05/27/2024] [Indexed: 06/15/2024]
Abstract
BACKGROUND The prevalence of chronic diseases has shifted the burden of disease from incidental acute inpatient admissions to long-term coordinated care across healthcare institutions and the patient's home. Digital healthcare ecosystems emerge to target increasing healthcare costs and invest in standard Application Programming Interfaces (API), such as HL7 Fast Healthcare Interoperability Resources (HL7 FHIR) for trusted data flows. OBJECTIVES This scoping review assessed the role and impact of HL7 FHIR and associated Implementation Guides (IGs) in digital healthcare ecosystems focusing on chronic disease management. METHODS To study trends and developments relevant to HL7 FHIR, a scoping review of the scientific and gray English literature from 2017 to 2023 was used. RESULTS The selection of 93 of 524 scientific papers reviewed in English indicates that the popularity of HL7 FHIR as a robust technical interface standard for the health sector has been steadily rising since its inception in 2010, reaching a peak in 2021. Digital Health applications use HL7 FHIR in cancer (45 %), cardiovascular disease (CVD) (more than 15 %), and diabetes (almost 15 %). The scoping review revealed that references to HL7 FHIR IGs are limited to ∼ 20 % of articles reviewed. HL7 FHIR R4 was most frequently referenced when the HL7 FHIR version was mentioned. In HL7 FHIR IGs registries and the internet, we found 35 HL7 FHIR IGs addressing chronic disease management, i.e., cancer (40 %), chronic disease management (25 %), and diabetes (20 %). HL7 FHIR IGs frequently complement the information in the article. CONCLUSIONS HL7 FHIR matures with each revision of the standard as HL7 FHIR IGs are developed with validated data sets, common shared HL7 FHIR resources, and supporting tools. Referencing HL7 FHIR IGs cataloged in official registries and in scientific publications is recommended to advance data quality and facilitate mutual learning in growing digital healthcare ecosystems that nurture interoperability in digital health innovation.
Collapse
Affiliation(s)
- Roberta Gazzarata
- HL7 Europe Foundation, 38-40 Square de Meeus, Brussels, 1000, Belgium; Healthropy Srl, Corso Vittorio Veneto 14B, Savona, 17100, Italy.
| | - Joao Almeida
- HL7 Europe Foundation, 38-40 Square de Meeus, Brussels, 1000, Belgium; MEDCIDS - Faculty of Medicine of University of Porto, Porto, Portugal; PDH - Pharma Data Hub, Porto, Portugal.
| | - Lars Lindsköld
- European Federation for Medical Informatics, Ch de Maillefer 37, CH-1052 Le Mont-sur-Lausanne, Switzerland; SciLifeLab Datacenter, University of Uppsala, S-752 37 Uppsala, Sweden.
| | - Giorgio Cangioli
- HL7 Europe Foundation, 38-40 Square de Meeus, Brussels, 1000, Belgium.
| | - Eugenio Gaeta
- Life Supporting Technologies, Universidad Politécnica de Madrid, Avenida Complutense 30, 28040 Madrid, Spain.
| | - Giuseppe Fico
- Life Supporting Technologies, Universidad Politécnica de Madrid, Avenida Complutense 30, 28040 Madrid, Spain.
| | - Catherine E Chronaki
- HL7 Europe Foundation, 38-40 Square de Meeus, Brussels, 1000, Belgium; European Federation for Medical Informatics, Ch de Maillefer 37, CH-1052 Le Mont-sur-Lausanne, Switzerland.
| |
Collapse
|
5
|
Davoodi Nik B, Hashemi Karoii D, Favaedi R, Ramazanali F, Jahangiri M, Movaghar B, Shahhoseini M. Differential expression of ion channel coding genes in the endometrium of women experiencing recurrent implantation failures. Sci Rep 2024; 14:19822. [PMID: 39192025 PMCID: PMC11349755 DOI: 10.1038/s41598-024-70778-9] [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: 04/14/2024] [Accepted: 08/21/2024] [Indexed: 08/29/2024] Open
Abstract
Our study probed the differences in ion channel gene expression in the endometrium of women with Recurrent Implantation Failure (RIF) compared to fertile women. We analyzed the relative expression of genes coding for T-type Ca2+, ENaC, CFTR, and KCNQ1 channels in endometrial samples from 20 RIF-affected and 10 control women, aged 22-35, via microarray analysis and quantitative real-time PCR. Additionally, we examined DNA methylation in the regulatory region of KCNQ1 using ChIP real-time PCR. The bioinformatics component of our research included Gene Ontology analysis, protein-protein interaction networks, and signaling pathway mapping to identify key biological processes and pathways implicated in RIF. This led to the discovery of significant alterations in the expression of ion channel genes in RIF women's endometrium, most notably an overexpression of CFTR and reduced expression of SCNN1A, SCNN1B, SCNN1G, CACNA1H, and KCNQ1. A higher DNA methylation level of KCNQ1's regulatory region was also observed in RIF patients. Gene-set enrichment analysis highlighted a significant presence of genes involved with ion transport and membrane potential regulation, particularly in sodium and calcium channel complexes, which are vital for cation movement across cell membranes. Genes were also enriched in broader ion channel and transmembrane transporter complexes, underscoring their potential extensive role in cellular ion homeostasis and signaling. These findings suggest a potential involvement of ion channels in the pathology of implantation failure, offering new insights into the mechanisms behind RIF and possible therapeutic targets.
Collapse
Affiliation(s)
- Bahar Davoodi Nik
- Department of Genetics, Faculty of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Danial Hashemi Karoii
- Department of Cell and Molecular Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Raha Favaedi
- Department of Genetics, Reproductive Biomedicine Research Centre, Royan Institute for Reproductive Biomedicine, ACECR, No. 12, Hafez St., Banihashem Sq, Resalat Ave., P.O. Box: 19395-4644, Tehran, Iran
| | - Fariba Ramazanali
- Department of Endocrinology and Female Infertility, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Maryam Jahangiri
- Department of Embryology, Reproductive Biomedicine Research Centre, Royan Institute for Reproductive Biomedicine, ACECR, No. 12, Hafez St., Banihashem Sq, Resalat Ave., P.O. Box: 19395-4644, Tehran, Iran
| | - Bahar Movaghar
- Department of Embryology, Reproductive Biomedicine Research Centre, Royan Institute for Reproductive Biomedicine, ACECR, No. 12, Hafez St., Banihashem Sq, Resalat Ave., P.O. Box: 19395-4644, Tehran, Iran.
| | - Maryam Shahhoseini
- Department of Genetics, Faculty of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran.
- Department of Cell and Molecular Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran.
- Department of Genetics, Reproductive Biomedicine Research Centre, Royan Institute for Reproductive Biomedicine, ACECR, No. 12, Hafez St., Banihashem Sq, Resalat Ave., P.O. Box: 19395-4644, Tehran, Iran.
| |
Collapse
|
6
|
Chakrabarty N, Mahajan A. Imaging Analytics using Artificial Intelligence in Oncology: A Comprehensive Review. Clin Oncol (R Coll Radiol) 2024; 36:498-513. [PMID: 37806795 DOI: 10.1016/j.clon.2023.09.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 08/09/2023] [Accepted: 09/21/2023] [Indexed: 10/10/2023]
Abstract
The present era has seen a surge in artificial intelligence-related research in oncology, mainly using deep learning, because of powerful computer hardware, improved algorithms and the availability of large amounts of data from open-source domains and the use of transfer learning. Here we discuss the multifaceted role of deep learning in cancer care, ranging from risk stratification, the screening and diagnosis of cancer, to the prediction of genomic mutations, treatment response and survival outcome prediction, through the use of convolutional neural networks. Another role of artificial intelligence is in the generation of automated radiology reports, which is a boon in high-volume centres to minimise report turnaround time. Although a validated and deployable deep-learning model for clinical use is still in its infancy, there is ongoing research to overcome the barriers for its universal implementation and we also delve into this aspect. We also briefly describe the role of radiomics in oncoimaging. Artificial intelligence can provide answers pertaining to cancer management at baseline imaging, saving cost and time. Imaging biobanks, which are repositories of anonymised images, are also briefly described. We also discuss the commercialisation and ethical issues pertaining to artificial intelligence. The latest generation generalist artificial intelligence model is also briefly described at the end of the article. We believe this article will not only enrich knowledge, but also promote research acumen in the minds of readers to take oncoimaging to another level using artificial intelligence and also work towards clinical translation of such research.
Collapse
Affiliation(s)
- N Chakrabarty
- Department of Radiodiagnosis, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Parel, Mumbai, Maharashtra, India.
| | - A Mahajan
- The Clatterbridge Cancer Centre NHS Foundation Trust, Liverpool, UK.
| |
Collapse
|
7
|
Osterman TJ, Ye J. The importance of studying the implementation of cancer data standards. Cancer 2024. [PMID: 38872412 DOI: 10.1002/cncr.35441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Data standards promise better quality measurement, discovery, population health management, and interoperability. For oncology practices to make informed decisions about implementation, it is paramount that the cancer informatics community continue to study and publish diverse methods to structure data.
Collapse
Affiliation(s)
| | - Jiarong Ye
- Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
8
|
Riba M, Sala C, Culhane AC, Flobak Å, Patocs A, Boye K, Plevova K, Pospíšilová Š, Gandolfi G, Morelli MJ, Bucci G, Edsjö A, Lassen U, Al-Shahrour F, Lopez-Bigas N, Hovland R, Cuppen E, Valencia A, Poirel HA, Rosenquist R, Scollen S, Arenas Marquez J, Belien J, De Nicolo A, De Maria R, Torrents D, Tonon G. The 1+Million Genomes Minimal Dataset for Cancer. Nat Genet 2024; 56:733-736. [PMID: 38702538 DOI: 10.1038/s41588-024-01721-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2024]
Affiliation(s)
- Michela Riba
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Cinzia Sala
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Aedin C Culhane
- Limerick Digital Cancer Research Centre, Health Research Institute, School of Medicine, University of Limerick, Limerick, Ireland
| | - Åsmund Flobak
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- The Cancer Clinic, St. Olav's University Hospital, Trondheim, Norway
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Attila Patocs
- Department of Molecular Genetics and the National Tumour Biology Laboratory, National Institute of Oncology, Budapest, Hungary
- Department of Oncology Biobank, National Institute of Oncology, Budapest, Hungary
- Hereditary Tumours Research Group, Hungarian Academy of Sciences, Semmelweis University, Budapest, Hungary
| | - Kjetil Boye
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Karla Plevova
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
- Department of Internal Medicine - Hematology and Oncology, Faculty of Medicine, Masaryk University and University Hospital, Brno, Czech Republic
- Department of Medical Genetics and Genomics, University Hospital and Faculty of Medicine, Brno, Czech Republic
| | - Šárka Pospíšilová
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
- Department of Internal Medicine - Hematology and Oncology, Faculty of Medicine, Masaryk University and University Hospital, Brno, Czech Republic
- Department of Medical Genetics and Genomics, University Hospital and Faculty of Medicine, Brno, Czech Republic
| | - Giorgia Gandolfi
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marco J Morelli
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Gabriele Bucci
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Anders Edsjö
- Department of Clinical Genetics, Pathology and Molecular Diagnostics, Office for Medical Services, Region Skåne, Lund, Sweden
- Division of Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Ulrik Lassen
- Department of Oncology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Fátima Al-Shahrour
- Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Nuria Lopez-Bigas
- Institución Catalana de Investigación y Estudios Avanzados (ICREA), Barcelona, Spain
| | - Randi Hovland
- Section of Cancer Genomics Haukeland University Hospital, Bergen, Norway
| | - Edwin Cuppen
- Center for Molecular Medicine, Oncode Institute, University Medical Center Utrecht, Utrecht, the Netherlands
- Hartwig Medical Foundation, Amsterdam, the Netherlands
| | - Alfonso Valencia
- Institución Catalana de Investigación y Estudios Avanzados (ICREA), Barcelona, Spain
| | | | - Richard Rosenquist
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| | - Serena Scollen
- ELIXIR Hub, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | | | - Jeroen Belien
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Arcangela De Nicolo
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ruggero De Maria
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario 'A. Gemelli' - Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - David Torrents
- Institución Catalana de Investigación y Estudios Avanzados (ICREA), Barcelona, Spain
| | - Giovanni Tonon
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Functional Genomics of Cancer Unit, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
9
|
Kim DY, Shin DY, Oh S, Kim I, Kim EJ. Gene Expression and DNA Methylation Profiling Suggest Potential Biomarkers for Azacitidine Resistance in Myelodysplastic Syndrome. Int J Mol Sci 2024; 25:4723. [PMID: 38731939 PMCID: PMC11083267 DOI: 10.3390/ijms25094723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 04/22/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
Myelodysplastic syndrome/neoplasm (MDS) comprises a group of heterogeneous hematopoietic disorders that present with genetic mutations and/or cytogenetic changes and, in the advanced stage, exhibit wide-ranging gene hypermethylation. Patients with higher-risk MDS are typically treated with repeated cycles of hypomethylating agents, such as azacitidine. However, some patients fail to respond to this therapy, and fewer than 50% show hematologic improvement. In this context, we focused on the potential use of epigenetic data in clinical management to aid in diagnostic and therapeutic decision-making. First, we used the F-36P MDS cell line to establish an azacitidine-resistant F-36P cell line. We performed expression profiling of azacitidine-resistant and parental F-36P cells and used biological and bioinformatics approaches to analyze candidate azacitidine-resistance-related genes and pathways. Eighty candidate genes were identified and found to encode proteins previously linked to cancer, chronic myeloid leukemia, and transcriptional misregulation in cancer. Interestingly, 24 of the candidate genes had promoter methylation patterns that were inversely correlated with azacitidine resistance, suggesting that DNA methylation status may contribute to azacitidine resistance. In particular, the DNA methylation status and/or mRNA expression levels of the four genes (AMER1, HSPA2, NCX1, and TNFRSF10C) may contribute to the clinical effects of azacitidine in MDS. Our study provides information on azacitidine resistance diagnostic genes in MDS patients, which can be of great help in monitoring the effectiveness of treatment in progressing azacitidine treatment for newly diagnosed MDS patients.
