1
|
Chen X, Xu W, Pan J, Yang H, Li Y, Chen X, Sun Y, Liu Q, Qiu S. m6A methylation profiling as a prognostic marker in nasopharyngeal carcinoma: insights from MeRIP-Seq and RNA-Seq. Front Immunol 2024; 15:1492648. [PMID: 39726587 PMCID: PMC11669702 DOI: 10.3389/fimmu.2024.1492648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 11/21/2024] [Indexed: 12/28/2024] Open
Abstract
Background Nasopharyngeal carcinoma (NPC) is a type of malignant tumors commonly found in Southeast Asia and China, with insidious onset and clinical symptoms. N6-methyladenosine (m6A) modification significantly contributes to tumorigenesis and progression by altering RNA secondary structure and influencing RNA-protein binding at the transcriptome level. However, the mechanism and role of abnormal m6A modification in nasopharyngeal carcinoma remain unclear. Methods Nasopharyngeal Carcinoma tissues from 3 patients and non-cancerous nasopharyngeal tissues from 3 individuals, all from Fujian Cancer Hospital, were sequenced for m6A methylation. These were combined with transcriptome sequencing data from 192 nasopharyngeal cancer tissues. Genes linked to prognosis were discovered using differential analysis and univariate Cox regression. Subsequently, a prognostic model associated with m6A was developed through the application of LASSO regression analysis. The model's accuracy was verified using both internal transcriptome databases and external databases. An extensive evaluation of the tumor's immune microenvironment and signaling pathways was performed, analyzing both transcriptomic and single-cell data. Results The m6A methylation sequencing analysis revealed 194 genes with varying expression levels, many of which are predominantly associated with immune pathways. By integrating transcriptome sequencing data, 19 m6A-modified genes were found to be upregulated in tumor tissues, leading to the development of a three-gene (EME1, WNT4, SHISA2) risk prognosis model. The group with lower risk exhibited notable enrichment in pathways related to immunity, displaying traits like enhanced survival rates, stronger immune profiles, and increased responsiveness to immunotherapy when compared to the higher-risk group. Single-cell analysis revealed that malignant cells exhibited the highest risk score levels compared to immune cells, with a high-risk score indicating worse biological behavior. The three hub genes demonstrated significant correlation with m6A modification regulators, and MeRIP-RT-PCR confirmed the occurrence of m6A methylation in these genes within nasopharyngeal carcinoma cells. Conclusions A prognostic model for nasopharyngeal carcinoma risk based on m6A modification genes was developed, and its prognostic value was confirmed through self-assessment data. The study highlighted the crucial impact of m6A modification on the immune landscape of nasopharyngeal cancer.
Collapse
Affiliation(s)
- Xiaochuan Chen
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Wenqian Xu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Junping Pan
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Hanxuan Yang
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Yi Li
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Xin Chen
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Yingming Sun
- Department of Radiation and Medical Oncology, Affiliated Sanming First Hospital of Fujian Medical University, Sanming, China
| | - Qinying Liu
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Sufang Qiu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| |
Collapse
|
2
|
Lutz S, D'Angelo A, Hammerl S, Schmutz M, Claus R, Fischer NM, Kramer F, Hammoud Z. Unveiling the Digital Evolution of Molecular Tumor Boards. Target Oncol 2024:10.1007/s11523-024-01109-1. [PMID: 39609355 DOI: 10.1007/s11523-024-01109-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2024] [Indexed: 11/30/2024]
Abstract
Molecular tumor boards (MTB) are interdisciplinary conferences involving various experts discussing patients with advanced tumors, to derive individualized treatment suggestions based on molecular variants. These discussions involve using heterogeneous internal data, such as patient clinical data, but also external resources such as knowledge databases for annotations and search for relevant clinical studies. This imposes a certain level of complexity that requires huge effort to homogenize the data and use it in a speedy manner to reach the needed treatment. For this purpose, most institutions involving an MTB are heading toward automation and digitalization of the process, hence reducing manual work requiring human intervention and subsequently time in deriving personalized treatment suggestions. The tools are also used to better visualize the patient's data, which allows a refined overview for the board members. In this paper, we present the results of our thorough literature research about MTBs, their process, the most common knowledge bases, and tools used to support this decision-making process.
Collapse
Affiliation(s)
- Sebastian Lutz
- IT-Infrastructure for Translational Medical Research, University of Augsburg, Augsburg, Germany.
| | - Alicia D'Angelo
- IT-Infrastructure for Translational Medical Research, University of Augsburg, Augsburg, Germany
| | - Sonja Hammerl
- IT-Infrastructure for Translational Medical Research, University of Augsburg, Augsburg, Germany
| | - Maximilian Schmutz
- Institute of Digital Medicine (IDM), Medical Faculty, University of Augsburg, Augsburg, Germany
- Hematology and Oncology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany
| | - Rainer Claus
- Hematology and Oncology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany
- Pathology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Comprehensive Cancer Center Augsburg (CCCA), Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Nina M Fischer
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany
- Comprehensive Cancer Center Augsburg (CCCA), Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Frank Kramer
- IT-Infrastructure for Translational Medical Research, University of Augsburg, Augsburg, Germany
| | - Zaynab Hammoud
- IT-Infrastructure for Translational Medical Research, University of Augsburg, Augsburg, Germany
| |
Collapse
|
3
|
Nagy P, Papp J, Grolmusz VK, Bozsik A, Pócza T, Oláh E, Patócs A, Butz H. Comprehensive Clinical Genetics, Molecular and Pathological Evaluation Efficiently Assist Diagnostics and Therapy Selection in Breast Cancer Patients with Hereditary Genetic Background. Int J Mol Sci 2024; 25:12546. [PMID: 39684258 DOI: 10.3390/ijms252312546] [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: 11/04/2024] [Revised: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Using multigene panel testing for the diagnostic evaluation of patients with hereditary breast and ovarian cancer (HBOC) syndrome often identifies clinically actionable variants in genes with varying levels of penetrance. High-penetrance genes (BRCA1, BRCA2, CDH1, PALB2, PTEN, STK11, TP53) inform specific clinical surveillance and therapeutic decisions, while recommendations for moderate-penetrance genes (ATM, BARD1, BRIP1, CHEK2, MLH1, MSH2, MSH6, PMS2, EPCAM, NF1, RAD51C, RAD51D) are more limited. A detailed disease history, including pedigree data, helps formulate the most appropriate and personalised management strategies. In this study, we evaluated the clinical benefits of comprehensive hereditary cancer gene panel testing and a pre-sent questionnaire in Hungarian patients with suspected HBOC syndrome. We prospectively enrolled 513 patients referred for HBOC testing. Of these, 463 met the genetic testing criteria, while 50 did not but were tested due to potential therapeutic indications. Additionally, a retrospective cohort of 47 patients who met the testing criteria but had previously only been tested for BRCA1/2 was also analysed. Among the 463 patients in the prospective cohort, 96 (20.7%) harboured pathogenic/likely pathogenic (P/LP) variants-67 in high-penetrance genes and 29 in moderate-penetrance genes. This ratio was similar in the retrospective cohort (6/47; 12.7%). In patients who did not meet the testing criteria, no mutations in high-penetrance genes were found, and only 3 of 50 (6%) harboured P/LP variants in moderate-penetrance genes. Secondary findings (P/LP variants in non-HBOC-associated genes) were identified in two patients. In the prospective cohort, P/LP variants in BRCA1 and BRCA2 were the most prevalent (56/96; 58.3%), and the extended testing doubled the P/LP detection ratio. Among moderate-penetrance genes, five cases (three in the prospective and two in the retrospective cohorts) had P/LP variants in Lynch syndrome-associated genes. Further immunohistochemistry analysis of breast tumour tissues helped clarify the causative role of these variants. Comprehensive clinical and molecular genetic evaluation is beneficial for the diagnosis and management of patients with P/LP variants in hereditary tumour-predisposing genes and can serve as a basis for effective therapy selection, such as PARP inhibitors or immunotherapy.
Collapse
Affiliation(s)
- Petra Nagy
- Department of Molecular Genetics and The National Tumour Biology Laboratory, National Institute of Oncology, Comprehensive Cancer Centre, Ráth György u. 7-9, 1122 Budapest, Hungary
| | - János Papp
- Department of Molecular Genetics and The National Tumour Biology Laboratory, National Institute of Oncology, Comprehensive Cancer Centre, Ráth György u. 7-9, 1122 Budapest, Hungary
- HUN-REN-SE Hereditary Tumours Research Group, Hungarian Research Network, Nagyvárad tér 4, 1089 Budapest, Hungary
| | - Vince Kornél Grolmusz
- Department of Molecular Genetics and The National Tumour Biology Laboratory, National Institute of Oncology, Comprehensive Cancer Centre, Ráth György u. 7-9, 1122 Budapest, Hungary
- HUN-REN-SE Hereditary Tumours Research Group, Hungarian Research Network, Nagyvárad tér 4, 1089 Budapest, Hungary
| | - Anikó Bozsik
- Department of Molecular Genetics and The National Tumour Biology Laboratory, National Institute of Oncology, Comprehensive Cancer Centre, Ráth György u. 7-9, 1122 Budapest, Hungary
- HUN-REN-SE Hereditary Tumours Research Group, Hungarian Research Network, Nagyvárad tér 4, 1089 Budapest, Hungary
| | - Tímea Pócza
- Department of Molecular Genetics and The National Tumour Biology Laboratory, National Institute of Oncology, Comprehensive Cancer Centre, Ráth György u. 7-9, 1122 Budapest, Hungary
| | - Edit Oláh
- Department of Molecular Genetics and The National Tumour Biology Laboratory, National Institute of Oncology, Comprehensive Cancer Centre, Ráth György u. 7-9, 1122 Budapest, Hungary
| | - Attila Patócs
- Department of Molecular Genetics and The National Tumour Biology Laboratory, National Institute of Oncology, Comprehensive Cancer Centre, Ráth György u. 7-9, 1122 Budapest, Hungary
- HUN-REN-SE Hereditary Tumours Research Group, Hungarian Research Network, Nagyvárad tér 4, 1089 Budapest, Hungary
- Department of Laboratory Medicine, Semmelweis University, Nagyvárad tér 4, 1089 Budapest, Hungary
| | - Henriett Butz
- Department of Molecular Genetics and The National Tumour Biology Laboratory, National Institute of Oncology, Comprehensive Cancer Centre, Ráth György u. 7-9, 1122 Budapest, Hungary
- HUN-REN-SE Hereditary Tumours Research Group, Hungarian Research Network, Nagyvárad tér 4, 1089 Budapest, Hungary
- Department of Laboratory Medicine, Semmelweis University, Nagyvárad tér 4, 1089 Budapest, Hungary
- Department of Oncology Biobank, National Institute of Oncology, Comprehensive Cancer Centre, Ráth György u. 7-9, 1122 Budapest, Hungary
| |
Collapse
|
4
|
Liu S, Obert C, Yu YP, Zhao J, Ren BG, Liu JJ, Wiseman K, Krajacich BJ, Wang W, Metcalfe K, Smith M, Ben-Yehezkel T, Luo JH. Utility analyses of AVITI sequencing chemistry. BMC Genomics 2024; 25:778. [PMID: 39127634 DOI: 10.1186/s12864-024-10686-4] [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: 01/31/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND DNA sequencing is a critical tool in modern biology. Over the last two decades, it has been revolutionized by the advent of massively parallel sequencing, leading to significant advances in the genome and transcriptome sequencing of various organisms. Nevertheless, challenges with accuracy, lack of competitive options and prohibitive costs associated with high throughput parallel short-read sequencing persist. RESULTS Here, we conduct a comparative analysis using matched DNA and RNA short-reads assays between Element Biosciences' AVITI and Illumina's NextSeq 550 chemistries. Similar comparisons were evaluated for synthetic long-read sequencing for RNA and targeted single-cell transcripts between the AVITI and Illumina's NovaSeq 6000. For both DNA and RNA short-read applications, the study found that the AVITI produced significantly higher per sequence quality scores. For PCR-free DNA libraries, we observed an average 89.7% lower experimentally determined error rate when using the AVITI chemistry, compared to the NextSeq 550. For short-read RNA quantification, AVITI platform had an average of 32.5% lower error rate than that for NextSeq 550. With regards to synthetic long-read mRNA and targeted synthetic long read single cell mRNA sequencing, both platforms' respective chemistries performed comparably in quantification of genes and isoforms. The AVITI displayed a marginally lower error rate for long reads, with fewer chemistry-specific errors and a higher mutation detection rate. CONCLUSION These results point to the potential of the AVITI platform as a competitive candidate in high-throughput short read sequencing analyses when juxtaposed with the Illumina NextSeq 550.