Collapse
Affiliation(s)
- Da Yeon Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea;
- Department of Radiological and Medico-Oncological Sciences, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Dong-Yeop Shin
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (D.-Y.S.); (S.O.)
- Center for Medical Innovation, Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Republic of Korea
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Somi Oh
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (D.-Y.S.); (S.O.)
| | - Inho Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (D.-Y.S.); (S.O.)
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Eun Ju Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea;
- Department of Radiological and Medico-Oncological Sciences, University of Science and Technology, Daejeon 34113, Republic of Korea
- Institute for Molecular Bioscience, The University of Queensland, Carmody Rd., St Lucia, Brisbane, QLD 4072, Australia
- Genomics and Machine Learning Lab, QIMR Berghofer Medical Research Institute, Herston Rd., Herston, Brisbane, QLD 4006, Australia
| |
Collapse
|
10
|
Chen Y, Fu D, Wu X, Zhang Y, Chen Y, Zhou Y, Lu M, Liu Q, Huang J. Biomimetic biphasic microsphere preparation based on the thermodynamic incompatibility of glycosaminoglycan with gelatin methacrylate for hair regeneration. Int J Biol Macromol 2024; 261:129934. [PMID: 38311145 DOI: 10.1016/j.ijbiomac.2024.129934] [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: 12/13/2023] [Revised: 01/28/2024] [Accepted: 01/31/2024] [Indexed: 02/08/2024]
Abstract
Hair follicle (HF) tissue engineering is promising for hair loss treatment especially for androgenetic alopecia. Physiologically, the initiation of HF morphogenesis relies on the interactions between hair germ mesenchymal and epithelial layers. To simulate this intricate process, in this study, a co-flowing microfluidic-assisted technology was developed to produce dual aqueous microdroplets capturing growth factors and double-layer cells for subsequent use in hair regeneration. Microspheres, called G/HAD, were generated using glycosaminoglycan-based photo-crosslinkable biological macromolecule (HAD) shells and gelatin methacrylate (GelMA) cores to enclose mesenchymal cells (MSCs) and mouse epidermal cells (EPCs). The findings indicated that the glycosaminoglycan-based HAD shells display thermodynamic incompatibility with GelMA cores, resulting in the aqueous phase separation of G/HAD cell spheres. These G/HAD microspheres exhibited favorable characteristics, including sustained growth factor release and wet adhesion properties. After transplantation into the dorsal skin of BALB/c nude mice, G/HAD cell microspheres efficiently induced the regeneration of HFs. This approach enables the mass production of approximately 250 dual-layer microspheres per minute. Thus, this dual-layer microsphere fabrication method holds great potential in improving current hair regeneration techniques and can also be combined with other tissue engineering techniques for various regenerative purposes.
Collapse
Affiliation(s)
- Yangpeng Chen
- Department of Plastic and Aesthetic Surgery, Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Danlan Fu
- Department of Plastic and Aesthetic Surgery, Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Xiaoqi Wu
- Department of Urology and Andrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China
| | - Yufan Zhang
- Department of Plastic and Aesthetic Surgery, Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yuxin Chen
- Department of Plastic and Aesthetic Surgery, Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yi Zhou
- Department of Plastic and Aesthetic Surgery, Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Mujun Lu
- Department of Urology and Andrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China.
| | - Qifa Liu
- Department of Plastic and Aesthetic Surgery, Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Junfei Huang
- Department of Plastic and Aesthetic Surgery, Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China.
| |
Collapse
|
11
|
O'Sullivan-Steben K, Galarneau L, Judd S, Laizner AM, Williams T, Kildea J. Design and implementation of a prototype radiotherapy menu in a patient portal. J Appl Clin Med Phys 2024; 25:e14201. [PMID: 37942985 DOI: 10.1002/acm2.14201] [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: 10/03/2023] [Revised: 10/10/2023] [Accepted: 10/31/2023] [Indexed: 11/10/2023] Open
Abstract
PURPOSE Radiotherapy patients often face undue anxiety due to misconceptions about radiation and their inability to visualize their upcoming treatments. Access to their personal treatment plans is one way in which pre-treatment anxiety may be reduced. But radiotherapy data are quite complex, requiring specialized software for display and necessitating personalized explanations for patients to understand them. Therefore, our goal was to design and implement a novel radiotherapy menu in a patient portal to improve patient access to and understanding of their radiotherapy treatment plans. METHODS A prototype radiotherapy menu was developed in our institution's patient portal following a participatory stakeholder co-design methodology. Customizable page templates were designed to render key radiotherapy data in the portal's patient-facing mobile phone app. DICOM-RT data were used to provide patients with relevant treatment parameters and generate pre-treatment 3D visualizations of planned treatment beams, while the mCODE data standard was used to provide post-treatment summaries of the delivered treatments. A focus group was conducted to gather initial patient feedback on the menu. RESULTS Pre-treatment: the radiotherapy menu provides patients with a personalized treatment plan overview, including a personalized explanation of their treatment, along with an interactive 3D rendering of their body, and treatment beams for visualization. Post-treatment: a summary of the delivered radiotherapy is provided, allowing patients to retain a concise personal record of their treatment that can easily be shared with future healthcare providers. Focus group feedback was overwhelmingly positive. Patients highlighted how the intuitive presentation of their complex radiotherapy data would better prepare them for their radiation treatments. CONCLUSIONS We successfully designed and implemented a prototype radiotherapy menu in our institution's patient portal that improves patient access to and understanding of their radiotherapy data. We used the mCODE data standard to generate post-treatment summaries in a way that is easily shareable and interoperable.
Collapse
Affiliation(s)
| | - Luc Galarneau
- Medical Physics Unit, McGill University, Montreal, Quebec, Canada
| | - Susie Judd
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Andrea M Laizner
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- Ingram School of Nursing, McGill University, Montreal, Quebec, Canada
| | - Tristan Williams
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - John Kildea
- Medical Physics Unit, McGill University, Montreal, Quebec, Canada
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- Gerald Bronfman Department of Oncology, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
12
|
Aryal B, Bizhanova Z, Joseph EA, Yin Y, Wagner PL, Dalton E, LaFramboise WA, Bartlett DL, Allen CJ. Navigating Precision Oncology: Insights from an Integrated Clinical Data and Biobank Repository Initiative across a Network Cancer Program. Cancers (Basel) 2024; 16:760. [PMID: 38398150 PMCID: PMC10886699 DOI: 10.3390/cancers16040760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/03/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Advancing cancer treatment relies on the rapid translation of new scientific discoveries to patient care. To facilitate this, an oncology biobank and data repository program, also referred to as the "Moonshot" program, was launched in 2021 within the Integrated Network Cancer Program of the Allegheny Health Network. A clinical data program (CDP) and biospecimen repository were established, and patient data and blood and tissue samples have been collected prospectively. To date, the study has accrued 2920 patients, predominantly female (61%) and Caucasian (90%), with a mean age of 64 ± 13 years. The most common cancer sites were the endometrium/uterus (12%), lung/bronchus (12%), breast (11%), and colon/rectum (11%). Of patients diagnosed with cancer, 34% were diagnosed at stage I, 25% at stage II, 26% at stage III, and 15% at stage IV. The CDP is designed to support our initiative in advancing personalized cancer research by providing a comprehensive array of patient data, encompassing demographic characteristics, diagnostic details, and treatment responses. The "Moonshot" initiative aims to predict therapy responses and clinical outcomes through cancer-related biomarkers. The CDP facilitates this initiative by fostering data sharing, enabling comparative analyses, and informing the development of novel diagnostic and therapeutic methods.
Collapse
Affiliation(s)
- Bibek Aryal
- Allegheny Singer Research Institute, Allegheny Health Network, Pittsburgh, PA 15212, USA; (B.A.); (Z.B.); (E.A.J.); (Y.Y.)
| | - Zhadyra Bizhanova
- Allegheny Singer Research Institute, Allegheny Health Network, Pittsburgh, PA 15212, USA; (B.A.); (Z.B.); (E.A.J.); (Y.Y.)
| | - Edward A. Joseph
- Allegheny Singer Research Institute, Allegheny Health Network, Pittsburgh, PA 15212, USA; (B.A.); (Z.B.); (E.A.J.); (Y.Y.)
| | - Yue Yin
- Allegheny Singer Research Institute, Allegheny Health Network, Pittsburgh, PA 15212, USA; (B.A.); (Z.B.); (E.A.J.); (Y.Y.)
| | - Patrick L. Wagner
- Division of Surgical Oncology, Institute of Surgery, Allegheny Health Network, Pittsburgh, PA 15212, USA;
| | | | | | - David L. Bartlett
- Allegheny Health Network Cancer Institute, Pittsburgh, PA 15212, USA;
| | - Casey J. Allen
- Division of Surgical Oncology, Institute of Surgery, Allegheny Health Network, Pittsburgh, PA 15212, USA;
| |
Collapse
|
13
|
Kočo L, Siebers CCN, Schlooz M, Meeuwis C, Oldenburg HSA, Prokop M, Mann RM. The Facilitators and Barriers of the Implementation of a Clinical Decision Support System for Breast Cancer Multidisciplinary Team Meetings-An Interview Study. Cancers (Basel) 2024; 16:401. [PMID: 38254891 PMCID: PMC10813995 DOI: 10.3390/cancers16020401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/07/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND AI-driven clinical decision support systems (CDSSs) hold promise for multidisciplinary team meetings (MDTMs). This study aimed to uncover the hurdles and aids in implementing CDSSs during breast cancer MDTMs. METHODS Twenty-four core team members from three hospitals engaged in semi-structured interviews, revealing a collective interest in experiencing CDSS workflows in clinical practice. All interviews were audio recorded, transcribed verbatim and analyzed anonymously. A standardized approach, 'the framework method', was used to create an analytical framework for data analysis, which was performed by two independent researchers. RESULTS Positive aspects included improved data visualization, time-saving features, automated trial matching, and enhanced documentation transparency. However, challenges emerged, primarily concerning data connectivity, guideline updates, the accuracy of AI-driven suggestions, and the risk of losing human involvement in decision making. Despite the complexities involved in CDSS development and integration, clinicians demonstrated enthusiasm to explore its potential benefits. CONCLUSIONS Acknowledging the multifaceted nature of this challenge, insights into the barriers and facilitators identified in this study offer a potential roadmap for smoother future implementations. Understanding these factors could pave the way for more effective utilization of CDSSs in breast cancer MDTMs, enhancing patient care through informed decision making.
Collapse
Affiliation(s)
- Lejla Kočo
- Department of Imaging, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Carmen C. N. Siebers
- Department of Imaging, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Margrethe Schlooz
- Department of Surgery, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Carla Meeuwis
- Department of Radiology, Rijnstate, Wagnerlaan 55, 6815 AD Arnhem, The Netherlands;
| | - Hester S. A. Oldenburg
- Department of Surgery, The Netherlands Cancer Institute (Antoni van Leeuwenhoek), Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Mathias Prokop
- Department of Imaging, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Ritse M. Mann
- Department of Imaging, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
- Department of Surgery, The Netherlands Cancer Institute (Antoni van Leeuwenhoek), Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| |
Collapse
|
14
|
Chu J, Liu W, Hu X, Zhang H, Jiang J. P2RY13 is a prognostic biomarker and associated with immune infiltrates in renal clear cell carcinoma: A comprehensive bioinformatic study. Health Sci Rep 2023; 6:e1646. [PMID: 38045624 PMCID: PMC10691167 DOI: 10.1002/hsr2.1646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 09/03/2023] [Accepted: 10/10/2023] [Indexed: 12/05/2023] Open
Abstract
Background and Aims Clear cell renal cell carcinoma (ccRCC) is a common and aggressive form of cancer with a high incidence globally. This study aimed to investigate the role of P2RY13 in the progression of ccRCC and elucidate its mechanism of action. Methods Gene Expression Omnibus and The Cancer Genome Atlas databases were used to extract gene expression profiles of ccRCC. These profiles were annotated and visualized by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) functional enrichment analyses, as well as Gene Set Enrichment Analysis (GSEA). The STRING database was used to establish a protein-protein interaction network and to analyze the functional similarity. The GEPIA2 database was used to predict survival associated with hub genes. Meanwhile, the TIMER2.0 database was used to assess immune cell infiltration and its link with the hub genes. Immunohistochemistry (IHC) was used to determine the difference between ccRCC and adjacent normal tissue. Results We identified 272 differentially expressed genes (DEGs). GO and KEGG analyses suggested that DEGs were primarily involved in lymphocyte activation, inflammatory response, immunological effector mechanism pathways. By cytohubba, the 20 highest-scoring hub genes were screened to identify critical genes in the protein-protein interaction network linked with ccRCC. Resting dendritic cells, CD8 T cells, and activated mast cells all showed a significant positive correlation with these hub genes. Moreover, a higher immune score was associated with increased prognostic risk scores, which in turn correlated with a poorer prognosis. IHC revealed that P2RY13 was expressed at higher levels in ccRCC compared to para-cancer tissues. Conclusion Identifying the DEGs will aid in the understanding of the causes and molecular mechanisms involved in ccRCC. P2RY13 may play a pivotal role in the progression and prognosis of ccRCC, potentially driving carcinogenesis though immune system mechanisms.
Collapse
Affiliation(s)
- Jie Chu
- Department of OncologyThe First People's Hospital of ZiyangZiyangChina
| | - Wei Liu
- Department of General Family MedicineThe First People's Hospital of NeiJiangNeiJiangChina
| | - Xinyue Hu
- Department of Clinical Laboratory, Kunming First People's HospitalKunming Medical UniversityKunmingChina
| | - Huiling Zhang
- Department of OncologyThe First People's Hospital of ZiyangZiyangChina
| | - Jiudong Jiang
- Department of SurgeryThe First People's Hospital of ZiYangZiyangChina
| |
Collapse
|
15
|
Franzoi MA, Gillanders E, Vaz-Luis I. Unlocking digitally enabled research in oncology: the time is now. ESMO Open 2023; 8:101633. [PMID: 37660408 PMCID: PMC10482746 DOI: 10.1016/j.esmoop.2023.101633] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 08/07/2023] [Indexed: 09/05/2023] Open
Affiliation(s)
- M A Franzoi
- Cancer Survivorship Group, Inserm Unit 981, Gustave Roussy, Villejuif
| | - E Gillanders
- Cancer Survivorship Group, Inserm Unit 981, Gustave Roussy, Villejuif
| | - I Vaz-Luis
- Cancer Survivorship Group, Inserm Unit 981, Gustave Roussy, Villejuif; Department for the Organization of Patient Pathways, DIOPP, Gustave Roussy, Villejuif, France.
| |
Collapse
|
16
|
Walesa MB, Denny A, Patel A, Mulcahy M, Kircher S, George C, Tsarwhas D, Ross A, Platanias LC, Poylin V, Yang AD, Barnard C, Bilimoria KY, Merkow RP. Enhancement and Implementation of a Health Information Technology Module to Improve the Discrete Capture of Cancer Staging in a Diverse Regional Health System. JCO Oncol Pract 2023; 19:e957-e966. [PMID: 37527464 DOI: 10.1200/op.23.00104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/10/2023] [Accepted: 05/30/2023] [Indexed: 08/03/2023] Open
Abstract
PURPOSE Cancer staging is the foundation for all cancer management decisions. For real-time use, stage must be embedded in the electronic health record as a discrete data element. The objectives of this quality improvement (QI) initiative were to (1) identify barriers to utilization of an existing discrete cancer staging module, (2) identify health information technology (HIT) solutions to support discrete capture of cancer staging data, and (3) increase capture across the oncology enterprise in our diverse health system. METHODS Six sigma QI methodologies were used to define barriers and solutions to improve discrete cancer staging. Design thinking principles informed solution development to test prototypes. Two multidisciplinary teams of disease-specific clinicians within GI and genitourinary conducted phased testing pilots to determine health system solutions. Solutions were expanded to all oncology specialties across our health system. RESULTS Baseline average discrete staging capture across our health system was 31%. Poor workflow efficiency, limited accountability, and technical design gaps were key barriers to timely, complete staging. Implementation of more than 25 design enhancements to a HIT solution and passive user alerts led to a postimplementation capture rate of 58% across 55 outpatient clinics involving more than 400 clinicians. CONCLUSION We identified key barriers to discrete data capture and designed solutions through iterative use of QI methodologies and disease-specific pilots. After implementation, discrete capture of cancer staging nearly doubled across our diverse health system. This approach is scalable and transferable to other initiatives to develop and implement clinically relevant HIT solutions across a diverse health system.
Collapse
Affiliation(s)
| | | | | | - Mary Mulcahy
- Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL
| | - Sheetal Kircher
- Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL
| | | | - Dean Tsarwhas
- Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL
| | - Ashley Ross
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Leonidas C Platanias
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Vitaly Poylin
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Anthony D Yang
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | | | - Karl Y Bilimoria
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN
| | - Ryan P Merkow
- Division of Surgical Oncology, University of Chicago, Chicago, IL
| |
Collapse
|
17
|
Zhang S, Li X, Zheng Y, Liu J, Hu H, Zhang S, Kuang W. Single cell and bulk transcriptome analysis identified oxidative stress response-related features of Hepatocellular Carcinoma. Front Cell Dev Biol 2023; 11:1191074. [PMID: 37842089 PMCID: PMC10568628 DOI: 10.3389/fcell.2023.1191074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 09/18/2023] [Indexed: 10/17/2023] Open
Abstract
Background: Hepatocellular Carcinoma (HCC) is a common lethal digestive system tumor. The oxidative stress mechanism is crucial in the HCC genesis and progression. Methods: Our study analyzed single-cell and bulk sequencing data to compare the microenvironment of non-tumor liver tissues and HCC tissues. Through these analyses, we aimed to investigate the effect of oxidative stress on cells in the HCC microenvironment and identify critical oxidative stress response-related genes that impact the survival of HCC patients. Results: Our results showed increased oxidative stress in HCC tissue compared to non-tumor tissue. Immune cells in the HCC microenvironment exhibited higher oxidative detoxification capacity, and oxidative stress-induced cell death of dendritic cells was attenuated. HCC cells demonstrated enhanced communication with immune cells through the MIF pathway in a highly oxidative hepatoma microenvironment. Meanwhile, using machine learning and Cox regression screening, we identified PRDX1 as a predictor of early occurrence and prognosis in patients with HCC. The expression level of PRDX1 in HCC was related to dysregulated ribosome biogenesis and positively correlated with the expression of immunological checkpoints (PDCD1LG2, CTLA4, TIGIT, LAIR1). High PRDX1 expression in HCC patients correlated with better sensitivity to immunotherapy agents such as sorafenib, IGF-1R inhibitor, and JAK inhibitor. Conclusion: In conclusion, our study unveiled variations in oxidative stress levels between non-tumor liver and HCC tissues. And we identified oxidative stress gene markers associated with hepatocarcinogenesis development, offering novel insights into the oxidative stress response mechanism in HCC.
Collapse
Affiliation(s)
- Shuqiao Zhang
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xinyu Li
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yilu Zheng
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jiahui Liu
- Department of Traditional Chinese Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hao Hu
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Shijun Zhang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Weihong Kuang
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Dongguan Key Laboratory of Chronic Inflammatory Diseases, School of Pharmacy, The First Dongguan Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong, China
| |
Collapse
|
18
|
Dreyer NA, Mack CD. Tactical Considerations for Designing Real-World Studies: Fit-for-Purpose Designs That Bridge Research and Practice. Pragmat Obs Res 2023; 14:101-110. [PMID: 37786592 PMCID: PMC10541678 DOI: 10.2147/por.s396024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 09/19/2023] [Indexed: 10/04/2023] Open
Abstract
Real-world evidence (RWE) is being used to provide information on diverse groups of patients who may be highly impacted by disease but are not typically studied in traditional randomized clinical trials (RCT) and to obtain insights from everyday care settings and real-world adherence to inform clinical practice. RWE is derived from so-called real-world data (RWD), ie, information generated by clinicians in the course of everyday patient care, and is sometimes coupled with systematic input from patients in the form of patient-reported outcomes or from wearable biosensors. Studies using RWD are conducted to evaluate how well medical interventions, services, and diagnostics perform under conditions of real-world use, and may include long-term follow-up. Here, we describe the main types of studies used to generate RWE and offer pointers for clinicians interested in study design and execution. Our tactical guidance addresses (1) opportunistic study designs, (2) considerations about representativeness of study participants, (3) expectations for transparency about data provenance, handling and quality assessments, and (4) considerations for strengthening studies using record linkage and/or randomization in pragmatic clinical trials. We also discuss likely sources of bias and suggest mitigation strategies. We see a future where clinical records - patient-generated data and other RWD - are brought together and harnessed by robust study design with efficient data capture and strong data curation. Traditional RCT will remain the mainstay of drug development, but RWE will play a growing role in clinical, regulatory, and payer decision-making. The most meaningful RWE will come from collaboration with astute clinicians with deep practice experience and questioning minds working closely with patients and researchers experienced in the development of RWE.
Collapse
|
19
|
Jamir E, Sarma H, Priyadarsinee L, Kiewhuo K, Nagamani S, Sastry GN. Polypharmacology guided drug repositioning approach for SARS-CoV2. PLoS One 2023; 18:e0289890. [PMID: 37556478 PMCID: PMC10411734 DOI: 10.1371/journal.pone.0289890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 07/27/2023] [Indexed: 08/11/2023] Open
Abstract
Drug repurposing has emerged as an important strategy and it has a great potential in identifying therapeutic applications for COVID-19. An extensive virtual screening of 4193 FDA approved drugs has been carried out against 24 proteins of SARS-CoV2 (NSP1-10 and NSP12-16, envelope, membrane, nucleoprotein, spike, ORF3a, ORF6, ORF7a, ORF8, and ORF9b). The drugs were classified into top 10 and bottom 10 drugs based on the docking scores followed by the distribution of their therapeutic indications. As a result, the top 10 drugs were found to have therapeutic indications for cancer, pain, neurological disorders, and viral and bacterial diseases. As drug resistance is one of the major challenges in antiviral drug discovery, polypharmacology and network pharmacology approaches were employed in the study to identify drugs interacting with multiple targets and drugs such as dihydroergotamine, ergotamine, bisdequalinium chloride, midostaurin, temoporfin, tirilazad, and venetoclax were identified among the multi-targeting drugs. Further, a pathway analysis of the genes related to the multi-targeting drugs was carried which provides insight into the mechanism of drugs and identifying targetable genes and biological pathways involved in SARS-CoV2.
Collapse
Affiliation(s)
- Esther Jamir
- Advanced Computation and Data Sciences Division, CSIR–North East Institute of Science and Technology, Jorhat, Assam, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Himakshi Sarma
- Advanced Computation and Data Sciences Division, CSIR–North East Institute of Science and Technology, Jorhat, Assam, India
| | - Lipsa Priyadarsinee
- Advanced Computation and Data Sciences Division, CSIR–North East Institute of Science and Technology, Jorhat, Assam, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Kikrusenuo Kiewhuo
- Advanced Computation and Data Sciences Division, CSIR–North East Institute of Science and Technology, Jorhat, Assam, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Selvaraman Nagamani
- Advanced Computation and Data Sciences Division, CSIR–North East Institute of Science and Technology, Jorhat, Assam, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - G. Narahari Sastry
- Advanced Computation and Data Sciences Division, CSIR–North East Institute of Science and Technology, Jorhat, Assam, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
20
|
Wood WA, Anderson KC, Kumar SK, Semmel EA, Hewitt K, Plovnick RM, Pappas G. A pandemic preparedness network for individuals living with compromised immune systems. Blood Adv 2023; 7:3925-3927. [PMID: 37023227 PMCID: PMC10405186 DOI: 10.1182/bloodadvances.2023010035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/22/2023] [Accepted: 04/05/2023] [Indexed: 04/08/2023] Open
Affiliation(s)
- William A. Wood
- Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | | | | | - Emily A. Semmel
- American Society of Hematology Research Collaborative, Washington, DC
| | - Kathleen Hewitt
- American Society of Hematology Research Collaborative, Washington, DC
| | | | | |
Collapse
|
21
|
Lee J, Ha HJ, Kim DY, Koh JS, Kim EJ. Analysis of Under-Diagnosed Malignancy during Fine Needle Aspiration Cytology of Lymphadenopathies. Int J Mol Sci 2023; 24:12394. [PMID: 37569769 PMCID: PMC10418811 DOI: 10.3390/ijms241512394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/25/2023] [Accepted: 08/02/2023] [Indexed: 08/13/2023] Open
Abstract
Fine needle aspiration cytology (FNAC) is a useful tool in the evaluation of lymphadenopathy. It is a safe and minimally invasive procedure that provides preoperative details for subsequent treatment. It can also diagnose the majority of malignant tumors. However, there are some instances where the diagnosis of tumors remains obscure. To address this, we re-analyzed the misinterpreted patients' samples using mRNA sequencing technology and then identified the characteristics of non-Hodgkin's lymphoma that tend to be under-diagnosed. To decipher the involved genes and pathways, we used bioinformatic and biological analysis approaches, identifying the response to oxygen species, inositol phosphate metabolic processes, and peroxisome and PPAR pathways as possibly being involved with this type of tumor. Notably, these analyses identified FOS, ENDOG, and PRKAR2B as hub genes. cBioPortal, a multidimensional cancer genomics database, also confirmed that these genes were associated with lymphoma patients. These results thus point to candidate genes that could be used as biomarkers to minimize the false-negative rate of FNAC diagnosis. We are currently pursuing the development of a gene chip to improve the diagnosis of lymphadenopathy patients with the ultimate goal of improving their prognosis.
Collapse
Affiliation(s)
- Jeeyong Lee
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea; (J.L.); (D.Y.K.)
| | - Hwa Jeong Ha
- Department of Pathology, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea; (H.J.H.); (J.S.K.)
- Convergence Institute of Biomedical Engineering and Biomaterials, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea
| | - Da Yeon Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea; (J.L.); (D.Y.K.)
- Department of Radiological and Medico-Oncological Sciences, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Jae Soo Koh
- Department of Pathology, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea; (H.J.H.); (J.S.K.)
| | - Eun Ju Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea; (J.L.); (D.Y.K.)