Collapse
Affiliation(s)
- Silvia Liu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA.
- High Throughput Genome Center, University of Pittsburgh School of Medicine, Pittsburgh, USA.
- Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, Pittsburgh, USA.
| | - Caroline Obert
- Element Biosciences Inc, 10055 Barnes Canyon Road, Suite 100, San Diego, CA, 92121, USA
| | - Yan-Ping Yu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
- High Throughput Genome Center, University of Pittsburgh School of Medicine, Pittsburgh, USA
- Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Junhua Zhao
- Element Biosciences Inc, 10055 Barnes Canyon Road, Suite 100, San Diego, CA, 92121, USA
| | - Bao-Guo Ren
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
- High Throughput Genome Center, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Jia-Jun Liu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
- High Throughput Genome Center, University of Pittsburgh School of Medicine, Pittsburgh, USA
- Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Kelly Wiseman
- Element Biosciences Inc, 10055 Barnes Canyon Road, Suite 100, San Diego, CA, 92121, USA
| | - Benjamin J Krajacich
- Element Biosciences Inc, 10055 Barnes Canyon Road, Suite 100, San Diego, CA, 92121, USA
| | - Wenjia Wang
- Department of Biostatistics, University of Pittsburgh School of Public Health, Pittsburgh, USA
| | - Kyle Metcalfe
- Element Biosciences Inc, 10055 Barnes Canyon Road, Suite 100, San Diego, CA, 92121, USA
| | - Mat Smith
- Element Biosciences Inc, 10055 Barnes Canyon Road, Suite 100, San Diego, CA, 92121, USA
| | - Tuval Ben-Yehezkel
- Element Biosciences Inc, 10055 Barnes Canyon Road, Suite 100, San Diego, CA, 92121, USA
| | - Jian-Hua Luo
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA.
- High Throughput Genome Center, University of Pittsburgh School of Medicine, Pittsburgh, USA.
- Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, Pittsburgh, USA.
| |
Collapse
|
5
|
Thein KZ, Myat YM, Park BS, Panigrahi K, Kummar S. Target-Driven Tissue-Agnostic Drug Approvals-A New Path of Drug Development. Cancers (Basel) 2024; 16:2529. [PMID: 39061168 PMCID: PMC11274498 DOI: 10.3390/cancers16142529] [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: 06/22/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024] Open
Abstract
The regulatory approvals of tumor-agnostic therapies have led to the re-evaluation of the drug development process. The conventional models of drug development are histology-based. On the other hand, the tumor-agnostic drug development of a new drug (or combination) focuses on targeting a common genomic biomarker in multiple cancers, regardless of histology. The basket-like clinical trials with multiple cohorts allow clinicians to evaluate pan-cancer efficacy and toxicity. There are currently eight tumor agnostic approvals granted by the Food and Drug Administration (FDA). This includes two immune checkpoint inhibitors, and five targeted therapy agents. Pembrolizumab is an anti-programmed cell death protein-1 (PD-1) antibody that was the first FDA-approved tumor-agnostic treatment for unresectable or metastatic microsatellite instability-high (MSI-H) or deficient mismatch repair (dMMR) solid tumors in 2017. It was later approved for tumor mutational burden-high (TMB-H) solid tumors, although the TMB cut-off used is still debated. Subsequently, in 2021, another anti-PD-1 antibody, dostarlimab, was also approved for dMMR solid tumors in the refractory setting. Patients with fusion-positive cancers are typically difficult to treat due to their rare prevalence and distribution. Gene rearrangements or fusions are present in a variety of tumors. Neurotrophic tyrosine kinase (NTRK) fusions are present in a range of pediatric and adult solid tumors in varying frequency. Larotrectinib and entrectinib were approved for neurotrophic tyrosine kinase (NTRK) fusion-positive cancers. Similarly, selpercatinib was approved for rearranged during transfection (RET) fusion-positive solid tumors. The FDA approved the first combination therapy of dabrafenib, a B-Raf proto-oncogene serine/threonine kinase (BRAF) inhibitor, plus trametinib, a mitogen-activated protein kinase (MEK) inhibitor for patients 6 months or older with unresectable or metastatic tumors (except colorectal cancer) carrying a BRAFV600E mutation. The most recent FDA tumor-agnostic approval is of fam-trastuzumab deruxtecan-nxki (T-Dxd) for HER2-positive solid tumors. It is important to identify and expeditiously develop drugs that have the potential to provide clinical benefit across tumor types.
Collapse
Affiliation(s)
- Kyaw Z. Thein
- Division of Hematology and Medical Oncology, Comprehensive Cancer Centers of Nevada—Central Valley, 3730 S Eastern Ave, Las Vegas, NV 89169, USA
- Department of Medicine, Kirk Kerkorian School of Medicine, University of Nevada Las Vegas (UNLV), 4505 S, Maryland Pkwy, Las Vegas, NV 89154, USA
- College of Osteopathic Medicine, Touro University Nevada, Touro College and University System, 874 American Pacific Dr, Henderson, NV 89014, USA
| | - Yin M. Myat
- Belfield Campus, University College Dublin (UCD) School of Medicine, D04 V1W8 Dublin, Ireland;
- Department of Internal Medicine, One Brooklyn Health—Interfaith Medical Center Campus, 1545, Atlantic Avenue, Brooklyn, NY 11213, USA;
| | - Byung S. Park
- OHSU-PSU School of Public Health, Portland, OR 97201, USA;
- Biostatistics Shared Resource, OHSU Knight Cancer Institute, OHSU School of Medicine, Portland, OR 97239, USA
| | - Kalpana Panigrahi
- Department of Internal Medicine, One Brooklyn Health—Interfaith Medical Center Campus, 1545, Atlantic Avenue, Brooklyn, NY 11213, USA;
| | - Shivaani Kummar
- Division of Hematology & Medical Oncology, Center for Experimental Therapeutics, Knight Cancer Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, USA;
| |
Collapse
|
6
|
Liu S, Obert C, Yu YP, Zhao J, Ren BG, Liu JJ, Wiseman K, Krajacich BJ, Wang W, Metcalfe K, Smith M, Ben-Yehezkel T, Luo JH. Utility Analyses of AVITI Sequencing Chemistry. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.18.590136. [PMID: 38712138 PMCID: PMC11071311 DOI: 10.1101/2024.04.18.590136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Background DNA sequencing is a critical tool in modern biology. Over the last two decades, it has been revolutionized by the advent of massively parallel sequencing, leading to significant advances in the genome and transcriptome sequencing of various organisms. Nevertheless, challenges with accuracy, lack of competitive options and prohibitive costs associated with high throughput parallel short-read sequencing persist. Results Here, we conduct a comparative analysis using matched DNA and RNA short-reads assays between Element Biosciences' AVITI and Illumina's NextSeq 550 chemistries. Similar comparisons were evaluated for synthetic long-read sequencing for RNA and targeted single-cell transcripts between the AVITI and Illumina's NovaSeq 6000. For both DNA and RNA short-read applications, the study found that the AVITI produced significantly higher per sequence quality scores. For PCR-free DNA libraries, we observed an average 89.7% lower experimentally determined error rate when using the AVITI chemistry, compared to the NextSeq 550. For short-read RNA quantification, AVITI platform had an average of 32.5% lower error rate than that for NextSeq 550. With regards to synthetic long-read mRNA and targeted synthetic long read single cell mRNA sequencing, both platforms' respective chemistries performed comparably in quantification of genes and isoforms. The AVITI displayed a marginally lower error rate for long reads, with fewer chemistry-specific errors and a higher mutation detection rate. Conclusion These results point to the potential of the AVITI platform as a competitive candidate in high-throughput short read sequencing analyses when juxtaposed with the Illumina NextSeq 550.
Collapse
Affiliation(s)
- Silvia Liu
- Department of Pathology, University of Pittsburgh School of Medicine, United States
- High Throughput Genome Center, University of Pittsburgh School of Medicine, United States
- Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, United States
| | - Caroline Obert
- Element Biosciences Inc, 10055 Barnes Canyon Road, Suite 100, San Diego, CA 92121, United States
| | - Yan-Ping Yu
- Department of Pathology, University of Pittsburgh School of Medicine, United States
- High Throughput Genome Center, University of Pittsburgh School of Medicine, United States
- Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, United States
| | - Junhua Zhao
- Element Biosciences Inc, 10055 Barnes Canyon Road, Suite 100, San Diego, CA 92121, United States
| | - Bao-Guo Ren
- Department of Pathology, University of Pittsburgh School of Medicine, United States
- High Throughput Genome Center, University of Pittsburgh School of Medicine, United States
| | - Jia-Jun Liu
- Department of Pathology, University of Pittsburgh School of Medicine, United States
- High Throughput Genome Center, University of Pittsburgh School of Medicine, United States
- Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, United States
| | - Kelly Wiseman
- Element Biosciences Inc, 10055 Barnes Canyon Road, Suite 100, San Diego, CA 92121, United States
| | - Benjamin J. Krajacich
- Element Biosciences Inc, 10055 Barnes Canyon Road, Suite 100, San Diego, CA 92121, United States
| | - Wenjia Wang
- Department of Biostatistics, University of Pittsburgh School of Public Health, United States
| | - Kyle Metcalfe
- Element Biosciences Inc, 10055 Barnes Canyon Road, Suite 100, San Diego, CA 92121, United States
| | - Mat Smith
- Element Biosciences Inc, 10055 Barnes Canyon Road, Suite 100, San Diego, CA 92121, United States
| | - Tuval Ben-Yehezkel
- Element Biosciences Inc, 10055 Barnes Canyon Road, Suite 100, San Diego, CA 92121, United States
| | - Jian-Hua Luo
- Department of Pathology, University of Pittsburgh School of Medicine, United States
- High Throughput Genome Center, University of Pittsburgh School of Medicine, United States
- Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, United States
| |
Collapse
|
7
|
Deschildre J, Vandemoortele B, Loers JU, De Preter K, Vermeirssen V. Evaluation of single-sample network inference methods for precision oncology. NPJ Syst Biol Appl 2024; 10:18. [PMID: 38360881 PMCID: PMC10869342 DOI: 10.1038/s41540-024-00340-w] [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: 07/11/2023] [Accepted: 01/17/2024] [Indexed: 02/17/2024] Open
Abstract
A major challenge in precision oncology is to detect targetable cancer vulnerabilities in individual patients. Modeling high-throughput omics data in biological networks allows identifying key molecules and processes of tumorigenesis. Traditionally, network inference methods rely on many samples to contain sufficient information for learning, resulting in aggregate networks. However, to implement patient-tailored approaches in precision oncology, we need to interpret omics data at the level of individual patients. Several single-sample network inference methods have been developed that infer biological networks for an individual sample from bulk RNA-seq data. However, only a limited comparison of these methods has been made and many methods rely on 'normal tissue' samples as reference, which are not always available. Here, we conducted an evaluation of the single-sample network inference methods SSN, LIONESS, SWEET, iENA, CSN and SSPGI using transcriptomic profiles of lung and brain cancer cell lines from the CCLE database. The methods constructed functional gene networks with distinct network characteristics. Hub gene analyses revealed different degrees of subtype-specificity across methods. Single-sample networks were able to distinguish between tumor subtypes, as exemplified by node strength clustering, enrichment of known subtype-specific driver genes among hubs and differential node strength. We also showed that single-sample networks correlated better to other omics data from the same cell line as compared to aggregate networks. We conclude that single-sample network inference methods can reflect sample-specific biology when 'normal tissue' samples are absent and we point out peculiarities of each method.