- Department of Radiological and Medico-Oncological Sciences, University of Science and Technology, Daejeon 34113, Republic of Korea
- Institute for Molecular Bioscience, The University of Queensland, Carmody Rd., St Lucia, Brisbane, QLD 4072, Australia
| |
Collapse
|
22
|
Burke HB. Standardized Documentation Is Not the Solution to Reduce Physician Time in the Electronic Health Record. JAMA Oncol 2023; 9:1151-1152. [PMID: 37289445 DOI: 10.1001/jamaoncol.2023.1523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Affiliation(s)
- Harry B Burke
- Uniformed Services University of the Health Sciences, Bethesda, Maryland
| |
Collapse
|
23
|
Aneja S, Avesta A, Xu H, Machado LO. Clinical Informatics Approaches to Facilitate Cancer Data Sharing. Yearb Med Inform 2023; 32:104-110. [PMID: 37414028 PMCID: PMC10751108 DOI: 10.1055/s-0043-1768721] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/08/2023] Open
Abstract
OBJECTIVES Despite growing enthusiasm surrounding the utility of clinical informatics to improve cancer outcomes, data availability remains a persistent bottleneck to progress. Difficulty combining data with protected health information often limits our ability to aggregate larger more representative datasets for analysis. With the rise of machine learning techniques that require increasing amounts of clinical data, these barriers have magnified. Here, we review recent efforts within clinical informatics to address issues related to safely sharing cancer data. METHODS We carried out a narrative review of clinical informatics studies related to sharing protected health data within cancer studies published from 2018-2022, with a focus on domains such as decentralized analytics, homomorphic encryption, and common data models. RESULTS Clinical informatics studies that investigated cancer data sharing were identified. A particular focus of the search yielded studies on decentralized analytics, homomorphic encryption, and common data models. Decentralized analytics has been prototyped across genomic, imaging, and clinical data with the most advances in diagnostic image analysis. Homomorphic encryption was most often employed on genomic data and less on imaging and clinical data. Common data models primarily involve clinical data from the electronic health record. Although all methods have robust research, there are limited studies showing wide scale implementation. CONCLUSIONS Decentralized analytics, homomorphic encryption, and common data models represent promising solutions to improve cancer data sharing. Promising results thus far have been limited to smaller settings. Future studies should be focused on evaluating the scalability and efficacy of these methods across clinical settings of varying resources and expertise.
Collapse
Affiliation(s)
- Sanjay Aneja
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, USA
- Center for Outcomes Research and Evaluation at Yale, New Haven, CT, USA
- Department of Bioinformatics and Data Science, Yale School of Medicine, New Haven, CT, USA
| | - Arman Avesta
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, USA
- Center for Outcomes Research and Evaluation at Yale, New Haven, CT, USA
| | - Hua Xu
- Department of Bioinformatics and Data Science, Yale School of Medicine, New Haven, CT, USA
| | - Lucila Ohno Machado
- Department of Bioinformatics and Data Science, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
24
|
Jin L, Ma J, Chen Z, Wang F, Li Z, Shang Z, Dong J. Osteoarthritis related epigenetic variations in miRNA expression and DNA methylation. BMC Med Genomics 2023; 16:163. [PMID: 37434153 PMCID: PMC10337191 DOI: 10.1186/s12920-023-01597-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 07/01/2023] [Indexed: 07/13/2023] Open
Abstract
Osteoarthritis (OA) is chronic arthritis characterized by articular cartilage degradation. However, a comprehensive regulatory network for OA-related microRNAs and DNA methylation modifications has yet to be established. Thus, we aimed to identify epigenetic changes in microRNAs and DNA methylation and establish the regulatory network between miRNAs and DNA methylation. The mRNA, miRNA, and DNA methylation expression profiles of healthy or osteoarthritis articular cartilage samples were downloaded from Gene Expression Omnibus (GEO) database, including GSE169077, GSE175961, and GSE162484. The differentially expressed genes (DEGs), differentially expressed miRNAs (DEMs), and differentially methylated genes (DMGs) were analyzed by the online tool GEO2R. DAVID and STRING databases were applied for functional enrichment analysis and protein-protein interaction (PPI) network. Potential therapeutic compounds for the treatment of OA were identified by Connectivity map (CMap) analysis. A total of 1424 up-regulated DEGs, 1558 down-regulated DEGs, 5 DEMs with high expression, 6 DEMs with low expression, 1436 hypermethylated genes, and 455 hypomethylated genes were selected. A total of 136 up-regulated and 65 downregulated genes were identified by overlapping DEGs and DEMs predicted target genes which were enriched in apoptosis and circadian rhythm. A total of 39 hypomethylated and 117 hypermethylated genes were obtained by overlapping DEGs and DMGs, which were associated with ECM receptor interactions and cellular metabolic processes, cell connectivity, and transcription. Moreover, The PPI network showed COL5A1, COL6A1, LAMA4, T3GAL6A, and TP53 were the most connective proteins. After overlapping of DEGs, DMGs and DEMs predicted targeted genes, 4 up-regulated genes and 11 down-regulated genes were enriched in the Axon guidance pathway. The top ten genes ranked by PPI network connectivity degree in the up-regulated and downregulated overlapping genes of DEGs and DMGs were further analyzed by the CMap database, and nine chemicals were predicted as potential drugs for the treatment of OA. In conclusion, TP53, COL5A1, COL6A1, LAMA4, and ST3GAL6 may play important roles in OA genesis and development.
Collapse
Affiliation(s)
- Lingpeng Jin
- Department of Orthopedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, China
| | - Jun Ma
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei, 050017, China
| | - Zhen Chen
- Department of Orthopedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, China
| | - Fei Wang
- Department of Orthopedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, China
| | - Zhikuan Li
- Department of Orthopedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, China
| | - Ziqi Shang
- Department of Orthopedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, China
| | - Jiangtao Dong
- Department of Orthopedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, China.
| |
Collapse
|
25
|
Noyd DH, Chen S, Bailey A, Janitz A, Baker A, Beasley W, Etzold N, Kendrick D, Kibbe W, Oeffinger K. Informatics tools to implement late cardiovascular risk prediction modeling for population management of high-risk childhood cancer survivors. Pediatr Blood Cancer 2023; 70:e30474. [PMID: 37283294 PMCID: PMC11110462 DOI: 10.1002/pbc.30474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 05/04/2023] [Accepted: 05/17/2023] [Indexed: 06/08/2023]
Abstract
BACKGROUND Clinical informatics tools to integrate data from multiple sources have the potential to catalyze population health management of childhood cancer survivors at high risk for late heart failure through the implementation of previously validated risk calculators. METHODS The Oklahoma cohort (n = 365) harnessed data elements from Passport for Care (PFC), and the Duke cohort (n = 274) employed informatics methods to automatically extract chemotherapy exposures from electronic health record (EHR) data for survivors 18 years old and younger at diagnosis. The Childhood Cancer Survivor Study (CCSS) late cardiovascular risk calculator was implemented, and risk groups for heart failure were compared to the Children's Oncology Group (COG) and the International Guidelines Harmonization Group (IGHG) recommendations. Analysis within the Oklahoma cohort assessed disparities in guideline-adherent care. RESULTS The Oklahoma and Duke cohorts both observed good overall concordance between the CCSS and COG risk groups for late heart failure, with weighted kappa statistics of .70 and .75, respectively. Low-risk groups showed excellent concordance (kappa > .9). Moderate and high-risk groups showed moderate concordance (kappa .44-.60). In the Oklahoma cohort, adolescents at diagnosis were significantly less likely to receive guideline-adherent echocardiogram surveillance compared with survivors younger than 13 years old at diagnosis (odds ratio [OD] 0.22; 95% confidence interval [CI]: 0.10-0.49). CONCLUSIONS Clinical informatics tools represent a feasible approach to leverage discrete treatment-related data elements from PFC or the EHR to successfully implement previously validated late cardiovascular risk prediction models on a population health level. Concordance of CCSS, COG, and IGHG risk groups using real-world data informs current guidelines and identifies inequities in guideline-adherent care.
Collapse
Affiliation(s)
- David H. Noyd
- Department of Pediatrics, The University of Oklahoma Health Sciences Center, College of Medicine, Oklahoma City, Oklahoma, USA
| | - Sixia Chen
- Department of Biostatistics and Epidemiology, The University of Oklahoma Health Sciences Center, Hudson College of Public Health, Oklahoma City, Oklahoma, USA
| | - Anna Bailey
- Department of Biostatistics and Epidemiology, The University of Oklahoma Health Sciences Center, Hudson College of Public Health, Oklahoma City, Oklahoma, USA
| | - Amanda Janitz
- Department of Biostatistics and Epidemiology, The University of Oklahoma Health Sciences Center, Hudson College of Public Health, Oklahoma City, Oklahoma, USA
| | - Ashley Baker
- Department of Pediatrics, The University of Oklahoma Health Sciences Center, College of Medicine, Oklahoma City, Oklahoma, USA
| | - William Beasley
- Department of Pediatrics, The University of Oklahoma Health Sciences Center, College of Medicine, Oklahoma City, Oklahoma, USA
| | - Nancy Etzold
- Biostatistics and Epidemiology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - David Kendrick
- Department of Medical Informatics, The University of Oklahoma Health Sciences Center, Tulsa, Oklahoma, USA
| | - Warren Kibbe
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Kevin Oeffinger
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
26
|
Osterman TJ, Yao JC, Krzyzanowska MK. Implementing Innovation: Informatics-Based Technologies to Improve Care Delivery and Clinical Research. Am Soc Clin Oncol Educ Book 2023; 43:e389880. [PMID: 37216629 DOI: 10.1200/edbk_389880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Improving technology has promised to improved health care delivery and the lives of patients. The realized benefits of technology, however, are delayed or less than anticipated. Three recent technology initiatives are reviewed: the Clinical Trials Rapid Activation Consortium (CTRAC), minimal Common Oncology Data Elements (mCODE), and electronic Patient-Reported Outcomes. Each initiative is at a different stage of maturity but promises to improve the delivery of cancer care. CTRAC is an ambitious initiative funded by the National Cancer Institute (NCI) to develop processes across multiple NCI-supported cancer centers to facilitate the development of centralized electronic health record (EHR) treatment plans. Facilitating interoperability of treatment regimens has the potential to improve sharing between centers and decrease the time to begin clinical trials. The mCODE initiative began in 2019 and is currently Standard for Trial Use version 2. This data standard provides an abstraction layer on top of EHR data and has been implemented across more than 60 organizations. Patient-reported outcomes have been shown to improve patient care in numerous studies. Best practices for how to leverage these in an oncology practice continue to evolve. These three examples show how innovative has diffused into practice and evolved cancer care delivery and highlight a movement toward patient-centered data and interoperability.
Collapse
Affiliation(s)
| | - James C Yao
- University of Texas MD Anderson Cancer Center, Houston, TX
| | | |
Collapse
|
27
|
Kempf E, Vaterkowski M, Leprovost D, Griffon N, Ouagne D, Breant S, Serre P, Mouchet A, Rance B, Chatellier G, Bellamine A, Frank M, Guerin J, Tannier X, Livartowski A, Hilka M, Daniel C. How to Improve Cancer Patients ENrollment in Clinical Trials From rEal-Life Databases Using the Observational Medical Outcomes Partnership Oncology Extension: Results of the PENELOPE Initiative in Urologic Cancers. JCO Clin Cancer Inform 2023; 7:e2200179. [PMID: 37167578 DOI: 10.1200/cci.22.00179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2023] Open
Abstract
PURPOSE To compare the computability of Observational Medical Outcomes Partnership (OMOP)-based queries related to prescreening of patients using two versions of the OMOP common data model (CDM; v5.3 and v5.4) and to assess the performance of the Greater Paris University Hospital (APHP) prescreening tool. MATERIALS AND METHODS We identified the prescreening information items being relevant for prescreening of patients with cancer. We randomly selected 15 academic and industry-sponsored urology phase I-IV clinical trials (CTs) launched at APHP between 2016 and 2021. The computability of the related prescreening criteria (PC) was defined by their translation rate in OMOP-compliant queries and by their execution rate on the APHP clinical data warehouse (CDW) containing data of 205,977 patients with cancer. The overall performance of the prescreening tool was assessed by the rate of true- and false-positive cases of three randomly selected CTs. RESULTS We defined a list of 15 minimal information items being relevant for patients' prescreening. We identified 83 PC of the 534 eligibility criteria from the 15 CTs. We translated 33 and 62 PC in queries on the basis of OMOP CDM v5.3 and v5.4, respectively (translation rates of 40% and 75%, respectively). Of the 33 PC translated in the v5.3 of the OMOP CDM, 19 could be executed on the APHP CDW (execution rate of 58%). Of 83 PC, the computability rate on the APHP CDW reached 23%. On the basis of three CTs, we identified 17, 32, and 63 patients as being potentially eligible for inclusion in those CTs, resulting in positive predictive values of 53%, 41%, and 21%, respectively. CONCLUSION We showed that PC could be formalized according to the OMOP CDM and that the oncology extension increased their translation rate through better representation of cancer natural history.
Collapse
Affiliation(s)
- Emmanuelle Kempf
- Sorbonne Université, Inserm, Université Sorbonne Paris Nord, Laboratoire d'Informatique Médicale et d'Ingénierie des Connaissances pour la e-Santé, LIMICS, Paris, France
- Department of Medical Oncology, Assistance Publique Hôpitaux de Paris, Henri Mondor Teaching Hospital, Créteil, France
| | - Morgan Vaterkowski
- Innovation and Data, Paris, IT Department, Assistance Publique Hôpitaux de Paris, Paris, France
- EPITA School of Engineering and Computer Science, Paris, France
| | - Damien Leprovost
- Sorbonne Université, Inserm, Université Sorbonne Paris Nord, Laboratoire d'Informatique Médicale et d'Ingénierie des Connaissances pour la e-Santé, LIMICS, Paris, France
- Innovation and Data, Paris, IT Department, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Nicolas Griffon
- Sorbonne Université, Inserm, Université Sorbonne Paris Nord, Laboratoire d'Informatique Médicale et d'Ingénierie des Connaissances pour la e-Santé, LIMICS, Paris, France
- Innovation and Data, Paris, IT Department, Assistance Publique Hôpitaux de Paris, Paris, France
| | - David Ouagne
- Innovation and Data, Paris, IT Department, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Stéphane Breant
- Innovation and Data, Paris, IT Department, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Patricia Serre
- Innovation and Data, Paris, IT Department, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Alexandre Mouchet
- Innovation and Data, Paris, IT Department, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Bastien Rance
- Department of Medical Informatics, Assistance Publique Hôpitaux de Paris, Centre-Université de Paris (APHP-CUP), Université de Paris, Paris, France
| | - Gilles Chatellier
- Department of Medical Informatics, Assistance Publique Hôpitaux de Paris, Centre-Université de Paris (APHP-CUP), Université de Paris, Paris, France
| | - Ali Bellamine
- Innovation and Data, Paris, IT Department, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Marie Frank
- Department of Medical Information, Paris Saclay Teaching Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | | | - Xavier Tannier
- Sorbonne Université, Inserm, Université Sorbonne Paris Nord, Laboratoire d'Informatique Médicale et d'Ingénierie des Connaissances pour la e-Santé, LIMICS, Paris, France
| | | | - Martin Hilka
- Innovation and Data, Paris, IT Department, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Christel Daniel
- Sorbonne Université, Inserm, Université Sorbonne Paris Nord, Laboratoire d'Informatique Médicale et d'Ingénierie des Connaissances pour la e-Santé, LIMICS, Paris, France
- Innovation and Data, Paris, IT Department, Assistance Publique Hôpitaux de Paris, Paris, France
| |
Collapse
|
28
|
Sweeney SM, Hamadeh HK, Abrams N, Adam SJ, Brenner S, Connors DE, Davis GJ, Fiore L, Gawel SH, Grossman RL, Hanlon SE, Hsu K, Kelloff GJ, Kirsch IR, Louv B, McGraw D, Meng F, Milgram D, Miller RS, Morgan E, Mukundan L, O'Brien T, Robbins P, Rubin EH, Rubinstein WS, Salmi L, Schaller T, Shi G, Sigman CC, Srivastava S. Challenges to Using Big Data in Cancer. Cancer Res 2023; 83:1175-1182. [PMID: 36625843 PMCID: PMC10102837 DOI: 10.1158/0008-5472.can-22-1274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 07/29/2022] [Accepted: 12/05/2022] [Indexed: 01/11/2023]
Abstract
Big data in healthcare can enable unprecedented understanding of diseases and their treatment, particularly in oncology. These data may include electronic health records, medical imaging, genomic sequencing, payor records, and data from pharmaceutical research, wearables, and medical devices. The ability to combine datasets and use data across many analyses is critical to the successful use of big data and is a concern for those who generate and use the data. Interoperability and data quality continue to be major challenges when working with different healthcare datasets. Mapping terminology across datasets, missing and incorrect data, and varying data structures make combining data an onerous and largely manual undertaking. Data privacy is another concern addressed by the Health Insurance Portability and Accountability Act, the Common Rule, and the General Data Protection Regulation. The use of big data is now included in the planning and activities of the FDA and the European Medicines Agency. The willingness of organizations to share data in a precompetitive fashion, agreements on data quality standards, and institution of universal and practical tenets on data privacy will be crucial to fully realizing the potential for big data in medicine.
Collapse
Affiliation(s)
- Shawn M. Sweeney
- American Association for Cancer Research, Philadelphia, Pennsylvania
| | | | - Natalie Abrams
- Division of Cancer Prevention, Early Detection Research Network, National Cancer Institute, Rockville, Maryland
| | - Stacey J. Adam
- Foundation for the National Institutes of Health, Bethesda, Maryland
| | - Sara Brenner
- Office of In Vitro Diagnostics, Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Dana E. Connors
- Foundation for the National Institutes of Health, Bethesda, Maryland
| | - Gerard J. Davis
- Abbott Diagnostics Division, Abbott Laboratories, Lake Forest, Illinois
| | - Louis Fiore
- Boston University School of Medicine, Boston and New England Department of Veterans Affairs, Bedford, Massachusetts
| | - Susan H. Gawel
- Abbott Diagnostics Division, Abbott Laboratories, Lake Forest, Illinois
| | - Robert L. Grossman
- Center for Translational Data Science, The University of Chicago, Chicago, Illinois
| | - Sean E. Hanlon
- Center for Strategic Scientific Initiatives, National Cancer Institute, Bethesda, Maryland
| | | | - Gary J. Kelloff
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | | | - Bill Louv
- Project Data Sphere, Morrisville, North Carolina
| | - Deven McGraw
- Ciitizen Platform at Invitae, San Francisco, California
| | - Frank Meng
- Boston University and Veterans Administration Boston Healthcare System, Boston, Massachusetts
| | | | - Robert S. Miller
- CancerLinQ, American Society of Clinical Oncology, Alexandria, Virginia
| | - Emily Morgan
- Foundation for the National Institutes of Health, Bethesda, Maryland
| | | | | | | | | | - Wendy S. Rubinstein
- Office of In Vitro Diagnostics, Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Liz Salmi
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | | | - George Shi
- Abbott Diagnostics Division, Abbott Laboratories, Lake Forest, Illinois
| | - Caroline C. Sigman
- Boston University and Veterans Administration Boston Healthcare System, Boston, Massachusetts
| | - Sudhir Srivastava
- Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, Maryland
| |
Collapse
|
29
|
Zhang S, Zheng Y, Li X, Zhang S, Hu H, Kuang W. Cellular senescence-related gene signature as a valuable predictor of prognosis in hepatocellular carcinoma. Aging (Albany NY) 2023; 15:3064-3093. [PMID: 37059592 DOI: 10.18632/aging.204658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 03/28/2023] [Indexed: 04/16/2023]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a lethal tumor. Its prognosis prediction remains a challenge. Meanwhile, cellular senescence, one of the hallmarks of cancer, and its related prognostic genes signature can provide critical information for clinical decision-making. METHOD Using bulk RNA sequencing and microarray data of HCC samples, we established a senescence score model via multi-machine learning algorithms to predict the prognosis of HCC. Single-cell and pseudo-time trajectory analyses were used to explore the hub genes of the senescence score model in HCC sample differentiation. RESULT A machine learning model based on cellular senescence gene expression profiles was identified in predicting HCC prognosis. The feasibility and accuracy of the senescence score model were confirmed in external validation and comparison with other models. Moreover, we analyzed the immune response, immune checkpoints, and sensitivity to immunotherapy drugs of HCC patients in different prognostic risk groups. Pseudo-time analyses identified four hub genes in HCC progression, including CDCA8, CENPA, SPC25, and TTK, and indicated related cellular senescence. CONCLUSIONS This study identified a prognostic model of HCC by cellular senescence-related gene expression and insight into novel potential targeted therapies.
Collapse
Affiliation(s)
- Shuqiao Zhang
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yilu Zheng
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xinyu Li
- Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Shijun Zhang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hao Hu
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Weihong Kuang
- Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, The First Dongguan Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong, China
| |
Collapse
|
30
|
Botsis T, Murray JC, Ghanem P, Balan A, Kernagis A, Hardart K, He T, Spiker J, Kreimeyer K, Tao J, Baras AS, Yegnasubramanian S, Canzoniero J, Anagnostou V. Precision Oncology Core Data Model to Support Clinical Genomics Decision Making. JCO Clin Cancer Inform 2023; 7:e2200108. [PMID: 37040583 PMCID: PMC10281442 DOI: 10.1200/cci.22.00108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 10/26/2022] [Accepted: 01/20/2023] [Indexed: 04/13/2023] Open
Abstract
PURPOSE Precision oncology mandates developing standardized common data models (CDMs) to facilitate analyses and enable clinical decision making. Expert-opinion-based precision oncology initiatives are epitomized in Molecular Tumor Boards (MTBs), which process large volumes of clinical-genomic data to match genotypes with molecularly guided therapies. METHODS We used the Johns Hopkins University MTB as a use case and developed a precision oncology core data model (Precision-DM) to capture key clinical-genomic data elements. We leveraged existing CDMs, building upon the Minimal Common Oncology Data Elements model (mCODE). Our model was defined as a set of profiles with multiple data elements, focusing on next-generation sequencing and variant annotations. Most elements were mapped to terminologies or code sets and the Fast Healthcare Interoperability Resources (FHIR). We subsequently compared our Precision-DM with existing CDMs, including the National Cancer Institute's Genomic Data Commons (NCI GDC), mCODE, OSIRIS, the clinical Genome Data Model (cGDM), and the genomic CDM (gCDM). RESULTS Precision-DM contained 16 profiles and 355 data elements. 39% of the elements derived values from selected terminologies or code sets, and 61% were mapped to FHIR. Although we used most elements contained in mCODE, we significantly expanded the profiles to include genomic annotations, resulting in a partial overlap of 50.7% between our core model and mCODE. Limited overlap was noted between Precision-DM and OSIRIS (33.2%), NCI GDC (21.4%), cGDM (9.3%), and gCDM (7.9%). Precision-DM covered most of the mCODE elements (87.7%), with less coverage for OSIRIS (35.8%), NCI GDC (11%), cGDM (26%) and gCDM (33.3%). CONCLUSION Precision-DM supports clinical-genomic data standardization to support the MTB use case and may allow for harmonized data pulls across health care systems, academic institutions, and community medical centers.
Collapse
Affiliation(s)
- Taxiarchis Botsis
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Joseph C. Murray
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Paola Ghanem
- Department of Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Archana Balan
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Alexander Kernagis
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Kent Hardart
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ting He
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jonathan Spiker
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Kory Kreimeyer
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jessica Tao
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Alexander S. Baras
- Department of Pathology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Srinivasan Yegnasubramanian
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jenna Canzoniero
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Valsamo Anagnostou
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
31
|
Gabriel PE, Singh AP, Shulman LN. Re-envisioning the Paradigm for Oncology Electronic Health Record Documentation by Paying for What Matters for Patients, Quality, and Research. JAMA Oncol 2023; 9:299-300. [PMID: 36633843 DOI: 10.1001/jamaoncol.2022.6842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
This Viewpoint discusses re-envisioning and incentivizing a unique approach to oncology electronic health record documentation.
Collapse
Affiliation(s)
- Peter E Gabriel
- Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Aditi P Singh
- Hematology-Oncology Division, University of Pennsylvania, Philadelphia
| | - Lawrence N Shulman
- Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| |
Collapse
|
32
|
Milgrom ZZ, Milgrom DP, Han Y, Hui SL, Haggstrom DA, Fisher CS, Mendonca EA. Breast Cancer Screening, Diagnosis, and Surgery during the Pre- and Peri-pandemic: Experience of Patients in a Statewide Health Information Exchange. Ann Surg Oncol 2023; 30:2883-2894. [PMID: 36749504 PMCID: PMC9904246 DOI: 10.1245/s10434-023-13119-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 01/04/2023] [Indexed: 02/08/2023]
Abstract
BACKGROUND Measures taken to address the COVID-19 pandemic interrupted routine diagnosis and care for breast cancer. The aim of this study was to characterize the effects of the pandemic on breast cancer care in a statewide cohort. PATIENTS AND METHODS Using data from a large health information exchange, we retrospectively analyzed the timing of breast cancer screening, and identified a cohort of newly diagnosed patients with any stage of breast cancer to further access the information available about their surgical treatments. We compared data for four subgroups: pre-lockdown (preLD) 25 March to 16 June 2019; lockdown (LD) 23 March to 3 May 2020; reopening (RO) 4 May to 14 June 2020; and post-lockdown (postLD) 22 March to 13 June 2021. RESULTS During LD and RO, screening mammograms in the cohort decreased by 96.3% and 36.2%, respectively. The overall breast cancer diagnosis and surgery volumes decreased up to 38.7%, and the median time to surgery was prolonged from 1.5 months to 2.4 for LD and 1.8 months for RO. Interestingly, higher mean DCIS diagnosis (5.0 per week vs. 3.1 per week, p < 0.05) and surgery volume (14.8 vs. 10.5, p < 0.05) were found for postLD compared with preLD, while median time to surgery was shorter (1.2 months vs. 1.5 months, p < 0.0001). However, the postLD average weekly screening and diagnostic mammogram did not fully recover to preLD levels (2055.3 vs. 2326.2, p < 0.05; 574.2 vs. 624.1, p < 0.05). CONCLUSIONS Breast cancer diagnosis and treatment patterns were interrupted during the lockdown and still altered 1 year after. Screening in primary care should be expanded to mitigate possible longer-term effects of these interruptions.
Collapse
Affiliation(s)
- Zheng Z. Milgrom
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN USA
| | | | - Yan Han
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN USA
| | - Siu L. Hui
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN USA ,Regenstrief Institute, Indianapolis, IN USA
| | - David A. Haggstrom
- Regenstrief Institute, Indianapolis, IN USA ,Richard L. Roudebush VA Medical Center, Indianapolis, IN USA ,Department of Internal Medicine, Indiana University School of Medicine, Indianapolis, IN USA
| | - Carla S. Fisher
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN USA
| | - Eneida A. Mendonca
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| |
Collapse
|
33
|
Wyatt KD, Noyd DH, Wood NM, Phillips CA, Miller TP, Rubin EM, Winestone LE, Waanders AJ, Perentesis JP, Aplenc R. Data standards in pediatric oncology: Past, present, and future. Pediatr Blood Cancer 2023; 70:e30128. [PMID: 36495256 DOI: 10.1002/pbc.30128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/19/2022] [Accepted: 11/10/2022] [Indexed: 12/14/2022]
Abstract
In this commentary, we highlight the central role that data standards play in facilitating data-driven efforts to advance research in pediatric oncology. We discuss the current state of data standards for pediatric oncology and propose five steps to achieve an improved future state with benefits for clinicians, researchers, and patients.