Collapse
Affiliation(s)
- Joke Deschildre
- Lab for Computational Biology, Integromics and Gene Regulation (CBIGR), Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Boris Vandemoortele
- Lab for Computational Biology, Integromics and Gene Regulation (CBIGR), Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Jens Uwe Loers
- Lab for Computational Biology, Integromics and Gene Regulation (CBIGR), Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Katleen De Preter
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Lab of Translational Onco-genomics and Bio-informatics, Center for Medical Biotechnology (VIB-UGent), Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Vanessa Vermeirssen
- Lab for Computational Biology, Integromics and Gene Regulation (CBIGR), Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium.
| |
Collapse
|
8
|
Kwon R, Yeung CCS. Advances in next-generation sequencing and emerging technologies for hematologic malignancies. Haematologica 2024; 109:379-387. [PMID: 37584286 PMCID: PMC10828783 DOI: 10.3324/haematol.2022.282442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 08/17/2023] [Indexed: 08/17/2023] Open
Abstract
Innovations in molecular diagnostics have often evolved through the study of hematologic malignancies. Examples include the pioneering characterization of the Philadelphia chromosome by cytogenetics in the 1970s, the implementation of polymerase chain reaction for high-sensitivity detection and monitoring of mutations and, most recently, targeted next- generation sequencing to drive the prognostic and therapeutic assessment of leukemia. Hematologists and hematopath- ologists have continued to advance in the past decade with new innovations improving the type, amount, and quality of data generated for each molecule of nucleic acid. In this review article, we touch on these new developments and discuss their implications for diagnostics in hematopoietic malignancies. We review advances in sequencing platforms and library preparation chemistry that can lead to faster turnaround times, novel sequencing techniques, the development of mobile laboratories with implications for worldwide benefits, the current status of sample types, improvements to quality and reference materials, bioinformatic pipelines, and the integration of machine learning and artificial intelligence into mol- ecular diagnostic tools for hematologic malignancies.
Collapse
Affiliation(s)
- Regina Kwon
- Department of Laboratory Medicine and Pathology, University of Washington
| | - Cecilia C. S. Yeung
- Department of Laboratory Medicine and Pathology, University of Washington
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| |
Collapse
|
9
|
Luo D, Wang X, Feng W. Comprehensive analysis of cuproptosis and copper homeostasis genotyping and related immune land scape in lung adenocarcinoma. Sci Rep 2023; 13:16554. [PMID: 37783723 PMCID: PMC10545825 DOI: 10.1038/s41598-023-43795-3] [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: 06/29/2023] [Accepted: 09/28/2023] [Indexed: 10/04/2023] Open
Abstract
Cuproptosis is a manner of cell death which is related to the homeostasis of copper ions in the cellular environment and is expected to open a new direction of anti-tumor therapy. However, the studies on cuproptosis and copper homeostasis in lung adenocarcinoma (LUAD) are still limited. In this study, we identified new cuproptosis and copper homeostasis related genes (CHRGs) which were effective in stratifying genotyping clusters with survival differences based on transcriptomic data obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO). Weighted Gene Co-expression Network Analysis (WGCNA) further expands the screening boundary of CHRGs, and finally we established a 10-CHRGs-based prognostic signature using lasso-penalized cox regression method, which were validated in GSE30219. Comprehensive bioinformatics analysis revealed these genes are potential regulators of modulating immunotherapy efficacy, drug resistance, tumor microenvironment infiltration, and tumor mutation patterns. Lastly, the scRNA-seq datasets GSE183219 and GSE203360 offers the evidences that CHRGs signature are mainly distributed in cancer epithelial cells, real time quantitative polymerase chain reaction (RT-qPCR) also confirmed the differential expression of these genes between normal lung cell line and lung adenocarcinoma cell lines. Collectively, our findings revealed new cuproptosis and copper homeostasis related genotyping clusters and genes which may play important roles in predicting prognosis, influencing tumor microenvironment and drug efficacy in LUAD patients.
Collapse
Affiliation(s)
- Dayuan Luo
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Xiang Wang
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Wei Feng
- Department of Cardiothoracic Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China.
| |
Collapse
|
10
|
Nelakurthi VM, Paul P, Reche A. Bioinformatics in Early Cancer Detection. Cureus 2023; 15:e46931. [PMID: 38021627 PMCID: PMC10640668 DOI: 10.7759/cureus.46931] [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: 08/03/2023] [Accepted: 10/12/2023] [Indexed: 12/01/2023] Open
Abstract
Bioinformatics is a pretty recent branch of biology that encompasses the use of algebraic, analytic, and computing approaches to the processing and interpretation of biological information. A wide term, "bioinformatics" refers to the use of digital technology to study biological processes using high-dimensional data collected from many resources. The design and testing of the software tools required to evaluate the information are the core of bioinformatics research, which is conducted in great portions in silico and typically involves the synthesis of new learning from available data. Early diagnosis of cancer results in improved prognosis, but at the same time, it is difficult to conform to diagnosis at a very early stage. The use of DNA microarrays and proteomics studies for large-scale gene expression research has advanced technology, thus elevating the significance of bioinformatics tools. In today's research, wet experimentation and the application of bioinformatics analytics go side by side. Molecular profiling of tumor biopsies is becoming more and more crucial to both cancer research and the treatment of cancer.
Collapse
Affiliation(s)
- Vidya Maheswari Nelakurthi
- Public Health Dentistry, Sharad Pawar Dental College and Hospital, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Priyanka Paul
- Public Health Dentistry, Sharad Pawar Dental College and Hospital, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Amit Reche
- Public Health Dentistry, Sharad Pawar Dental College and Hospital, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
11
|
Wang X, Liu D, Luo J, Kong D, Zhang Y. Exploring the Role of Enhancer-Mediated Transcriptional Regulation in Precision Biology. Int J Mol Sci 2023; 24:10843. [PMID: 37446021 PMCID: PMC10342031 DOI: 10.3390/ijms241310843] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/18/2023] [Accepted: 06/25/2023] [Indexed: 07/15/2023] Open
Abstract
The emergence of precision biology has been driven by the development of advanced technologies and techniques in high-resolution biological research systems. Enhancer-mediated transcriptional regulation, a complex network of gene expression and regulation in eukaryotes, has attracted significant attention as a promising avenue for investigating the underlying mechanisms of biological processes and diseases. To address biological problems with precision, large amounts of data, functional information, and research on the mechanisms of action of biological molecules is required to address biological problems with precision. Enhancers, including typical enhancers and super enhancers, play a crucial role in gene expression and regulation within this network. The identification and targeting of disease-associated enhancers hold the potential to advance precision medicine. In this review, we present the concepts, progress, importance, and challenges in precision biology, transcription regulation, and enhancers. Furthermore, we propose a model of transcriptional regulation for multi-enhancers and provide examples of their mechanisms in mammalian cells, thereby enhancing our understanding of how enhancers achieve precise regulation of gene expression in life processes. Precision biology holds promise in providing new tools and platforms for discovering insights into gene expression and disease occurrence, ultimately benefiting individuals and society as a whole.
Collapse
Affiliation(s)
- Xueyan Wang
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China;
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518120, China; (D.L.); (J.L.); (D.K.)
| | - Danli Liu
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518120, China; (D.L.); (J.L.); (D.K.)
| | - Jing Luo
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518120, China; (D.L.); (J.L.); (D.K.)
| | - Dashuai Kong
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518120, China; (D.L.); (J.L.); (D.K.)
| | - Yubo Zhang
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China;
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518120, China; (D.L.); (J.L.); (D.K.)
| |
Collapse
|
12
|
Chen X, Ding Q, Lin T, Sun Y, Huang Z, Li Y, Hong W, Chen X, Wang D, Qiu S. An immune-related prognostic model predicts neoplasm-immunity interactions for metastatic nasopharyngeal carcinoma. Front Immunol 2023; 14:1109503. [PMID: 37063853 PMCID: PMC10102363 DOI: 10.3389/fimmu.2023.1109503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 03/21/2023] [Indexed: 04/03/2023] Open
Abstract
BackgroundThe prognosis of nasopharyngeal carcinoma (NPC) has been recognized to improve immensely owing to radiotherapy combined with chemotherapy. However, patients with metastatic NPC have a poor prognosis. Immunotherapy has dramatically prolonged the survival of patients with NPC. Hence, further research on immune-related biomarkers is imperative to establish the prognosis of metastatic NPC.Methods10 NPC RNA expression profiles were generated from patients with or without distant metastasis after chemoradiotherapy from the Fujian Cancer Hospital. The differential immune-related genes were identified and validated by immunohistochemistry analysis. The method of least absolute shrinkage and selection operator (LASSO)was used to further establish the immune-related prognostic model in an external GEO database (GSE102349, n=88). The immune microenvironment and signal pathways were evaluated in multiple dimensions at the transcriptome and single-cell levels.Results1328 differential genes were identified, out of which 520 were upregulated and 808 were downregulated. Notably, most of the immune genes and pathways were down-regulated in the metastasis group. A prognostic immune model involving nine hub genes. Patients in low-risk group were characterized by survival advantage, hot immune phenotype and benefit from immunotherapy. Compared with immune cells, malignant cell exhibited the most active levels of risk score by ssGSEA. Accordingly, intercellular communications including LT, CD70, CD40 and SPP1, and the like, between high-risk and low-risk were explored by the R package “Cellchat”.ConclusionWe have constructed a model based on immunity of metastatic NPC and determined its prognostic value. The model identified the level of immune cell infiltration, cell-cell communication, along with potential immunotherapy for metastatic NPC.
Collapse
Affiliation(s)
- Xiaochuan Chen
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Qin Ding
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Ting Lin
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Yingming Sun
- Department of Radiation and Medical Oncology, Affiliated Sanming First Hospital of Fujian Medical University, Sanming, China
| | - Zongwei Huang
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Ying Li
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Wenquan Hong
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Xin Chen
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Desheng Wang
- Department of Otolaryngology, Fujian Medical University Union Hospital, Fuzhou, China
- *Correspondence: Sufang Qiu, ; Desheng Wang,
| | - Sufang Qiu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- *Correspondence: Sufang Qiu, ; Desheng Wang,
| |
Collapse
|
13
|
Comprehensive pan-cancer analysis identifies centromere associated protein E as a novel prognostic and immunological biomarker in human tumors. Biochim Biophys Acta Gen Subj 2023; 1867:130346. [PMID: 36931353 DOI: 10.1016/j.bbagen.2023.130346] [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: 09/14/2022] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023]
Abstract
Centromere-associated protein E (CENP-E), a core component of the kinetochore, mediates chromosome congression and spindle microtubule capture during mitosis. Partial experimental evidence has illustrated the carcinogenic effects of CENPE in tumors, but the corresponding pan-cancer analysis of CENPE still lacking. Based on public databases, including the Cancer Genome Atlas (TCGA), Genotype-Tissue Expression (GTEx), and Human Protein Atlas (HPA), we take an array of bioinformatics methods to investigate the potential oncogenic roles of CENPE. Then, we validated CENPE, cell cycle-related proteins, and immune checkpoint molecule findings expression in clinical colon cancer samples by western blot. Our results showed that CENPE was up-regulated in almost all tumors, and the expression level of CENPE was associated with worse overall survival (OS) and disease-specific survival (DSS) in patients. The strong relationship between CENPE with gene mutation and MMR has also been validated. Moreover, CENPE gene expression was positively correlated with immune checkpoint molecular, and reversely correlated with infiltration levels of most immune cells. In the human colon cancer tissues, the expression of CENPE, cell cycle-related proteins, and immune checkpoint molecules were significantly higher than in the adjacent normal tissues. Our results indicated that CENPE can function as an oncogene in various cancers, and may be regarded as a promising prognostic and diagnostic biomarker in cancer treatment.
Collapse
|
14
|
Rodríguez Ruiz N, Abd Own S, Ekström Smedby K, Eloranta S, Koch S, Wästerlid T, Krstic A, Boman M. Data-driven support to decision-making in molecular tumour boards for lymphoma: A design science approach. Front Oncol 2022; 12:984021. [PMID: 36457495 PMCID: PMC9705761 DOI: 10.3389/fonc.2022.984021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/03/2022] [Indexed: 09/10/2024] Open
Abstract
Background The increasing amount of molecular data and knowledge about genomic alterations from next-generation sequencing processes together allow for a greater understanding of individual patients, thereby advancing precision medicine. Molecular tumour boards feature multidisciplinary teams of clinical experts who meet to discuss complex individual cancer cases. Preparing the meetings is a manual and time-consuming process. Purpose To design a clinical decision support system to improve the multimodal data interpretation in molecular tumour board meetings for lymphoma patients at Karolinska University Hospital, Stockholm, Sweden. We investigated user needs and system requirements, explored the employment of artificial intelligence, and evaluated the proposed design with primary stakeholders. Methods Design science methodology was used to form and evaluate the proposed artefact. Requirements elicitation was done through a scoping review followed by five semi-structured interviews. We used UML Use Case diagrams to model user interaction and UML Activity diagrams to inform the proposed flow of control in the system. Additionally, we modelled the current and future workflow for MTB meetings and its proposed machine learning pipeline. Interactive sessions with end-users validated the initial requirements based on a fictive patient scenario which helped further refine the system. Results The analysis showed that an interactive secure Web-based information system supporting the preparation of the meeting, multidisciplinary discussions, and clinical decision-making could address the identified requirements. Integrating artificial intelligence via continual learning and multimodal data fusion were identified as crucial elements that could provide accurate diagnosis and treatment recommendations. Impact Our work is of methodological importance in that using artificial intelligence for molecular tumour boards is novel. We provide a consolidated proof-of-concept system that could support the end-to-end clinical decision-making process and positively and immediately impact patients. Conclusion Augmenting a digital decision support system for molecular tumour boards with retrospective patient material is promising. This generates realistic and constructive material for human learning, and also digital data for continual learning by data-driven artificial intelligence approaches. The latter makes the future system adaptable to human bias, improving adequacy and decision quality over time and over tasks, while building and maintaining a digital log.
Collapse
Affiliation(s)
- Núria Rodríguez Ruiz
- Department of Learning, Informatics, Management and Ethics (LIME), Health Informatics Centre, Karolinska Institutet, Stockholm, Sweden
| | - Sulaf Abd Own
- Department of Medicine Solna, Clinical Epidemiology Division, Karolinska Institutet, Stockholm, Sweden
- Department of Laboratory Medicine, Division of Pathology, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Karin Ekström Smedby
- Department of Medicine Solna, Clinical Epidemiology Division, Karolinska Institutet, Stockholm, Sweden
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Sandra Eloranta
- Department of Medicine Solna, Clinical Epidemiology Division, Karolinska Institutet, Stockholm, Sweden
| | - Sabine Koch
- Department of Learning, Informatics, Management and Ethics (LIME), Health Informatics Centre, Karolinska Institutet, Stockholm, Sweden
| | - Tove Wästerlid
- Department of Medicine Solna, Clinical Epidemiology Division, Karolinska Institutet, Stockholm, Sweden
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Aleksandra Krstic
- Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Magnus Boman
- Department of Learning, Informatics, Management and Ethics (LIME), Health Informatics Centre, Karolinska Institutet, Stockholm, Sweden
- School of Electrical Engineering and Computer Science (EECS)/Software and Computer Systems, KTH Royal Institute of Technology, Stockholm, Sweden
| |
Collapse
|
15
|
Alsaleh L, Li C, Couetil JL, Ye Z, Huang K, Zhang J, Chen C, Johnson TS. Spatial Transcriptomic Analysis Reveals Associations between Genes and Cellular Topology in Breast and Prostate Cancers. Cancers (Basel) 2022; 14:4856. [PMID: 36230778 PMCID: PMC9562681 DOI: 10.3390/cancers14194856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/26/2022] [Accepted: 09/28/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Cancer is the leading cause of death worldwide with breast and prostate cancer the most common among women and men, respectively. Gene expression and image features are independently prognostic of patient survival; but until the advent of spatial transcriptomics (ST), it was not possible to determine how gene expression of cells was tied to their spatial relationships (i.e., topology). METHODS We identify topology-associated genes (TAGs) that correlate with 700 image topological features (ITFs) in breast and prostate cancer ST samples. Genes and image topological features are independently clustered and correlated with each other. Themes among genes correlated with ITFs are investigated by functional enrichment analysis. RESULTS Overall, topology-associated genes (TAG) corresponding to extracellular matrix (ECM) and Collagen Type I Trimer gene ontology terms are common to both prostate and breast cancer. In breast cancer specifically, we identify the ZAG-PIP Complex as a TAG. In prostate cancer, we identify distinct TAGs that are enriched for GI dysmotility and the IgA immunoglobulin complex. We identified TAGs in every ST slide regardless of cancer type. CONCLUSIONS These TAGs are enriched for ontology terms, illustrating the biological relevance to our image topology features and their potential utility in diagnostic and prognostic models.
Collapse
Affiliation(s)
- Lujain Alsaleh
- Department of Biostatistics and Health Data Science, Indiana University, Indianapolis, IN 46202, USA
| | - Chen Li
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, NY 11794, USA
| | - Justin L. Couetil
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, IN 46202, USA
| | - Ze Ye
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, NY 11794, USA
| | - Kun Huang
- Department of Biostatistics and Health Data Science, Indiana University, Indianapolis, IN 46202, USA
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, IN 46202, USA
- Regenstrief Institute, Indiana University, Indianapolis, IN 46202, USA
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN 46202, USA
| | - Jie Zhang
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, IN 46202, USA
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN 46202, USA
| | - Chao Chen
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, NY 11794, USA
| | - Travis S. Johnson
- Department of Biostatistics and Health Data Science, Indiana University, Indianapolis, IN 46202, USA
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN 46202, USA
- Indiana Biosciences Research Institute, Indianapolis, IN 46202, USA
| |
Collapse
|
16
|
Dotolo S, Esposito Abate R, Roma C, Guido D, Preziosi A, Tropea B, Palluzzi F, Giacò L, Normanno N. Bioinformatics: From NGS Data to Biological Complexity in Variant Detection and Oncological Clinical Practice. Biomedicines 2022; 10:biomedicines10092074. [PMID: 36140175 PMCID: PMC9495893 DOI: 10.3390/biomedicines10092074] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/12/2022] [Accepted: 08/22/2022] [Indexed: 11/22/2022] Open
Abstract
The use of next-generation sequencing (NGS) techniques for variant detection has become increasingly important in clinical research and in clinical practice in oncology. Many cancer patients are currently being treated in clinical practice or in clinical trials with drugs directed against specific genomic alterations. In this scenario, the development of reliable and reproducible bioinformatics tools is essential to derive information on the molecular characteristics of each patient’s tumor from the NGS data. The development of bioinformatics pipelines based on the use of machine learning and statistical methods is even more relevant for the determination of complex biomarkers. In this review, we describe some important technologies, computational algorithms and models that can be applied to NGS data from Whole Genome to Targeted Sequencing, to address the problem of finding complex cancer-associated biomarkers. In addition, we explore the future perspectives and challenges faced by bioinformatics for precision medicine both at a molecular and clinical level, with a focus on an emerging complex biomarker such as homologous recombination deficiency (HRD).
Collapse
Affiliation(s)
- Serena Dotolo
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, 80131 Naples, Italy
| | - Riziero Esposito Abate
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, 80131 Naples, Italy
| | - Cristin Roma
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, 80131 Naples, Italy
| | - Davide Guido
- Bioinformatics Research Core Facility, Gemelli Science and Technology Park (GSTeP), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo A. Gemelli, 8, 00168 Rome, Italy
| | - Alessia Preziosi
- Bioinformatics Research Core Facility, Gemelli Science and Technology Park (GSTeP), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo A. Gemelli, 8, 00168 Rome, Italy
| | - Beatrice Tropea
- Bioinformatics Research Core Facility, Gemelli Science and Technology Park (GSTeP), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo A. Gemelli, 8, 00168 Rome, Italy
| | - Fernando Palluzzi
- Bioinformatics Research Core Facility, Gemelli Science and Technology Park (GSTeP), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo A. Gemelli, 8, 00168 Rome, Italy
| | - Luciano Giacò
- Bioinformatics Research Core Facility, Gemelli Science and Technology Park (GSTeP), Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo A. Gemelli, 8, 00168 Rome, Italy
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori—IRCCS—Fondazione G. Pascale, 80131 Naples, Italy
- Correspondence:
| |
Collapse
|
17
|
Angaroni F, Guidi A, Ascolani G, d'Onofrio A, Antoniotti M, Graudenzi A. J-SPACE: a Julia package for the simulation of spatial models of cancer evolution and of sequencing experiments. BMC Bioinformatics 2022; 23:269. [PMID: 35804300 PMCID: PMC9270769 DOI: 10.1186/s12859-022-04779-8] [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: 02/23/2022] [Accepted: 06/09/2022] [Indexed: 11/15/2022] Open
Abstract
Background The combined effects of biological variability and measurement-related errors on cancer sequencing data remain largely unexplored. However, the spatio-temporal simulation of multi-cellular systems provides a powerful instrument to address this issue. In particular, efficient algorithmic frameworks are needed to overcome the harsh trade-off between scalability and expressivity, so to allow one to simulate both realistic cancer evolution scenarios and the related sequencing experiments, which can then be used to benchmark downstream bioinformatics methods. Result We introduce a Julia package for SPAtial Cancer Evolution (J-SPACE), which allows one to model and simulate a broad set of experimental scenarios, phenomenological rules and sequencing settings.Specifically, J-SPACE simulates the spatial dynamics of cells as a continuous-time multi-type birth-death stochastic process on a arbitrary graph, employing different rules of interaction and an optimised Gillespie algorithm. The evolutionary dynamics of genomic alterations (single-nucleotide variants and indels) is simulated either under the Infinite Sites Assumption or several different substitution models, including one based on mutational signatures. After mimicking the spatial sampling of tumour cells, J-SPACE returns the related phylogenetic model, and allows one to generate synthetic reads from several Next-Generation Sequencing (NGS) platforms, via the ART read simulator. The results are finally returned in standard FASTA, FASTQ, SAM, ALN and Newick file formats. Conclusion J-SPACE is designed to efficiently simulate the heterogeneous behaviour of a large number of cancer cells and produces a rich set of outputs. Our framework is useful to investigate the emergent spatial dynamics of cancer subpopulations, as well as to assess the impact of incomplete sampling and of experiment-specific errors. Importantly, the output of J-SPACE is designed to allow the performance assessment of downstream bioinformatics pipelines processing NGS data. J-SPACE is freely available at: https://github.com/BIMIB-DISCo/J-Space.jl.
Collapse
Affiliation(s)
- Fabrizio Angaroni
- Dept. of Informatics, Systems and Communication, Univ. of Milan-Bicocca, Milan, Italy.
| | - Alessandro Guidi
- Dept. of Informatics, Systems and Communication, Univ. of Milan-Bicocca, Milan, Italy
| | - Gianluca Ascolani
- Dept. of Informatics, Systems and Communication, Univ. of Milan-Bicocca, Milan, Italy
| | - Alberto d'Onofrio
- Department of Mathematics and Geosciences, Univ. of Trieste, Trieste, Italy
| | - Marco Antoniotti
- Dept. of Informatics, Systems and Communication, Univ. of Milan-Bicocca, Milan, Italy.,Bicocca Bioinformatics, Biostatistics and Bioimaging Centre (B4), Milan, Italy
| | - Alex Graudenzi
- Dept. of Informatics, Systems and Communication, Univ. of Milan-Bicocca, Milan, Italy.,Bicocca Bioinformatics, Biostatistics and Bioimaging Centre (B4), Milan, Italy.,Inst. of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), Segrate, Italy
| |
Collapse
|
18
|
Hamamoto R, Takasawa K, Machino H, Kobayashi K, Takahashi S, Bolatkan A, Shinkai N, Sakai A, Aoyama R, Yamada M, Asada K, Komatsu M, Okamoto K, Kameoka H, Kaneko S. Application of non-negative matrix factorization in oncology: one approach for establishing precision medicine. Brief Bioinform 2022; 23:6628783. [PMID: 35788277 PMCID: PMC9294421 DOI: 10.1093/bib/bbac246] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/06/2022] [Accepted: 05/25/2022] [Indexed: 12/19/2022] Open
Abstract
The increase in the expectations of artificial intelligence (AI) technology has led to machine learning technology being actively used in the medical field. Non-negative matrix factorization (NMF) is a machine learning technique used for image analysis, speech recognition, and language processing; recently, it is being applied to medical research. Precision medicine, wherein important information is extracted from large-scale medical data to provide optimal medical care for every individual, is considered important in medical policies globally, and the application of machine learning techniques to this end is being handled in several ways. NMF is also introduced differently because of the characteristics of its algorithms. In this review, the importance of NMF in the field of medicine, with a focus on the field of oncology, is described by explaining the mathematical science of NMF and the characteristics of the algorithm, providing examples of how NMF can be used to establish precision medicine, and presenting the challenges of NMF. Finally, the direction regarding the effective use of NMF in the field of oncology is also discussed.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Rina Aoyama
- Showa University Graduate School of Medicine School of Medicine
| | | | - Ken Asada
- RIKEN Center for Advanced Intelligence Project
| | | | | | | | | |
Collapse
|
19
|
Li AH, Li WW, Yu XQ, Zhang DM, Liu YR, Li D. Bioinformatic Analysis and Translational Validation of Psoriasis Candidate Genes for Precision Medicine. Clin Cosmet Investig Dermatol 2022; 15:1447-1458. [PMID: 35924255 PMCID: PMC9343179 DOI: 10.2147/ccid.s378143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/22/2022] [Indexed: 11/23/2022]
Affiliation(s)
- An-Hai Li
- Department of Dermatology, Qingdao Huangdao District Central Hospital, Qingdao, People’s Republic of China
| | - Wen-Wen Li
- Department of Hematology, Qingdao Women and Children’s Hospital, Qingdao, People’s Republic of China
| | - Xiao-Qian Yu
- Department of Dermatology, Qingdao Haici Hospital (Qingdao Traditional Chinese Medicine Hospital), Qingdao, People’s Republic of China
| | - Dai-Ming Zhang
- Department of Pharmacy, Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Yi-Ran Liu
- College of Traditional Chinese Medicine, Weifang Medical College, Weifang, People’s Republic of China
| | - Ding Li
- Department of Dermatology, Qingdao Huangdao District Central Hospital, Qingdao, People’s Republic of China
- Correspondence: Ding Li, Department of Dermatology, Qingdao Huangdao District Central Hospital, Qingdao, People’s Republic of China, Email
| |
Collapse
|
20
|
Sun SJ, Han JD, Liu W, Wu ZY, Zhao X, Yan X, Jiao SC, Fang J. Sequential chemotherapy and icotinib as first-line treatment for advanced epidermal growth factor receptor-mutated non-small cell lung cancer. World J Clin Cases 2022; 10:6069-6081. [PMID: 35949840 PMCID: PMC9254173 DOI: 10.12998/wjcc.v10.i18.6069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 03/13/2022] [Accepted: 04/15/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Icotinib could have potential effect and tolerability when used sequentially with chemotherapy for advanced epidermal growth factor receptor (EGFR)-mutated non-small cell lung cancer (NSCLC).