Collapse
Affiliation(s)
- Kirk D Wyatt
- Division of Pediatric Hematology/Oncology, Roger Maris Cancer Center, Sanford Health, Fargo, North Dakota, USA
| | - David H Noyd
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Nicole M Wood
- Department of Hematology/Oncology, Children's Mercy Kansas City, Kansas City, Missouri, USA.,Department of Health Informatics & Technology, Children's Mercy Kansas City, Kansas City, Missouri, USA
| | - Charles A Phillips
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Tamara P Miller
- Aflac Cancer & Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA.,Department of Pediatrics, Emory University, Atlanta, Georgia, USA
| | - Elyssa M Rubin
- Hundai Cancer Institute, Children's Hospital of Orange County, Orange, California, USA
| | - Lena E Winestone
- Division of Allergy, Immunology and BMT, Department of Pediatrics, University of California San Francisco Benioff Children's Hospitals, San Francisco, California, USA
| | - Angela J Waanders
- Division of Hematology, Oncology, Neurooncology and Stem Cell Transplant, Ann & Robert H Lurie Children's Hospital of Chicago, Chicago, Illinois, USA.,Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - John P Perentesis
- Cancer & Blood Diseases Institute, Cincinnati Children's Hospital, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Richard Aplenc
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| |
Collapse
|
34
|
Mathieu JE. Teams, Teaming, and Complex Systems in Cancer Care. JCO Oncol Pract 2023; 19:6-9. [PMID: 36516363 DOI: 10.1200/op.22.00650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- John E Mathieu
- Department of Management, School of Business, University of Connecticut, Storrs, CT
| |
Collapse
|
35
|
Dolin RH, Heale BSE, Alterovitz G, Gupta R, Aronson J, Boxwala A, Gothi SR, Haines D, Hermann A, Hongsermeier T, Husami A, Jones J, Naeymi-Rad F, Rapchak B, Ravishankar C, Shalaby J, Terry M, Xie N, Zhang P, Chamala S. Introducing HL7 FHIR Genomics Operations: a developer-friendly approach to genomics-EHR integration. J Am Med Inform Assoc 2022; 30:485-493. [PMID: 36548217 PMCID: PMC9933060 DOI: 10.1093/jamia/ocac246] [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] [Received: 09/27/2022] [Revised: 11/16/2022] [Accepted: 12/02/2022] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE Enabling clinicians to formulate individualized clinical management strategies from the sea of molecular data remains a fundamentally important but daunting task. Here, we describe efforts towards a new paradigm in genomics-electronic health record (HER) integration, using a standardized suite of FHIR Genomics Operations that encapsulates the complexity of molecular data so that precision medicine solution developers can focus on building applications. MATERIALS AND METHODS FHIR Genomics Operations essentially "wrap" a genomics data repository, presenting a uniform interface to applications. More importantly, operations encapsulate the complexity of data within a repository and normalize redundant data representations-particularly relevant in genomics, where a tremendous amount of raw data exists in often-complex non-FHIR formats. RESULTS Fifteen FHIR Genomics Operations have been developed, designed to support a wide range of clinical scenarios, such as variant discovery; clinical trial matching; hereditary condition and pharmacogenomic screening; and variant reanalysis. Operations are being matured through the HL7 balloting process, connectathons, pilots, and the HL7 FHIR Accelerator program. DISCUSSION Next-generation sequencing can identify thousands to millions of variants, whose clinical significance can change over time as our knowledge evolves. To manage such a large volume of dynamic and complex data, new models of genomics-EHR integration are needed. Qualitative observations to date suggest that freeing application developers from the need to understand the nuances of genomic data, and instead base applications on standardized APIs can not only accelerate integration but also dramatically expand the applications of Omic data in driving precision care at scale for all.
Collapse
Affiliation(s)
- Robert H Dolin
- Corresponding Author: Robert H. Dolin, MD, Elimu Informatics, 1709 Julian Ct, El Cerrito, CA 94530, USA;
| | | | - Gil Alterovitz
- Brigham and Women’s Hospital, Boston, Massachusetts, USA,Harvard/MIT Division of Health Sciences and Technology, Harvard Medical School, Boston, Massachusetts, USA
| | - Rohan Gupta
- Shri Mata Vaishno Devi University, Katra, Jammu and Kashmir, India
| | | | | | - Shaileshbhai R Gothi
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida, USA
| | - David Haines
- Leap of Faith Technologies, Libertyville, Illinois, USA
| | - Arthur Hermann
- Department of Health IT Strategy & Policy, Kaiser Permanente, Pasadena, California, USA
| | | | - Ammar Husami
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - James Jones
- Computational Health Informatics Program, Boston Children’s Hospital, Boston, Massachusetts, USA
| | | | | | | | | | - May Terry
- MITRE Corporation, McLean, Virginia, USA
| | - Ning Xie
- Biomedical Cybernetics Laboratory, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Powell Zhang
- Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Srikar Chamala
- Keck School of Medicine, Department of Pathology, University of Southern California, Los Angeles, California, USA,Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, California, USA
| |
Collapse
|
36
|
Lu Z, Chen Y, Chen S, Zhu X, Wang C, Wang Z, Yao Q. Comprehensive Prognostic Analysis of Immune Implication Value and Oxidative Stress Significance of NECAP2 in Low-Grade Glioma. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1494520. [PMID: 36531205 PMCID: PMC9750773 DOI: 10.1155/2022/1494520] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/20/2022] [Accepted: 09/22/2022] [Indexed: 12/07/2023]
Abstract
Adaptin ear-binding coat-associated protein 2 (NECAP2) belongs to the family of proteins encoding adaptin-ear-binding coat-associated proteins. However, its immune effect on tumors and its microenvironment are still unclear. Here, we systematically evaluated the differences (variations) in NECAP2 expression for low-grade glioma (LGG) and pan-cancer in the LGG dataset of The Cancer Genome Atlas (TCGA) utilizing bioinformatics methods. We found for the first time that NECAP2 level was elevated in gliomas and that this upregulation increased as the tumor grade increased. In addition, Pearson correlations of NECAP2 with five immune pathways and significant gene mutations associated with NECAP2 were also analyzed. Univariate survival and multivariate Cox analyses were used to compare the clinical characteristics and survival of the patients. Glioma patients with NECAP2 overexpression have a remarkably higher risk of developing malignant behavior and a worse prognosis. The correlation between the expression levels of NECAP2 and the prognosis of glioma patients was identified. Kaplan-Meier curves showed that patients with upregulated NECAP2 expression exhibited an unfavorable prognosis. Western blotting showed that NECAP2 was overexpressed in glioma patients. IHC staining results illustrated an elevation in the NECAP2 protein expression level with the development of tumor malignancy. Additionally, qRT-PCR verified that oxidative stress in glioma tissues reduced the expression of stress-related genes and oxidative stress capacity compared to normal tissues, which may be associated with tumor evasion of immune surveillance and tumor progression. In vitro wound-healing and Transwell assay confirmed that NECAP2 promotes glioma cell migration and invasion. Our study also thoroughly examined the immune significance of NECAP2 in the TCGA-LGG samples, using CIBERSORT and ESTIMATE to explore the correlation between NECAP2 and cancer immune infiltration. The NECAP2 expression levels were correlated with the infiltration degree of immune cells such as neutrophils, CD4+ T cells, macrophages, CD8+ T cells, and B cells. Therefore, our results indicate that NECAP2 strongly correlates with the overall immune infiltration level of glioma and could independently serve as a prognostic biological marker for glioma patients.
Collapse
Affiliation(s)
- Zhichao Lu
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Yixun Chen
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Siqi Chen
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Xingjia Zhu
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Chenxing Wang
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Ziheng Wang
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
- Department of Clinical Biobank & Institute of Oncology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
- Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Qi Yao
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| |
Collapse
|
37
|
Huang X, Lu Z, He M, Feng Y, Yu S, Shen B, Lu J, Wu P, Pan B, Ding H, Chen C, Sun Y. A Prognostic Risk Model of a Novel Oxidative Stress-Related Signature Predicts Clinical Prognosis and Demonstrates Immune Relevancy in Lung Adenocarcinoma. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2262014. [PMID: 36439693 PMCID: PMC9699774 DOI: 10.1155/2022/2262014] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 10/10/2022] [Accepted: 10/13/2022] [Indexed: 01/17/2024]
Abstract
Lung adenocarcinoma (LUAD) is among the most prevalent malignant lung cancers with a poor prognosis due to high invasiveness and lethality despite multiple treatments. Since the lung is an important organ associated with oxidative stress, and it has been confirmed that oxidative stress represents a potential cancer-specific depletion, it is of important significance to investigate and evaluate the clinical value of oxidative stress mechanisms regulating tumor cell apoptosis. Furthermore, there are few studies on the impact of the microenvironment on reaction to immune-checkpoint inhibitors (ICIs) in patients with LUAD. Based on the TCGA-LUAD dataset, which is stratified into a training set as well as a validation set in a ratio of 2 : 1, this investigation constructs and validates a prognostic predictive power of a gene signature model of oxidative stress-related prognostic signatures. To ascertain the differences between the high-risk score group and the low-risk score group in tumor-infiltrating lymphocytes and patients' response to ICI therapy. This oxidative stress-related prognostic gene signature is composed of MAP3K19 and NTSR1 and is an independent prognosis-related factor in the LUAD group. The outcome of patients having a low risk score is better, and the difference was statistically significant, and individuals with a low risk score had a larger number of infiltrating immune cell distribution in the tumor microenvironment, which was closely related to clinical outcome. Our study suggests that the synergistic effect of oxidative stress-related prognostic gene markers-MAP3K19 and NTSR1 has clinical significance in the prognosis identification and immunotherapy of LUAD patients. Thus, the results may help to better intersect the oxidative stress-related mechanisms in clinical value in LUAD but requires prospective validation.
Collapse
Affiliation(s)
- Xing Huang
- Department of Pathology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & Nanjing Medical University Affiliated Cancer Hospital, China
| | - Zhichao Lu
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Min He
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Yipeng Feng
- Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, 21009 Nanjing, China
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, China
| | - Shaorong Yu
- Department of Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing 210000, China
| | - Bo Shen
- Department of Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing 210000, China
| | - Jianwei Lu
- Department of Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing 210000, China
| | - Pingping Wu
- Department of Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing 210000, China
| | - Banzhou Pan
- Department of Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing 210000, China
| | - Hanlin Ding
- Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, 21009 Nanjing, China
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing, China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, China
| | - Chen Chen
- Department of Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing 210000, China
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, China
| | - Yidan Sun
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
38
|
Lyu HG, Kantor O, Laws AD, McDonald J, Pham L, Dominici LS, Vincuilla J, Raut CP, Danilchuk B, Novak L, Parker T, King TA, Mittendorf EA. Development of an Electronic Health Record Registry to Facilitate Collection of Commission on Cancer Metrics for Patients Undergoing Surgery for Breast Cancer. JCO Clin Cancer Inform 2022; 6:e2200012. [DOI: 10.1200/cci.22.00012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
PURPOSE Accurate and efficient data collection is a challenge for quality improvement initiatives and clinical research. We describe the development of a custom electronic health record (EHR)–based registry to automatically extract structured Commission on Cancer axillary surgery-specific metrics from a custom synoptic note template included in the operative reports for patients with breast cancer undergoing surgery. METHODS The smart functionality of our enterprise-based EHR system was leveraged to create a custom smart phrase to capture axillary surgery-specific variables. A multidisciplinary team developed structured data elements correlating to each axillary surgery-specific variable. These data elements were then included in a note template for the operative report. Each variable could be aggregated and converted into a single flat database through the EHR's reporting workbench and serve as a live, prospective registry for all users within the EHR. RESULTS The final axillary surgery-specific note template in a synoptic format allowed for efficient and easy entry and automatic collection of breast cancer–specific metrics. From initial adoption in February 2021-December 2021, there were 1,254 patients who underwent breast surgery with axillary surgery. The operative notes allowed for automatic capture of metrics from 60.5% (n = 759) of patients. Data capture improved from 37.6% in the initial adoption period of 6 months to 86.2% in the last 5 months. CONCLUSION We were able to demonstrate successful implementation of provider-driven structured data entry into EHR systems that permits automatic data capture. The end result is a custom synoptic note template and a real-time, prospective registry of breast cancer–specific Commission on Cancer metrics that are robust enough to use for quality improvement initiatives and clinical research.
Collapse
Affiliation(s)
- Heather G. Lyu
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, TX
| | - Olga Kantor
- Division of Breast Surgery, Brigham and Women's Hospital, Boston, MA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA
| | - Alison D. Laws
- Division of Breast Surgery, Brigham and Women's Hospital, Boston, MA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA
| | | | - Lisa Pham
- Mass General Brigham, Somerville, MA
| | - Laura S. Dominici
- Division of Breast Surgery, Brigham and Women's Hospital, Boston, MA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA
| | - Julie Vincuilla
- Division of Breast Surgery, Brigham and Women's Hospital, Boston, MA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA
| | - Chandrajit P. Raut
- Division of Surgical Oncology, Brigham and Women's Hospital, Boston, MA
- Sarcoma Center, Dana-Farber Brigham Cancer Center, Boston, MA
| | - Bryan Danilchuk
- Division of Breast Surgery, Brigham and Women's Hospital, Boston, MA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA
| | - Lara Novak
- Division of Breast Surgery, Brigham and Women's Hospital, Boston, MA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA
| | - Tonia Parker
- Division of Breast Surgery, Brigham and Women's Hospital, Boston, MA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA
| | - Tari A. King
- Division of Breast Surgery, Brigham and Women's Hospital, Boston, MA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA
| | - Elizabeth A. Mittendorf
- Division of Breast Surgery, Brigham and Women's Hospital, Boston, MA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA
| |
Collapse
|
39
|
Precision Oncology in Canada: Converting Vision to Reality with Lessons from International Programs. Curr Oncol 2022; 29:7257-7271. [PMID: 36290849 PMCID: PMC9600134 DOI: 10.3390/curroncol29100572] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 09/13/2022] [Accepted: 09/20/2022] [Indexed: 11/25/2022] Open
Abstract
Canada's healthcare system, like others worldwide, is immersed in a process of evolution, attempting to adapt conventional frameworks of health technology assessment (HTA) and funding models to a new landscape of precision medicine in oncology. In particular, the need for real-world evidence in Canada is not matched by the necessary infrastructure and technologies required to integrate genomic and clinical data. Since healthcare systems in many developed nations face similar challenges, we adopted a solutions-based approach and conducted a search of worldwide programs in personalized medicine, with an emphasis on precision oncology. This search strategy included review articles published between 1 January 2016 and 1 March 2021 and hand-searches of their reference lists for relevant publications back to 1 December 2005. Thirty-nine initiatives across 37 countries in Europe, Australasia, Africa, and the Americas had the potential to lead to real-world data (RWD) on the clinical utility of oncology biomarkers. We highlight four initiatives with helpful lessons for Canada: Genomic Medicine France 2025, UNICANCER, the German Medical Informatics Initiative, and CANCER-ID. Among the 35 other programs evaluated, the main themes included the need for collaboration and systems to support data harmonization across multiple jurisdictions. In order to generate RWD in precision oncology that will prove acceptable to HTA bodies, Canada must take a national approach to biomarker strategy and unite all stakeholders at the highest level to overcome jurisdictional and technological barriers.