AIM To evaluate the efficacy and safety of chemotherapy followed by icotinib maintenance therapy as first-line treatment for advanced EGFR-mutated NSCLC.
METHODS This multicenter, open-label, pilot randomized controlled trial enrolled 68 EGFR-mutated stage IIIB/IV NSCLC patients randomized 2:3 to the icotinib alone and chemotherapy + icotinib groups.
RESULTS The median progression-free survival in the icotinib alone and chemotherapy + icotinib groups was 8.0 mo (95%CI: 3.84-11.63) and 13.4 mo (95%CI: 10.18-16.33), respectively (P = 0.0249). No significant differences were found in the curative effect when considering different cycles of chemotherapy or chemotherapy regimen (all P > 0.05).
CONCLUSION A sequential combination of chemotherapy and EGFR-tyrosine kinase inhibitor is feasible for stage IV EGFR-mutated NSCLC patients.
Collapse
Affiliation(s)
- Sheng-Jie Sun
- Department of Medical Oncology, The Fifth Medical Center of General Hospital of Chinese People's Liberation Army, Beijing 100039, China
| | - Jin-Di Han
- Department of Internal Oncology of Chest, Beijing Cancer Hospital, Beijing 100142, China
| | - Wei Liu
- Peking Cancer Hospital Palliative Care Center, Beijing Cancer Hospital, Beijing 100142, China
| | - Zhi-Yong Wu
- Department of Medical Oncology, The Fifth Medical Center of General Hospital of Chinese People's Liberation Army, Beijing 100039, China
| | - Xiao Zhao
- Department of Medical Oncology, The Fifth Medical Center of General Hospital of Chinese People's Liberation Army, Beijing 100039, China
| | - Xiang Yan
- Department of Medical Oncology, The Fifth Medical Center of General Hospital of Chinese People's Liberation Army, Beijing 100039, China
| | - Shun-Chang Jiao
- Department of Oncology, The Fifth Medical Center of General Hospital of Chinese People's Liberation Army, Beijing 100039, China
| | - Jian Fang
- Department of Internal Oncology of Chest, Beijing Cancer Hospital, Beijing 100142, China
| |
Collapse
|
21
|
Wang K, Zhong W, Long Z, Guo Y, Zhong C, Yang T, Wang S, Lai H, Lu J, Zheng P, Mao X. 5-Methylcytosine RNA Methyltransferases-Related Long Non-coding RNA to Develop and Validate Biochemical Recurrence Signature in Prostate Cancer. Front Mol Biosci 2021; 8:775304. [PMID: 34926580 PMCID: PMC8672116 DOI: 10.3389/fmolb.2021.775304] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 11/01/2021] [Indexed: 02/02/2023] Open
Abstract
The effects of 5-methylcytosine in RNA (m5C) in various human cancers have been increasingly studied recently; however, the m5C regulator signature in prostate cancer (PCa) has not been well established yet. In this study, we identified and characterized a series of m5C-related long non-coding RNAs (lncRNAs) in PCa. Univariate Cox regression analysis and least absolute shrinkage and selector operation (LASSO) regression analysis were implemented to construct a m5C-related lncRNA prognostic signature. Consequently, a prognostic m5C-lnc model was established, including 17 lncRNAs: MAFG-AS1, AC012510.1, AC012065.3, AL117332.1, AC132192.2, AP001160.2, AC129510.1, AC084018.2, UBXN10-AS1, AC138956.2, ZNF32-AS2, AC017100.1, AC004943.2, SP2-AS1, Z93930.2, AP001486.2, and LINC01135. The high m5C-lnc score calculated by the model significantly relates to poor biochemical recurrence (BCR)-free survival (p < 0.0001). Receiver operating characteristic (ROC) curves and a decision curve analysis (DCA) further validated the accuracy of the prognostic model. Subsequently, a predictive nomogram combining the prognostic model with clinical features was created, and it exhibited promising predictive efficacy for BCR risk stratification. Next, the competing endogenous RNA (ceRNA) network and lncRNA–protein interaction network were established to explore the potential functions of these 17 lncRNAs mechanically. In addition, functional enrichment analysis revealed that these lncRNAs are involved in many cellular metabolic pathways. Lastly, MAFG-AS1 was selected for experimental validation; it was upregulated in PCa and probably promoted PCa proliferation and invasion in vitro. These results offer some insights into the m5C's effects on PCa and reveal a predictive model with the potential clinical value to improve the prognosis of patients with PCa.
Collapse
Affiliation(s)
- Ke Wang
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Department of Urology, The Hospital of Trade-Business in Hunan Province, Changsha, China
| | - Weibo Zhong
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Zining Long
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yufei Guo
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Chuanfan Zhong
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Taowei Yang
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Shuo Wang
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Houhua Lai
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jianming Lu
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Pengxiang Zheng
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Department of Urology, Fuqing City Hospital Affiliated with Fujian Medical University, Fuzhou, China
| | - Xiangming Mao
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
22
|
Borchert F, Mock A, Tomczak A, Hügel J, Alkarkoukly S, Knurr A, Volckmar AL, Stenzinger A, Schirmacher P, Debus J, Jäger D, Longerich T, Fröhling S, Eils R, Bougatf N, Sax U, Schapranow MP. Knowledge bases and software support for variant interpretation in precision oncology. Brief Bioinform 2021; 22:bbab134. [PMID: 33971666 PMCID: PMC8574624 DOI: 10.1093/bib/bbab134] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 03/10/2021] [Accepted: 03/30/2021] [Indexed: 12/12/2022] Open
Abstract
Precision oncology is a rapidly evolving interdisciplinary medical specialty. Comprehensive cancer panels are becoming increasingly available at pathology departments worldwide, creating the urgent need for scalable cancer variant annotation and molecularly informed treatment recommendations. A wealth of mainly academia-driven knowledge bases calls for software tools supporting the multi-step diagnostic process. We derive a comprehensive list of knowledge bases relevant for variant interpretation by a review of existing literature followed by a survey among medical experts from university hospitals in Germany. In addition, we review cancer variant interpretation tools, which integrate multiple knowledge bases. We categorize the knowledge bases along the diagnostic process in precision oncology and analyze programmatic access options as well as the integration of knowledge bases into software tools. The most commonly used knowledge bases provide good programmatic access options and have been integrated into a range of software tools. For the wider set of knowledge bases, access options vary across different parts of the diagnostic process. Programmatic access is limited for information regarding clinical classifications of variants and for therapy recommendations. The main issue for databases used for biological classification of pathogenic variants and pathway context information is the lack of standardized interfaces. There is no single cancer variant interpretation tool that integrates all identified knowledge bases. Specialized tools are available and need to be further developed for different steps in the diagnostic process.
Collapse
Affiliation(s)
- Florian Borchert
- Digital Health Center, Hasso Plattner Institute (HPI), University of Potsdam, Prof.-Dr.-Helmert-Str. 2-3, 14482 Potsdam, Germany
| | - Andreas Mock
- Department of Translational Medical Oncology (TMO), National Center for Tumor Diseases (NCT) Heidelberg, German Cancer Research Center (DKFZ) Heidelberg, Im Neuenheimer Feld 460, 69120 Heidelberg, Germany
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg University Hospital, Im Neuenheimer Feld 460, 69120 Heidelberg, Germany
| | - Aurelie Tomczak
- Institute of Pathology Heidelberg, Heidelberg University Hospital, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
- Liver Cancer Center Heidelberg, Heidelberg University Hospital, Im Neuenheimer Feld 460, 69120 Heidelberg, Germany
| | - Jonas Hügel
- Department of Medical Informatics, University Medical Center Göttingen, Von-Siebold-Str. 3, 37099 Göttingen, Germany
- Campus Institute Data Science, Göttingen, Germany
| | - Samer Alkarkoukly
- CECAD, Faculty of Medicine and University Hospital Cologne, University of Cologne, Joseph-Stelzmann-Straße 26, 50931 Cologne
| | - Alexander Knurr
- Division of Medical Informatics for Translational Oncology, German Cancer Research Center (DKFZ) Heidelberg, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Anna-Lena Volckmar
- Institute of Pathology Heidelberg, Heidelberg University Hospital, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
| | - Albrecht Stenzinger
- Institute of Pathology Heidelberg, Heidelberg University Hospital, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
| | - Peter Schirmacher
- Institute of Pathology Heidelberg, Heidelberg University Hospital, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
- Liver Cancer Center Heidelberg, Heidelberg University Hospital, Im Neuenheimer Feld 460, 69120 Heidelberg, Germany
| | - Jürgen Debus
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg University Hospital, Im Neuenheimer Feld 460, 69120 Heidelberg, Germany
- Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ) Heidelberg, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Center (HIT), Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 450, 69120 Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Dirk Jäger
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg University Hospital, Im Neuenheimer Feld 460, 69120 Heidelberg, Germany
- Clinical Coorporation Unit Applied Tumor-Immunity, German Cancer Research Center (DKFZ) Heidelberg, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Thomas Longerich
- Institute of Pathology Heidelberg, Heidelberg University Hospital, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
- Liver Cancer Center Heidelberg, Heidelberg University Hospital, Im Neuenheimer Feld 460, 69120 Heidelberg, Germany
| | - Stefan Fröhling
- Department of Translational Medical Oncology (TMO), National Center for Tumor Diseases (NCT) Heidelberg, German Cancer Research Center (DKFZ) Heidelberg, Im Neuenheimer Feld 460, 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
| | - Roland Eils
- Health Data Science Unit, Heidelberg University Hospital, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany
- Center for Digital Health, Berlin Institute of Health and Charité Universitötsmedizin Berlin, Kapelle-Ufer 2, 10117 Berlin, Germany
| | - Nina Bougatf
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg University Hospital, Im Neuenheimer Feld 460, 69120 Heidelberg, Germany
- Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ) Heidelberg, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Center (HIT), Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 450, 69120 Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Ulrich Sax
- Department of Medical Informatics, University Medical Center Göttingen, Von-Siebold-Str. 3, 37099 Göttingen, Germany
- Campus Institute Data Science, Göttingen, Germany
| | - Matthieu-P Schapranow
- Digital Health Center, Hasso Plattner Institute (HPI), University of Potsdam, Prof.-Dr.-Helmert-Str. 2-3, 14482 Potsdam, Germany
| |
Collapse
|
23
|
Jain SR, Sim W, Ng CH, Chin YH, Lim WH, Syn NL, Kamal NHBA, Gupta M, Heong V, Lee XW, Sapari NS, Koh XQ, Isa ZFA, Ho L, O'Hara C, Ulagapan A, Gu SY, Shroff K, Weng RC, Lim JSY, Lim D, Pang B, Ng LK, Wong A, Soo RA, Yong WP, Chee CE, Lee SC, Goh BC, Soong R, Tan DSP. Statistical Process Control Charts for Monitoring Next-Generation Sequencing and Bioinformatics Turnaround in Precision Medicine Initiatives. Front Oncol 2021; 11:736265. [PMID: 34631570 PMCID: PMC8498582 DOI: 10.3389/fonc.2021.736265] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 09/08/2021] [Indexed: 02/04/2023] Open
Abstract
Purpose Precision oncology, such as next generation sequencing (NGS) molecular analysis and bioinformatics are used to guide targeted therapies. The laboratory turnaround time (TAT) is a key performance indicator of laboratory performance. This study aims to formally apply statistical process control (SPC) methods such as CUSUM and EWMA to a precision medicine programme to analyze the learning curves of NGS and bioinformatics processes. Patients and Methods Trends in NGS and bioinformatics TAT were analyzed using simple regression models with TAT as the dependent variable and chronologically-ordered case number as the independent variable. The M-estimator "robust" regression and negative binomial regression were chosen to serve as sensitivity analyses to each other. Next, two popular statistical process control (SPC) approaches which are CUSUM and EWMA were utilized and the CUSUM log-likelihood ratio (LLR) charts were also generated. All statistical analyses were done in Stata version 16.0 (StataCorp), and nominal P < 0.05 was considered to be statistically significant. Results A total of 365 patients underwent successful molecular profiling. Both the robust linear model and negative binomial model showed statistically significant reductions in TAT with accumulating experience. The EWMA and CUSUM charts of overall TAT largely corresponded except that the EWMA chart consistently decreased while the CUSUM analyses indicated improvement only after a nadir at the 82nd case. CUSUM analysis found that the bioinformatics team took a lower number of cases (54 cases) to overcome the learning curve compared to the NGS team (85 cases). Conclusion As NGS and bioinformatics lead precision oncology into the forefront of cancer management, characterizing the TAT of NGS and bioinformatics processes improves the timeliness of data output by potentially spotlighting problems early for rectification, thereby improving care delivery.