Collapse
|
40
|
Wang L, Fu S, Wen A, Ruan X, He H, Liu S, Moon S, Mai M, Riaz IB, Wang N, Yang P, Xu H, Warner JL, Liu H. Assessment of Electronic Health Record for Cancer Research and Patient Care Through a Scoping Review of Cancer Natural Language Processing. JCO Clin Cancer Inform 2022; 6:e2200006. [PMID: 35917480 PMCID: PMC9470142 DOI: 10.1200/cci.22.00006] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/18/2022] [Accepted: 06/15/2022] [Indexed: 11/20/2022] Open
Abstract
PURPOSE The advancement of natural language processing (NLP) has promoted the use of detailed textual data in electronic health records (EHRs) to support cancer research and to facilitate patient care. In this review, we aim to assess EHR for cancer research and patient care by using the Minimal Common Oncology Data Elements (mCODE), which is a community-driven effort to define a minimal set of data elements for cancer research and practice. Specifically, we aim to assess the alignment of NLP-extracted data elements with mCODE and review existing NLP methodologies for extracting said data elements. METHODS Published literature studies were searched to retrieve cancer-related NLP articles that were written in English and published between January 2010 and September 2020 from main literature databases. After the retrieval, articles with EHRs as the data source were manually identified. A charting form was developed for relevant study analysis and used to categorize data including four main topics: metadata, EHR data and targeted cancer types, NLP methodology, and oncology data elements and standards. RESULTS A total of 123 publications were selected finally and included in our analysis. We found that cancer research and patient care require some data elements beyond mCODE as expected. Transparency and reproductivity are not sufficient in NLP methods, and inconsistency in NLP evaluation exists. CONCLUSION We conducted a comprehensive review of cancer NLP for research and patient care using EHRs data. Issues and barriers for wide adoption of cancer NLP were identified and discussed.
Collapse
Affiliation(s)
- Liwei Wang
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Rochester, MN
| | - Sunyang Fu
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Rochester, MN
| | - Andrew Wen
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Rochester, MN
| | - Xiaoyang Ruan
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Rochester, MN
| | - Huan He
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Rochester, MN
| | - Sijia Liu
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Rochester, MN
| | - Sungrim Moon
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Rochester, MN
| | - Michelle Mai
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Rochester, MN
| | - Irbaz B. Riaz
- Department of Hematology/Oncology, Mayo Clinic, Scottsdale, AZ
| | - Nan Wang
- Department of Computer Science and Engineering, College of Science and Engineering, University of Minnesota, Minneapolis, MN
| | - Ping Yang
- Department of Quantitative Health Sciences, Mayo Clinic, Scottsdale, AZ
| | - Hua Xu
- School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX
| | - Jeremy L. Warner
- Departments of Medicine (Hematology/Oncology), Vanderbilt University, Nashville, TN
- Department Biomedical Informatics, Vanderbilt University, Nashville, TN
| | - Hongfang Liu
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Rochester, MN
| |
Collapse
|
41
|
Huang M, Zhu Z, Nong C, Liang Z, Ma J, Li G. Bioinformatics analysis identifies diagnostic biomarkers and their correlation with immune infiltration in diabetic nephropathy. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:669. [PMID: 35845512 PMCID: PMC9279778 DOI: 10.21037/atm-22-1682] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 06/01/2022] [Indexed: 11/16/2022]
Abstract
Background Diabetic nephropathy (DN) is a major cause of end-stage renal disease (ESRD). Currently, microalbuminuria is mainly used as a diagnostic indicator of DN, but there are still limitations and lack of immune-related diagnostic markers. In this study, we aimed to explore diagnostic biomarkers associated with immune infiltration of DN. Methods Immune-related differentially expressed genes (DEGs) were derived from those at the intersection of the ImmPort database and DEGs identified from 3 datasets, which were based on the Gene Expression Omnibus (GEO). Functional enrichment analyses were performed; a protein-protein interaction (PPI) network was constructed; and hub genes were identified by Search Tool for the Retrieval of Interacting Genes/Proteins (STRING). After screening the key genes using least absolute shrinkage and selection operator (LASSO) and support vector machine recursive feature elimination (SVM-RFE), a prediction model for DN was constructed. The predictive performance of the model was quantified by receiver-operating characteristic curve, decision curve analysis, and nomogram. Next, infiltration of 22 types of immune cells in DN kidney tissue was evaluated using cell-type identification by estimating relative subsets of RNA transcripts (CIBERSORT). Expression of diagnostic markers was analyzed in DN and control patient groups to determine the genes with the maximum diagnostic potential. Finally, we explored the correlation between diagnostic markers and immune cells. Results Overall, 191 immune-related DEGs were identified, that primarily positively regulated with cell adhesion, T cell activation, leukocyte proliferation and migration, urogenital system development, lymphocyte differentiation and proliferation, and mononuclear cell proliferation. Gene sets were related to the PI3K-Akt, MAPK, Rap1, and WNT signaling pathways. Finally, CCL19, CD1C, and IL33 were identified as diagnostic markers of DN and recognized in the 3 datasets [area under the curve (AUC) =0.921]. Immune cell infiltration analysis demonstrated that CCL19 was positively correlated with macrophages M1 (R=0.47, P<0.001) and macrophages M2 (R=0.75, P<0.001). CD1C was positively correlated with macrophages M1 (R=0.47, P<0.05), macrophages M2 (R=0.75, P<0.01), and monocytes (R=0.42, P<0.01). IL33 was positively correlated with macrophages M1 (R=0.45, P<0.05), macrophages M2 (R=0.74, P<0.01), and monocytes (R=0.41, P<0.01). Conclusions Our results provide evidence that CCL19, CD1C, and IL33, which are associated with immune infiltration, are the potential diagnostic biomarkers for DN candidates.
Collapse
Affiliation(s)
- Menglan Huang
- Department of Nephrology, The People's Hospital of Baise, Baise, China
| | - Zhengxi Zhu
- Department of Nephrology, The People's Hospital of Baise, Baise, China
| | - Cong Nong
- Department of Nephrology, The People's Hospital of Baise, Baise, China
| | - Zhao Liang
- Department of Nephrology, The People's Hospital of Baise, Baise, China
| | - Jingxue Ma
- Department of Nephrology, The People's Hospital of Baise, Baise, China
| | - Guangzhi Li
- Department of General Medicine, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| |
Collapse
|
42
|
Emamekhoo H, Carroll CB, Stietz C, Pier JB, Lavitschke MD, Mulkerin D, Sesto ME, Tevaarwerk AJ. Supporting Structured Data Capture for Patients With Cancer: An Initiative of the University of Wisconsin Carbone Cancer Center Survivorship Program to Improve Capture of Malignant Diagnosis and Cancer Staging Data. JCO Clin Cancer Inform 2022; 6:e2200020. [PMID: 35802837 PMCID: PMC9296185 DOI: 10.1200/cci.22.00020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/27/2022] [Accepted: 05/16/2022] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Structured data elements within electronic health records are health-related information that can be entered, stored, and extracted in an organized manner at later time points. Tracking outcomes for cancer survivors is also enabled by structured data. We sought to increase structured data capture within oncology practices at multiple sites sharing the same electronic health records. METHODS Applying engineering approaches and the Plan-Do-Study-Act cycle, we launched dual quality improvement initiatives to ensure that a malignant diagnosis and stage were captured as structured data. Intervention: Close Visit Validation (CVV) requires providers to satisfy certain criteria before closing ambulatory encounters. CVV may be used to track open clinical encounters and chart delinquencies to encourage optimal clinical workflows. We added two cancer-specific required criteria at the time of closing encounters in oncology clinics: (1) the presence of at least one malignant diagnosis on the Problem List and (2) staging all the malignant diagnoses on the Problem List when appropriate. RESULTS Six months before the CVV implementation, the percentage of encounters with a malignant diagnosis on the Problem List at the time of the encounter was 65%, whereas the percentage of encounters with a staged diagnosis was 32%. Three months after cancer-specific CVV implementation, the percentages were 85% and 75%, respectively. Rates had increased to 90% and 88% more than 2 years after implementation. CONCLUSION Oncologist performance improved after the implementation of cancer-specific CVV criteria, with persistently high percentages of relevant malignant diagnoses and cancer stage structured data capture 2 years after the intervention.
Collapse
Affiliation(s)
- Hamid Emamekhoo
- University of Wisconsin, Madison, WI
- Carbone Cancer Center, Madison, WI
| | | | | | | | | | | | - Mary E. Sesto
- University of Wisconsin, Madison, WI
- Carbone Cancer Center, Madison, WI
| | | |
Collapse
|
43
|
Thanarajasingam G, Minasian LM, Bhatnagar V, Cavalli F, De Claro RA, Dueck AC, El-Galaly TC, Everest N, Geissler J, Gisselbrecht C, Gormley N, Gribben J, Horowitz M, Ivy SP, Jacobson CA, Keating A, Kluetz PG, Kwong YL, Little RF, Matasar MJ, Mateos MV, McCullough K, Miller RS, Mohty M, Moreau P, Morton LM, Nagai S, Nair A, Nastoupil L, Robertson K, Sidana S, Smedby KE, Sonneveld P, Tzogani K, van Leeuwen FE, Velikova G, Villa D, Wingard JR, Seymour JF, Habermann TM. Reaching beyond maximum grade: progress and future directions for modernising the assessment and reporting of adverse events in haematological malignancies. Lancet Haematol 2022; 9:e374-e384. [PMID: 35483398 PMCID: PMC9241484 DOI: 10.1016/s2352-3026(22)00045-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/20/2022] [Accepted: 02/02/2022] [Indexed: 12/15/2022]
Abstract
Remarkable improvements in outcomes for many haematological malignancies have been driven primarily by a proliferation of novel therapeutics over the past two decades. Targeted agents, immune and cellular therapies, and combination regimens have adverse event profiles distinct from conventional finite cytotoxic chemotherapies. In 2018, a Commission comprising patient advocates, clinicians, clinical investigators, regulators, biostatisticians, and pharmacists representing a broad range of academic and clinical cancer expertise examined issues of adverse event evaluation in the context of both newer and existing therapies for haematological cancers. The Commission proposed immediate actions and long-term solutions in the current processes in adverse event assessment, patient-reported outcomes in haematological malignancies, toxicities in cellular therapies, long-term toxicity and survivorship in haematological malignancies, issues in regulatory approval from an international perspective, and toxicity reporting in haematological malignancies and the real-world setting. In this follow-up report, the Commission describes progress that has been made in these areas since the initial report.
Collapse
Affiliation(s)
| | - Lori M Minasian
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Vishal Bhatnagar
- Oncology Center for Excellence, US Food and Drug Administration, Silver Spring, MD, USA
| | - Franco Cavalli
- Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - R Angelo De Claro
- Office of Oncologic Diseases, US Food and Drug Administration, Silver Spring, MD, USA
| | - Amylou C Dueck
- Division of Quantitative Health Sciences Research, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - Tarec C El-Galaly
- Department of Haematology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Neil Everest
- Health Resourcing Group, Australian Government Department of Health, Canberra, ACT, Australia
| | - Jan Geissler
- Leukaemia Patient Advocates Foundation, Bern, Switzerland
| | - Christian Gisselbrecht
- Haemato-Oncology Department, Hopital Saint-Louis, Institute Haematology, Paris Diderot University VII, Paris, France; European Medicines Agency, London, UK
| | - Nicole Gormley
- Office of Oncologic Diseases, US Food and Drug Administration, Silver Spring, MD, USA
| | - John Gribben
- Centre for Haemato-Oncology, Barts Cancer Institute, London, UK
| | - Mary Horowitz
- Center for International Blood and Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, WI, USA
| | - S Percy Ivy
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | - Paul G Kluetz
- Oncology Center for Excellence, US Food and Drug Administration, Silver Spring, MD, USA
| | - Yok Lam Kwong
- Department of Haematology and Haematologic Oncology, University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Richard F Little
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Matthew J Matasar
- Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | - Robert S Miller
- CancerLinQ, American Society of Clinical Oncology, Alexandria, VA, USA
| | - Mohamad Mohty
- Haematology and Cellular Therapy Department, Sorbonne University, Saint-Antoine Hospital (AP-HP), INSERM UMRs 938, Paris, France
| | - Philippe Moreau
- Department of Haematology, University Hospital Nantes, Nantes, France
| | - Lindsay M Morton
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sumimasa Nagai
- Department of Medical Development, Institute for Advancement of Clinical and Translational Science, Kyoto University Hospital, Kyoto, Japan; Pharmaceuticals and Medical Devices Agency, Tokyo, Japan
| | - Abhilasha Nair
- Oncology Center for Excellence, US Food and Drug Administration, Silver Spring, MD, USA
| | | | - Kaye Robertson
- Office of Product Review, Therapeutic Goods Administration, Canberra, ACT, Australia
| | - Surbhi Sidana
- Division of BMT and Cellular Therapy, Stanford University School of Medicine, Stanford, CA, USA
| | - Karin E Smedby
- Department of Medicine Solna, Division of Clinical Epidemiology, Karolinska Institutet, Stockholm, Sweden; Department of Haematology, Karolinska University Hospital, Stockholm, Sweden
| | - Pieter Sonneveld
- Department of Haematology, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | | | | | - Galina Velikova
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Diego Villa
- BC Cancer Centre for Lymphoid Cancer and University of British Columbia, Vancouver, BC, Canada
| | - John R Wingard
- Division of Haematology & Oncology, University of Florida College of Medicine, Gainesville, FL, USA
| | - John F Seymour
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Royal Melbourne Hospital, Melbourne, VIC, Australia; University of Melbourne, Melbourne, VIC, Australia
| | | |
Collapse
|
44
|
Crowson MG, Moukheiber D, Arévalo AR, Lam BD, Mantena S, Rana A, Goss D, Bates DW, Celi LA. A systematic review of federated learning applications for biomedical data. PLOS DIGITAL HEALTH 2022; 1:e0000033. [PMID: 36812504 PMCID: PMC9931322 DOI: 10.1371/journal.pdig.0000033] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/30/2022] [Indexed: 11/18/2022]
Abstract
OBJECTIVES Federated learning (FL) allows multiple institutions to collaboratively develop a machine learning algorithm without sharing their data. Organizations instead share model parameters only, allowing them to benefit from a model built with a larger dataset while maintaining the privacy of their own data. We conducted a systematic review to evaluate the current state of FL in healthcare and discuss the limitations and promise of this technology. METHODS We conducted a literature search using PRISMA guidelines. At least two reviewers assessed each study for eligibility and extracted a predetermined set of data. The quality of each study was determined using the TRIPOD guideline and PROBAST tool. RESULTS 13 studies were included in the full systematic review. Most were in the field of oncology (6 of 13; 46.1%), followed by radiology (5 of 13; 38.5%). The majority evaluated imaging results, performed a binary classification prediction task via offline learning (n = 12; 92.3%), and used a centralized topology, aggregation server workflow (n = 10; 76.9%). Most studies were compliant with the major reporting requirements of the TRIPOD guidelines. In all, 6 of 13 (46.2%) of studies were judged at high risk of bias using the PROBAST tool and only 5 studies used publicly available data. CONCLUSION Federated learning is a growing field in machine learning with many promising uses in healthcare. Few studies have been published to date. Our evaluation found that investigators can do more to address the risk of bias and increase transparency by adding steps for data homogeneity or sharing required metadata and code.
Collapse
Affiliation(s)
- Matthew G. Crowson
- Department of Otolaryngology-Head & Neck Surgery, Massachusetts Eye & Ear, Boston, Massachusetts, United States of America
- Department of Otolaryngology-Head & Neck Surgery, Harvard Medical School, Massachusetts, United States of America
| | - Dana Moukheiber
- Laboratory for Computational Physiology, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| | - Aldo Robles Arévalo
- IDMEC, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Data & Analytics, NTT DATA Portugal, Lisbon, Portugal
| | - Barbara D. Lam
- Department of Hematology & Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Sreekar Mantena
- Harvard College, Boston, Massachusetts, United States of America
| | - Aakanksha Rana
- Massachusetts Institute of Technology, Boston, Massachusetts, United States of America
| | - Deborah Goss
- Department of Otolaryngology-Head & Neck Surgery, Massachusetts Eye & Ear, Boston, Massachusetts, United States of America
| | - David W. Bates
- Division of General Internal Medicine and Primary Care, Brigham and Women’s Hospital, Boston, MA, United States of America
- Department of Health Policy and Management, Harvard T. H. Chan School of Public Health, Boston, MA, United States of America
| | - Leo Anthony Celi
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Division of Pulmonary, Critical Care and Sleep Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| |
Collapse
|
45
|
Shreve JT, Khanani SA, Haddad TC. Artificial Intelligence in Oncology: Current Capabilities, Future Opportunities, and Ethical Considerations. Am Soc Clin Oncol Educ Book 2022; 42:1-10. [PMID: 35687826 DOI: 10.1200/edbk_350652] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The promise of highly personalized oncology care using artificial intelligence (AI) technologies has been forecasted since the emergence of the field. Cumulative advances across the science are bringing this promise to realization, including refinement of machine learning- and deep learning algorithms; expansion in the depth and variety of databases, including multiomics; and the decreased cost of massively parallelized computational power. Examples of successful clinical applications of AI can be found throughout the cancer continuum and in multidisciplinary practice, with computer vision-assisted image analysis in particular having several U.S. Food and Drug Administration-approved uses. Techniques with emerging clinical utility include whole blood multicancer detection from deep sequencing, virtual biopsies, natural language processing to infer health trajectories from medical notes, and advanced clinical decision support systems that combine genomics and clinomics. Substantial issues have delayed broad adoption, with data transparency and interpretability suffering from AI's "black box" mechanism, and intrinsic bias against underrepresented persons limiting the reproducibility of AI models and perpetuating health care disparities. Midfuture projections of AI maturation involve increasing a model's complexity by using multimodal data elements to better approximate an organic system. Far-future positing includes living databases that accumulate all aspects of a person's health into discrete data elements; this will fuel highly convoluted modeling that can tailor treatment selection, dose determination, surveillance modality and schedule, and more. The field of AI has had a historical dichotomy between its proponents and detractors. The successful development of recent applications, and continued investment in prospective validation that defines their impact on multilevel outcomes, has established a momentum of accelerated progress.
Collapse
Affiliation(s)
| | | | - Tufia C Haddad
- Department of Oncology, Mayo Clinic, Rochester, MN.,Center for Digital Health, Mayo Clinic, Rochester, MN
| |
Collapse
|
46
|
Wheatley-Price P, Moore S, Lee CW. The Role of Real-World Evidence to Support Treatment Choices in Malignant Pleural Mesothelioma. JTO Clin Res Rep 2022; 3:100300. [PMID: 35510132 PMCID: PMC9059068 DOI: 10.1016/j.jtocrr.2022.100300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 11/22/2022] Open
Affiliation(s)
- Paul Wheatley-Price
- Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Sara Moore
- Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | | |
Collapse
|
47
|
Post AR, Burningham Z, Halwani AS. Electronic Health Record Data in Cancer Learning Health Systems: Challenges and Opportunities. JCO Clin Cancer Inform 2022; 6:e2100158. [PMID: 35353547 PMCID: PMC9005105 DOI: 10.1200/cci.21.00158] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 01/04/2022] [Accepted: 02/18/2022] [Indexed: 12/21/2022] Open
Affiliation(s)
- Andrew R. Post
- Research Informatics Shared Resource, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
- Department of Biomedical Informatics, University of Utah School of Medicine, Salt Lake City, UT
| | - Zachary Burningham
- Division of Epidemiology, Department of Internal Medicine, University of Utah, Salt Lake City, UT
| | - Ahmad S. Halwani
- Division of Hematology and Hematologic Malignancies, Department of Internal Medicine, University of Utah, Salt Lake City, UT
| |
Collapse
|
48
|
Huang X, Wu B, Zhang F, Chen F, Zhang Y, Guo H, Zhang H. Epigenetic Biomarkers Screening of Non-Coding RNA and DNA Methylation Based on Peripheral Blood Monocytes in Smokers. Front Genet 2022; 13:766553. [PMID: 35233217 PMCID: PMC8882369 DOI: 10.3389/fgene.2022.766553] [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: 11/11/2021] [Accepted: 01/17/2022] [Indexed: 11/13/2022] Open
Abstract
This study aims to use bioinformatics methods to determine the epigenetic changes in microRNA expression and DNA methylation caused by cigarette smoking. The data of mRNA, miRNA expression, and methylation microarray were obtained from the GEO database to filter differentially expressed genes (DEGs), differentially expressed miRNAs (DEMs), and methylated CpG probes (DMPs) through the limma package. The R clusterProfile package was used for functional annotation and enrichment analysis. The protein-protein interaction (PPI) network was constructed by the String database and visualized in Cytoscape software. Starbase database was employed to predict lncRNA and CirRNA based on the sequence of miRNA, and to establish a regulatory network of ceRNA. By overlapping DEG and DEM, 107 down-miRNA-targeted up-regulated genes and 65 up-miRNA-target down-regulated genes were obtained, which were mainly enriched in autophagy signaling pathways and protein ubiquitination pathways, respectively. In addition, 324 genes with low methylation and high expression and 204 genes with high methylation and low expression were respectively related to the degeneration of the nervous system and the function of the cardiovascular system. Interestingly, 43 genes were up-regulated under the dual regulation of reduced miRNA and hypomethylation, while 14 genes were down-regulated under the dual regulation of increased miRNA and hypermethylation. Ten chemicals have been identified as putative therapeutic agents for pathological conditions caused by smoking. In addition, among these genes, HSPA4, GRB2, PRKCA, and BCL2L1 could play a fundamental role in related diseases caused by smoking and may be used as the biomarkers for precise diagnosis and targets for future therapies of smoking-related diseases.
Collapse
Affiliation(s)
- Xiaowei Huang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Bian Wu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Fangxue Zhang
- Knee Surgery Department of the Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Peking University, Beijing, China
| | - Fancheng Chen
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yong Zhang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Huizhi Guo
- The First Institute of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hongtao Zhang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
- *Correspondence: Hongtao Zhang,
| |
Collapse
|
49
|
Upadhyay VA, Johnson BE, Landman AB, Hassett MJ. Real-World Analysis of Off-Label Use of Molecularly Targeted Therapy in a Large Academic Medical Center Cohort. JCO Precis Oncol 2022; 6:e2100232. [PMID: 35050710 DOI: 10.1200/po.21.00232] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE The primary objective of this study is to quantify the use of off-label molecularly targeted therapy and describe the clinical situations in which off-label targeted therapy are used. A key secondary objective is to report the outcomes of patients treated with off-label use of targeted therapy. PATIENTS AND METHODS We searched the electronic health record between 2000 and 2020 at our center to characterize the volume, clinical settings, and outcomes associated with off-label use of targeted therapies in different types of solid tumors. RESULTS Among 46,712 patients who received targeted therapies, we identified 119 instances of off-label use of targeted therapy. Colon cancer was the most common cancer type to receive off-label targeted therapy in 18 patients (15.1%), followed by 13 with non-small-cell lung cancer (10.9%), eight with cholangiocarcinoma (6.7%), and seven with glioblastoma (5.9%). The most frequent molecular rationale for off-label therapy came from a comprehensive next-generation sequencing test (53.7%). The most frequently mutated gene that provided the rationale for targeted therapy was BRAF (20.1%), with BRAFV600E being the most common molecular alteration overall (15.1%). The median duration of off-label targeted therapy was 3.58 months, and the overall survival of treated patients was 7.59 months. There were 37 patients (31.1%) treated for longer than 6 months, 23 patients (19.3%) who survived ≥ 2 years, and 13 patients who were still on therapy as of June 2020. CONCLUSION In this large cohort study of patients with solid tumors, off-label use of targeted therapy was uncommon. With that said, a notable proportion of patients had treatment durations ≥ 6 months and survivals of ≥ 2 years.
Collapse
Affiliation(s)
- Vivek A Upadhyay
- Dana-Farber Cancer Institute, Boston, MA.,Massachusetts General Hospital, Boston, MA.,Harvard Medical School, Boston, MA
| | - Bruce E Johnson
- Dana-Farber Cancer Institute, Boston, MA.,Harvard Medical School, Boston, MA
| | - Adam B Landman
- Harvard Medical School, Boston, MA.,Brigham and Women's Hospital, Boston, MA
| | - Michael J Hassett
- Dana-Farber Cancer Institute, Boston, MA.,Harvard Medical School, Boston, MA
| |
Collapse
|
50
|
Schorer AE, Moldwin R, Koskimaki J, Bernstam EV, Venepalli NK, Miller RS, Chen JL. Chasm Between Cancer Quality Measures and Electronic Health Record Data Quality. JCO Clin Cancer Inform 2022; 6:e2100128. [PMID: 34985912 PMCID: PMC9848533 DOI: 10.1200/cci.21.00128] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 10/27/2021] [Accepted: 11/15/2021] [Indexed: 01/26/2023] Open
Abstract
PURPOSE The Medicare Access and CHIP Reauthorization Act of 2015 (MACRA) requires eligible clinicians to report clinical quality measures (CQMs) in the Merit-Based Incentive Payment System (MIPS) to maximize reimbursement. To determine whether structured data in electronic health records (EHRs) were adequate to report MIPS CQMs, EHR data aggregated by ASCO's CancerLinQ platform were analyzed. MATERIALS AND METHODS Using the CancerLinQ health technology platform, 19 Oncology MIPS (oMIPS) CQMs were evaluated to determine the presence of data elements (DEs) necessary to satisfy each CQM and the DE percent population with patient data (fill rates). At the time of this analysis, the CancerLinQ network comprised 63 active practices, representing eight different EHR vendors and containing records for more than 1.63 million unique patients with one or more malignant neoplasms (1.73 million cancer cases). RESULTS Fill rates for the 63 oMIPS-associated DEs varied widely among the practices. The average site had at least one filled DE for 52% of the DEs. Only 35% of the DEs were populated for at least one patient record in 95% of the practices. However, the average DE fill rate of all practices was 23%. No data were found at any practice for 22% of the DEs. Since any oMIPS CQM with an unpopulated DE component resulted in an inability to compute the measure, only two (10.5%) of the 19 oMIPS CQMs were computable for more than 1% of the patients. CONCLUSION Although EHR systems had relatively high DE fill rates for some DEs, underfilling and inconsistency of DEs in EHRs render automated oncology MIPS CQM calculations impractical.
Collapse
Affiliation(s)
| | | | - Jacob Koskimaki
- CancerLinQ, American Society of Clinical Oncology, Alexandria, VA
| | - Elmer V. Bernstam
- The University of Texas School of Biomedical Informatics at Houston and Division of General Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, TX
| | | | - Robert S. Miller
- CancerLinQ, American Society of Clinical Oncology, Alexandria, VA
| | - James L. Chen
- Departments of Internal Medicine and Biomedical Informatics, The Ohio State University, Columbus, OH
| |
Collapse
|