Collapse
Affiliation(s)
- Sneha Rajiv Jain
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wilson Sim
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Cheng Han Ng
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yip Han Chin
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wen Hui Lim
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Nicholas L Syn
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | | | - Mehek Gupta
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Valerie Heong
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Xiao Wen Lee
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Nur Sabrina Sapari
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Xue Qing Koh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Zul Fazreen Adam Isa
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Lucius Ho
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Caitlin O'Hara
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Arvindh Ulagapan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Shi Yu Gu
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kashyap Shroff
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Rei Chern Weng
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Joey S Y Lim
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Diana Lim
- Department of Pathology, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore.,Department of Pathology, National University Hospital, National University Health System, Singapore, Singapore
| | - Brendan Pang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,Department of Pathology, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore.,Department of Pathology, National University Hospital, National University Health System, Singapore, Singapore
| | - Lai Kuan Ng
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Andrea Wong
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Ross Andrew Soo
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Wei Peng Yong
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Cheng Ean Chee
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Soo-Chin Lee
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Boon-Cher Goh
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore
| | - Richie Soong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,Department of Pathology, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore.,Department of Medicine, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore.,Pascific Laboratories, Singapore, Singapore
| | - David S P Tan
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,Department of Medicine, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore
| |
Collapse
|
24
|
Piyawajanusorn C, Nguyen LC, Ghislat G, Ballester PJ. A gentle introduction to understanding preclinical data for cancer pharmaco-omic modeling. Brief Bioinform 2021; 22:6343527. [PMID: 34368843 DOI: 10.1093/bib/bbab312] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/25/2021] [Accepted: 07/20/2021] [Indexed: 12/16/2022] Open
Abstract
A central goal of precision oncology is to administer an optimal drug treatment to each cancer patient. A common preclinical approach to tackle this problem has been to characterize the tumors of patients at the molecular and drug response levels, and employ the resulting datasets for predictive in silico modeling (mostly using machine learning). Understanding how and why the different variants of these datasets are generated is an important component of this process. This review focuses on providing such introduction aimed at scientists with little previous exposure to this research area.
Collapse
Affiliation(s)
- Chayanit Piyawajanusorn
- Cancer Research Center of Marseille, INSERM U1068, F-13009 Marseille, France.,Institut Paoli-Calmettes, F-13009 Marseille, France.,Aix-Marseille Université, F-13284 Marseille, France.,CNRS UMR7258, F-13009 Marseille, France.,Faculty of Medicine and Public Health, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Linh C Nguyen
- Cancer Research Center of Marseille, INSERM U1068, F-13009 Marseille, France.,Institut Paoli-Calmettes, F-13009 Marseille, France.,Aix-Marseille Université, F-13284 Marseille, France.,CNRS UMR7258, F-13009 Marseille, France.,Department of Life Sciences, University of Science and Technology of Hanoi, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | - Ghita Ghislat
- U1104, CNRS UMR7280, Centre d'Immunologie de Marseille-Luminy, Inserm, Marseille, France
| | - Pedro J Ballester
- Cancer Research Center of Marseille, INSERM U1068, F-13009 Marseille, France.,Institut Paoli-Calmettes, F-13009 Marseille, France.,Aix-Marseille Université, F-13284 Marseille, France.,CNRS UMR7258, F-13009 Marseille, France
| |
Collapse
|
25
|
Rafique R, Islam SR, Kazi JU. Machine learning in the prediction of cancer therapy. Comput Struct Biotechnol J 2021; 19:4003-4017. [PMID: 34377366 PMCID: PMC8321893 DOI: 10.1016/j.csbj.2021.07.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 12/15/2022] Open
Abstract
Resistance to therapy remains a major cause of cancer treatment failures, resulting in many cancer-related deaths. Resistance can occur at any time during the treatment, even at the beginning. The current treatment plan is dependent mainly on cancer subtypes and the presence of genetic mutations. Evidently, the presence of a genetic mutation does not always predict the therapeutic response and can vary for different cancer subtypes. Therefore, there is an unmet need for predictive models to match a cancer patient with a specific drug or drug combination. Recent advancements in predictive models using artificial intelligence have shown great promise in preclinical settings. However, despite massive improvements in computational power, building clinically useable models remains challenging due to a lack of clinically meaningful pharmacogenomic data. In this review, we provide an overview of recent advancements in therapeutic response prediction using machine learning, which is the most widely used branch of artificial intelligence. We describe the basics of machine learning algorithms, illustrate their use, and highlight the current challenges in therapy response prediction for clinical practice.
Collapse
Affiliation(s)
| | - S.M. Riazul Islam
- Department of Computer Science and Engineering, Sejong University, Seoul, South Korea
| | - Julhash U. Kazi
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Corresponding author at: Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon village Building 404:C3, Scheelevägen 8, 22363 Lund, Sweden.
| |
Collapse
|
26
|
Laßmann S, Hummel M. [Molecular tumor boards - insights and perspectives]. DER PATHOLOGE 2021; 42:357-362. [PMID: 34170386 DOI: 10.1007/s00292-021-00955-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/10/2021] [Indexed: 10/21/2022]
Abstract
The rapid development of molecular technologies and targeted therapies has fostered the implementation of specialized tumor conferences, known as molecular tumor boards (MTBs). MTBs become particularly important when treatment recommendations are needed based on molecular alterations beyond the approved targeted therapies. While an MTB's goals are based on individualized diagnostics and therapies of tumor patients using innovative technologies and biomarkers, the procedures of MTBs are still quite heterogeneous. This applies to the primary inclusion criteria for tumor patients, the composition of MTBs, the applied diagnostic tests and their assessment and reporting, the evaluation of their clinical value and implementation in a therapeutic strategy, and the associated quality assurance measurements as well as knowledge-gaining, economical, legal, and ethical aspects.This article provides an overview of the spectrum of MTBs, their challenges, and the potential for individualized cancer medicine.
Collapse
Affiliation(s)
- Silke Laßmann
- Institut für Klinische Pathologie, Universitätsklinikum Freiburg, Breisacher Str. 115A, 79106, Freiburg, Deutschland.
| | - Michael Hummel
- Institut für Pathologie, Charité - Universitätsmedizin Berlin, Berlin, Deutschland
| |
Collapse
|
27
|
Bagaev A, Kotlov N, Nomie K, Svekolkin V, Gafurov A, Isaeva O, Osokin N, Kozlov I, Frenkel F, Gancharova O, Almog N, Tsiper M, Ataullakhanov R, Fowler N. Conserved pan-cancer microenvironment subtypes predict response to immunotherapy. Cancer Cell 2021; 39:845-865.e7. [PMID: 34019806 DOI: 10.1016/j.ccell.2021.04.014] [Citation(s) in RCA: 578] [Impact Index Per Article: 144.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 09/14/2020] [Accepted: 04/23/2021] [Indexed: 12/18/2022]
Abstract
The clinical use of molecular targeted therapy is rapidly evolving but has primarily focused on genomic alterations. Transcriptomic analysis offers an opportunity to dissect the complexity of tumors, including the tumor microenvironment (TME), a crucial mediator of cancer progression and therapeutic outcome. TME classification by transcriptomic analysis of >10,000 cancer patients identifies four distinct TME subtypes conserved across 20 different cancers. The TME subtypes correlate with patient response to immunotherapy in multiple cancers, with patients possessing immune-favorable TME subtypes benefiting the most from immunotherapy. Thus, the TME subtypes act as a generalized immunotherapy biomarker across many cancer types due to the inclusion of malignant and microenvironment components. A visual tool integrating transcriptomic and genomic data provides a global tumor portrait, describing the tumor framework, mutational load, immune composition, anti-tumor immunity, and immunosuppressive escape mechanisms. Integrative analyses plus visualization may aid in biomarker discovery and the personalization of therapeutic regimens.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Nathan Fowler
- BostonGene, Waltham, MA 02453, USA; Department of Lymphoma and Myeloma, Unit 0429, the University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.
| |
Collapse
|
28
|
Özdoğan M, Papadopoulou E, Tsoulos N, Tsantikidi A, Mariatou VM, Tsaousis G, Kapeni E, Bourkoula E, Fotiou D, Kapetsis G, Boukovinas I, Touroutoglou N, Fassas A, Adamidis A, Kosmidis P, Trafalis D, Galani E, Lypas G, Orhan B, Tansan S, Özatlı T, Kırca O, Çakır O, Nasioulas G. Comprehensive tumor molecular profile analysis in clinical practice. BMC Med Genomics 2021; 14:105. [PMID: 33853586 PMCID: PMC8045191 DOI: 10.1186/s12920-021-00952-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 03/18/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Tumor molecular profile analysis by Next Generation Sequencing technology is currently widely applied in clinical practice and has enabled the detection of predictive biomarkers of response to targeted treatment. In parallel with targeted therapies, immunotherapies are also evolving, revolutionizing cancer therapy, with Programmed Death-ligand 1 (PD-L1), Microsatellite instability (MSI), and Tumor Mutational Burden (TMB) analysis being the biomarkers employed most commonly. METHODS In the present study, tumor molecular profile analysis was performed using a 161 gene NGS panel, containing the majority of clinically significant genes for cancer treatment selection. A variety of tumor types have been analyzed, including aggressive and hard to treat cancers such as pancreatic cancer. Besides, the clinical utility of immunotherapy biomarkers (TMB, MSI, PD-L1), was also studied. RESULTS Molecular profile analysis was conducted in 610 cancer patients, while in 393 of them a at least one biomarker for immunotherapy response was requested. An actionable alteration was detected in 77.87% of the patients. 54.75% of them received information related to on-label or off-label treatment (Tiers 1A.1, 1A.2, 2B, and 2C.1) and 21.31% received a variant that could be used for clinical trial inclusion. The addition to immunotherapy biomarker to targeted biomarkers' analysis in 191 cases increased the number of patients with an on-label treatment recommendation by 22.92%, while an option for on-label or off-label treatment was provided in 71.35% of the cases. CONCLUSIONS Tumor molecular profile analysis using NGS is a first-tier method for a variety of tumor types and provides important information for decision making in the treatment of cancer patients. Importantly, simultaneous analysis for targeted therapy and immunotherapy biomarkers could lead to better tumor characterization and offer actionable information in the majority of patients. Furthermore, our data suggest that one in two patients may be eligible for on-label ICI treatment based on biomarker analysis. However, appropriate interpretation of results from such analysis is essential for implementation in clinical practice and accurate refinement of treatment strategy.
Collapse
Affiliation(s)
- Mustafa Özdoğan
- Division of Medical Oncology, Memorial Hospital, Antalya, Turkey
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Eleni Galani
- Second Department of Medical Oncology, "Metropolitan" Hospital, Piraeus, Greece
| | - George Lypas
- Department of Genetic Oncology/Medical Oncology, Hygeia Hospital, Athens, Greece
| | - Bülent Orhan
- Department of Medical Oncology, Ceylan International Hospital, Bursa, Turkey
| | | | | | - Onder Kırca
- Division of Medical Oncology, Memorial Hospital, Antalya, Turkey
| | - Okan Çakır
- Applied Health Sciences, Edinburgh Napier University, Edinburgh, EH11 4BN, Scotland, UK
| | | |
Collapse
|
29
|
Shi Y, Chang D, Li W, Zhao F, Ren X, Hou B. Identification of core genes and clinical outcomes in tumors originated from endoderm (gastric cancer and lung carcinoma) via bioinformatics analysis. Medicine (Baltimore) 2021; 100:e25154. [PMID: 33761685 PMCID: PMC10545272 DOI: 10.1097/md.0000000000025154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 11/30/2020] [Accepted: 02/23/2021] [Indexed: 12/24/2022] Open
Abstract
ABSTRACT During last decade, bioinformatics analysis has provided an effective way to study the relationship between various genes and biological processes. In this study, we aimed to identify potential core candidate genes and underlying mechanisms of progression of lung and gastric carcinomas which both originated from endoderm. The expression profiles, GSE54129 (gastric carcinoma) and GSE27262 (lung carcinoma), were collected from GEO database. One hundred eleven patients with gastric carcinoma and 21 health people were included in this research. Meanwhile, there were 25 lung carcinoma patients. Then, 75 differentially expressed genes were selected via GEO2R online tool and Venn software, including 31 up-regulated genes and 44 down-regulated genes. Next, we used Database for Annotation, Visualization, and Integrated Discovery and Metascpe software to analyze Kyoto Encyclopedia of Gene and Genome pathway and gene ontology. Furthermore, Cytoscape software and MCODE App were performed to construct complex of these differentially expressed genes . Twenty core genes were identified, which mainly enriched in extracellular matrix-receptor interaction, focal adhesion, and PI3K-Akt pathway (P < .01). Finally, the significant difference of gene expression between cancer tissues and normal tissues in both lung and gastric carcinomas was examined by Gene Expression Profiling Interactive Analysis database. Twelve candidate genes with positive statistical significance (P < .01), COMP CTHRC1 COL1A1 SPP1 COL11A1 COL10A1 CXCL13 CLDN3 CLDN1 matrix metalloproteinases 7 ADAM12 PLAU, were picked out to further analysis. The Kaplan-Meier plotter website was applied to examine relationship among these genes and clinical outcomes. We found 4 genes (ADAM12, SPP1, COL1A1, COL11A1) were significantly associated with poor prognosis in both lung and gastric carcinoma patients (P < .05). In conclusion, these candidate genes may be potential therapeutic targets for cancer treatment.
Collapse
Affiliation(s)
- Yewen Shi
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Affiliated Hospital of Xi’an Jiaotong University
| | - Dongmin Chang
- Department of Surgical Oncology, the First Affiliated Hospital of Xi’an Jiaotong University
| | - Wenhan Li
- Department of Surgical Oncology, Shaanxi Provincial People's Hospital
- The Third Affiliated Hospital, the School of Medicine Xi’an Jiaotong University
| | - FengYu Zhao
- Department of Surgical Oncology, the First Affiliated Hospital of Xi’an Jiaotong University
| | - Xiaoyong Ren
- Department of Otorhinolaryngology Head and Neck Surgery, the Second Affiliated Hospital of Xi’an Jiaotong University
| | - Bin Hou
- The Third Affiliated Hospital, the School of Medicine Xi’an Jiaotong University
- Department of Thoracic Surgery, Shaanxi Provincial People's Hospital, Xi’an, Shaanxi, China
| |
Collapse
|
30
|
Aikemu B, Xue P, Hong H, Jia H, Wang C, Li S, Huang L, Ding X, Zhang H, Cai G, Lu A, Xie L, Li H, Zheng M, Sun J. Artificial Intelligence in Decision-Making for Colorectal Cancer Treatment Strategy: An Observational Study of Implementing Watson for Oncology in a 250-Case Cohort. Front Oncol 2021; 10:594182. [PMID: 33628729 PMCID: PMC7899045 DOI: 10.3389/fonc.2020.594182] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 12/21/2020] [Indexed: 12/15/2022] Open
Abstract
Background Personalized and novel evidence-based clinical treatment strategy consulting for colorectal cancer has been available through various artificial intelligence (AI) supporting systems such as Watson for Oncology (WFO) from IBM. However, the potential effects of this supporting tool in cancer care have not been thoroughly explored in real-world studies. This research aims to investigate the concordance between treatment recommendations for colorectal cancer patients made by WFO and a multidisciplinary team (MDT) at a major comprehensive gastrointestinal cancer center. Methods In this prospective study, both WFO and the blinded MDT's treatment recommendations were provided concurrently for enrolled colorectal cancers of stages II to IV between March 2017 and January 2018 at Shanghai Minimally Invasive Surgery Center. Concordance was achieved if the cancer team's decisions were listed in the "recommended" or "for consideration" classification in WFO. A review was carried out after 100 cases for all non-concordant patients to explain the inconsistency, and corresponding feedback was given to WFO's database. The concordance of the subsequent cases was analyzed to evaluate both the performance and learning ability of WFO. Results Overall, 250 patients met the inclusion criteria and were recruited in the study. Eighty-one were diagnosed with colon cancer and 189 with rectal cancer. The concordances for colon cancer, rectal cancer, or overall were all 91%. The overall rates were 83, 94, and 88% in subgroups of stages II, III, and IV. When categorized by treatment strategy, concordances were 97, 93, 89, 87, and 100% for neoadjuvant, surgery, adjuvant, first line, and second line treatment groups, respectively. After analyzing the main factors causing discordance, relative updates were made in the database accordingly, which led to the concordance curve rising in most groups compared with the initial rates. Conclusion Clinical recommendations made by WFO and the cancer team were highly matched for colorectal cancer. Patient age, cancer stage, and the consideration of previous therapy details had a significant influence on concordance. Addressing these perspectives will facilitate the use of the cancer decision-support systems to help oncologists achieve the promise of precision medicine.
Collapse
Affiliation(s)
- Batuer Aikemu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pei Xue
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hiju Hong
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongtao Jia
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chenxing Wang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuchun Li
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling Huang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyi Ding
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huan Zhang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Gang Cai
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Aiguo Lu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Xie
- Clinical Research Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hao Li
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Minhua Zheng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Sun
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
31
|
Liang R, Chen W, Chen XY, Fan HN, Zhang J, Zhu JS. Dihydroartemisinin inhibits the tumorigenesis and invasion of gastric cancer by regulating STAT1/KDR/MMP9 and P53/BCL2L1/CASP3/7 pathways. Pathol Res Pract 2021; 218:153318. [PMID: 33370709 DOI: 10.1016/j.prp.2020.153318] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/28/2020] [Accepted: 12/08/2020] [Indexed: 12/20/2022]
Abstract
Dihydroartemisinin (DHA), an effective antimalarial drug, has been widely investigated as an anti-tumor agent. Although previous studies have indicated the potential therapeutic effects of DHA on multiple malignancies, its detailed molecular mechanisms in gastric cancer (GC) are still undocumented. In the present study, we applied network pharmacology and bioinformatics (gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses) to obtain the collective targets of DHA and GC and analyzed their involvement in constructing a protein-protein interaction (PPI) network. The top 10% hub targets in this network were identified, and TCGA database was utilized for the single gene analysis of their correlation with the prognosis of GC. CCK8, EdU, Transwell, and flow cytometry analyses were conducted, and subcutaneous xenograft tumor models were constructed to assess the effects of DHA on the tumorigenesis and invasion of GC. Furthermore, the targets of DHA were verified by molecular docking, quantitative real-time PCR (qPCR) and western blot analyses in GC cells. The results indicated that the common targets of DHA and GC were enriched in multiple cancer-related pathways including KDR, STAT1 and apoptosis signaling pathways, where the core genes included KDR, MMP9, STAT1, TP53, CASP3/7 and BCL2L1. The lowered expression of KDR and increased expression of TP53 and CASP7 harbored a favorable survival for patients with GC patients. CASP7 showed a positive correlation with CASP3 but a negative correlation with KDR and could be regarded as an independent protective factor for overall survival in GC. Moreover, DHA treatment induced cell apoptosis and suppressed the cell proliferation, DNA synthesis, cycle progression and invasive capabilities both in vitro and in vivo. DHA also upregulated p53, CASP3, and cleaved-CASP3 and downregulated BCL2L1, MMP9, KDR, p-KDR, STAT1 and p-STAT1 in GC cell lines. In conclusion, DHA could suppress the tumorigenesis and invasion of GC by regulating STAT1/KDR/MMP9 and p53/BCL2L1/CASP3/7 pathways. Our findings might provide a novel approach for the treatment of GC.
Collapse
Affiliation(s)
- Rui Liang
- Department of Gastroenterology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Wei Chen
- Department of Gastroenterology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xiao-Yu Chen
- Department of Gastroenterology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Hui-Ning Fan
- Department of Gastroenterology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jing Zhang
- Department of Gastroenterology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| | - Jin-Shui Zhu
- Department of Gastroenterology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| |
Collapse
|
32
|
Jäger N. Bioinformatics workflows for clinical applications in precision oncology. Semin Cancer Biol 2021; 84:103-112. [PMID: 33476720 DOI: 10.1016/j.semcancer.2020.12.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/15/2020] [Accepted: 12/28/2020] [Indexed: 12/23/2022]
Abstract
High-throughput molecular profiling of tumors is a fundamental aspect of precision oncology, enabling the identification of genomic alterations that can be targeted therapeutically. In this context, a patient is matched to a specific drug or therapy based on the tumor's underlying genetic driver events rather than the histologic classification. This approach requires extensive bioinformatics methodology and workflows, including raw sequencing data processing and quality control, variant calling and annotation, integration of different molecular data types, visualization and finally reporting the data to physicians, cancer researchers and pharmacologists in a format that is readily interpretable for clinical decision making. This review comprises a broad overview of these bioinformatics aspects and discusses the multiple analytical, technical and interpretational challenges that remain to efficiently translate molecular findings into personalized treatment recommendations.
Collapse
Affiliation(s)
- Natalie Jäger
- Hopp Children's Cancer Center Heidelberg (KiTZ) & Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
33
|
Mo XC, Zhang ZT, Song MJ, Zhou ZQ, Zeng JX, Du YF, Sun FZ, Yang JY, He JY, Huang Y, Xia JC, Weng DS. Screening and identification of hub genes in bladder cancer by bioinformatics analysis and KIF11 is a potential prognostic biomarker. Oncol Lett 2021; 21:205. [PMID: 33574944 PMCID: PMC7816288 DOI: 10.3892/ol.2021.12466] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 12/18/2020] [Indexed: 12/20/2022] Open
Abstract
Bladder cancer (BC) is the ninth most common lethal malignancy worldwide. Great efforts have been devoted to clarify the pathogenesis of BC, but the underlying molecular mechanisms remain unclear. To screen for the genes associated with the progression and carcinogenesis of BC, three datasets were obtained from the Gene Expression Omnibus. A total of 37 tumor and 16 non-cancerous samples were analyzed to identify differentially expressed genes (DEGs). Subsequently, 141 genes were identified, including 55 upregulated and 86 downregulated genes. The protein-protein interaction network was established using the Search Tool for Retrieval of Interacting Genes database. Hub gene identification and module analysis were performed using Cytoscape software. Hierarchical clustering of hub genes was conducted using the University of California, Santa Cruz Cancer Genomics Browser. Among the hub genes, kinesin family member 11 (KIF11) was identified as one of the most significant prognostic biomarkers among all the candidates. The Kaplan Meier Plotter database was used for survival analysis of KIF11. The expression profile of KIF11 was analyzed using the ONCOMINE database. The expression levels of KIF11 in BC samples and bladder cells were measured using reverse transcription-quantitative pCR, immunohistochemistry and western blotting. In summary, KIF11 was significantly upregulated in BC and might act as a potential prognostic biomarker. The present identification of DEGs and hub genes in BC may provide novel insight for investigating the molecular mechanisms of BC.
Collapse
Affiliation(s)
- Xiao-Cong Mo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China.,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Zi-Tong Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China.,Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Meng-Jia Song
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China.,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Zi-Qi Zhou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China.,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Jian-Xiong Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China.,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Yu-Fei Du
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China.,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Feng-Ze Sun
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China.,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Jie-Ying Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China.,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Jun-Yi He
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China.,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Yue Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China.,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Jian-Chuan Xia
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China.,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - De-Sheng Weng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou, Guangdong 510060, P.R. China.,Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| |
Collapse
|
34
|
Establishment of a Molecular Tumor Board (MTB) and Uptake of Recommendations in a Community Setting. J Pers Med 2020; 10:jpm10040252. [PMID: 33260805 PMCID: PMC7711773 DOI: 10.3390/jpm10040252] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/22/2020] [Accepted: 11/23/2020] [Indexed: 01/02/2023] Open
Abstract
In the precision medicine era, molecular testing in advanced cancer is foundational to patient management. Molecular tumor boards (MTBs) can be effective in processing comprehensive genomic profiling (CGP) results and providing expert recommendations. We assessed an MTB and its role in a community setting. This retrospective analysis included patients with MTB recommendations at a community-based oncology practice January 2015 to December 2018; exclusions were death within 60 days of the MTB and/or no metastatic disease. Potentially actionable genomic alterations from CGP (immunohistochemistry, in-situ hybridization, next-generation sequencing) were reviewed bi-weekly by MTB practice experts, pathologists, genetic counselors, and other support staff, and clinical care recommendations were provided. Subsequent chart reviews determined implementation rates of recommendations. In 613 patients, the most common cancers were lung (23%), breast (19%), and colorectal (17%); others included ovarian, endometrial, bladder, and melanoma. Patients received 837 actionable recommendations: standard therapy (37%), clinical trial (31%), germline testing and genetic counseling (17%), off-label therapy (10%), subspecialty multidisciplinary tumor board review (2%), and advice for classifying tumor of unknown origin (2%). Of these recommendations, 36% to 78% were followed by the treating physician. For clinical trial recommendations (n = 262), 13% of patients enrolled in a clinical trial. The median time between CPG result availability and MTB presentation was 12 days. A community oncology-based comprehensive and high-throughput MTB provided useful clinical guidance in various treatment domains within an acceptable timeframe for patients with cancer in a large community setting.
Collapse
|
35
|
McNamara DM, Goldberg SL, Latts L, Atieh Graham DM, Waintraub SE, Norden AD, Landstrom C, Pecora AL, Hervey J, Schultz EV, Wang CK, Jungbluth N, Francis PM, Snowdon JL. Differential impact of cognitive computing augmented by real world evidence on novice and expert oncologists. Cancer Med 2019; 8:6578-6584. [PMID: 31509353 PMCID: PMC6825991 DOI: 10.1002/cam4.2548] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 08/01/2019] [Accepted: 08/21/2019] [Indexed: 01/10/2023] Open
Abstract
INTRODUCTION Cognitive computing point-of-care decision support tools which ingest patient attributes from electronic health records and display treatment options based on expert training and medical literature, supplemented by real world evidence (RWE), might prove useful to expert and novice oncologists. The concordance of augmented intelligence systems with best medical practices and potential influences on physician behavior remain unknown. METHODS Electronic health records from 88 breast cancer patients evaluated at a USA tertiary care center were presented to subspecialist experts and oncologists focusing on other disease states with and without reviewing the IBM Watson for Oncology with Cota RWE platform. RESULTS The cognitive computing "recommended" option was concordant with selection by breast cancer experts in 78.5% and "for consideration" option was selected in 9.4%, yielding agreements in 87.9%. Fifty-nine percent of non-concordant responses were generated from 8% of cases. In the Cota observational database 69.3% of matched controls were treated with "recommended," 11.4% "for consideration", and 19.3% "not recommended." Without guidance from Watson for Oncology (WfO)/Cota RWE, novice oncologists chose 75.5% recommended/for consideration treatments which improved to 95.3% with WfO/Cota RWE. The novices were more likely than experts to choose a non-recommended option (P < .01) without WfO/Cota RWE and changed decisions in 39% cases. CONCLUSIONS Watson for Oncology with Cota RWE options were largely concordant with disease expert judged best oncology practices, and was able to improve treatment decisions among breast cancer novices. The observation that nearly a fifth of patients with similar disease characteristics received non-recommended options in a real world database highlights a need for decision support.
Collapse
Affiliation(s)
- Donna M. McNamara
- Division of Breast OncologyJohn Theurer Cancer Center at Hackensack University Medical CenterHackensackNJUSA
| | | | | | - Deena M. Atieh Graham
- Division of Breast OncologyJohn Theurer Cancer Center at Hackensack University Medical CenterHackensackNJUSA
| | - Stanley E. Waintraub
- Division of Breast OncologyJohn Theurer Cancer Center at Hackensack University Medical CenterHackensackNJUSA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Review: Precision medicine and driver mutations: Computational methods, functional assays and conformational principles for interpreting cancer drivers. PLoS Comput Biol 2019; 15:e1006658. [PMID: 30921324 PMCID: PMC6438456 DOI: 10.1371/journal.pcbi.1006658] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
At the root of the so-called precision medicine or precision oncology, which is our focus here, is the hypothesis that cancer treatment would be considerably better if therapies were guided by a tumor’s genomic alterations. This hypothesis has sparked major initiatives focusing on whole-genome and/or exome sequencing, creation of large databases, and developing tools for their statistical analyses—all aspiring to identify actionable alterations, and thus molecular targets, in a patient. At the center of the massive amount of collected sequence data is their interpretations that largely rest on statistical analysis and phenotypic observations. Statistics is vital, because it guides identification of cancer-driving alterations. However, statistics of mutations do not identify a change in protein conformation; therefore, it may not define sufficiently accurate actionable mutations, neglecting those that are rare. Among the many thematic overviews of precision oncology, this review innovates by further comprehensively including precision pharmacology, and within this framework, articulating its protein structural landscape and consequences to cellular signaling pathways. It provides the underlying physicochemical basis, thereby also opening the door to a broader community.
Collapse
|
37
|
Singer F, Irmisch A, Toussaint NC, Grob L, Singer J, Thurnherr T, Beerenwinkel N, Levesque MP, Dummer R, Quagliata L, Rothschild SI, Wicki A, Beisel C, Stekhoven DJ. SwissMTB: establishing comprehensive molecular cancer diagnostics in Swiss clinics. BMC Med Inform Decis Mak 2018; 18:89. [PMID: 30373609 PMCID: PMC6206832 DOI: 10.1186/s12911-018-0680-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 10/18/2018] [Indexed: 12/18/2022] Open
Abstract
Background Molecular precision oncology is an emerging practice to improve cancer therapy by decreasing the risk of choosing treatments that lack efficacy or cause adverse events. However, the challenges of integrating molecular profiling into routine clinical care are manifold. From a computational perspective these include the importance of a short analysis turnaround time, the interpretation of complex drug-gene and gene-gene interactions, and the necessity of standardized high-quality workflows. In addition, difficulties faced when integrating molecular diagnostics into clinical practice are ethical concerns, legal requirements, and limited availability of treatment options beyond standard of care as well as the overall lack of awareness of their existence. Methods To the best of our knowledge, we are the first group in Switzerland that established a workflow for personalized diagnostics based on comprehensive high-throughput sequencing of tumors at the clinic. Our workflow, named SwissMTB (Swiss Molecular Tumor Board), links genetic tumor alterations and gene expression to therapeutic options and clinical trial opportunities. The resulting treatment recommendations are summarized in a clinical report and discussed in a molecular tumor board at the clinic to support therapy decisions. Results Here we present results from an observational pilot study including 22 late-stage cancer patients. In this study we were able to identify actionable variants and corresponding therapies for 19 patients. Half of the patients were analyzed retrospectively. In two patients we identified resistance-associated variants explaining lack of therapy response. For five out of eleven patients analyzed before treatment the SwissMTB diagnostic influenced treatment decision. Conclusions SwissMTB enables the analysis and clinical interpretation of large numbers of potentially actionable molecular targets. Thus, our workflow paves the way towards a more frequent use of comprehensive molecular diagnostics in Swiss hospitals. Electronic supplementary material The online version of this article (10.1186/s12911-018-0680-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Franziska Singer
- NEXUS Personalized Health Technologies, ETH Zurich, Otto-Stern-Weg 7, 8093, Zurich, Switzerland.,SIB Swiss Institute of Bioinformatics, 4058, Basel, Switzerland
| | - Anja Irmisch
- Department of Dermatology, University Hospital Zurich, 8091, Zurich, Switzerland
| | - Nora C Toussaint
- NEXUS Personalized Health Technologies, ETH Zurich, Otto-Stern-Weg 7, 8093, Zurich, Switzerland.,SIB Swiss Institute of Bioinformatics, 4058, Basel, Switzerland
| | - Linda Grob
- NEXUS Personalized Health Technologies, ETH Zurich, Otto-Stern-Weg 7, 8093, Zurich, Switzerland.,SIB Swiss Institute of Bioinformatics, 4058, Basel, Switzerland
| | - Jochen Singer
- SIB Swiss Institute of Bioinformatics, 4058, Basel, Switzerland.,Department of Biosystems Science and Engineering, ETH Zurich, 4058, Basel, Switzerland
| | - Thomas Thurnherr
- SIB Swiss Institute of Bioinformatics, 4058, Basel, Switzerland.,Department of Biosystems Science and Engineering, ETH Zurich, 4058, Basel, Switzerland
| | - Niko Beerenwinkel
- SIB Swiss Institute of Bioinformatics, 4058, Basel, Switzerland.,Department of Biosystems Science and Engineering, ETH Zurich, 4058, Basel, Switzerland
| | - Mitchell P Levesque
- Department of Dermatology, University Hospital Zurich, 8091, Zurich, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, 8091, Zurich, Switzerland
| | - Luca Quagliata
- Department of Pathology, University Hospital Basel, Schönbeinstrasse 40, 4056, Basel, Switzerland
| | - Sacha I Rothschild
- Division of Oncology, Department of Biomedicine, University Hospital Basel, Petersgraben 4, 4031, Basel, Switzerland
| | - Andreas Wicki
- Division of Oncology, Department of Biomedicine, University Hospital Basel, Petersgraben 4, 4031, Basel, Switzerland
| | - Christian Beisel
- Department of Biosystems Science and Engineering, ETH Zurich, 4058, Basel, Switzerland
| | - Daniel J Stekhoven
- NEXUS Personalized Health Technologies, ETH Zurich, Otto-Stern-Weg 7, 8093, Zurich, Switzerland. .,SIB Swiss Institute of Bioinformatics, 4058, Basel, Switzerland.
| |
Collapse
|
38
|
Hoefflin R, Geißler AL, Fritsch R, Claus R, Wehrle J, Metzger P, Reiser M, Mehmed L, Fauth L, Heiland DH, Erbes T, Stock F, Csanadi A, Miething C, Weddeling B, Meiss F, von Bubnoff D, Dierks C, Ge I, Brass V, Heeg S, Schäfer H, Boeker M, Rawluk J, Botzenhart EM, Kayser G, Hettmer S, Busch H, Peters C, Werner M, Duyster J, Brummer T, Boerries M, Lassmann S, von Bubnoff N. Personalized Clinical Decision Making Through Implementation of a Molecular Tumor Board: A German Single-Center Experience. JCO Precis Oncol 2018; 2:PO.18.00105. [PMID: 32913998 PMCID: PMC7446498 DOI: 10.1200/po.18.00105] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
PURPOSE Dramatic advances in our understanding of the molecular pathophysiology of cancer, along with a rapidly expanding portfolio of molecular targeted drugs, have led to a paradigm shift toward personalized, biomarker-driven cancer treatment. Here, we report the 2-year experience of the Comprehensive Cancer Center Freiburg Molecular Tumor Board (MTB), one of the first interdisciplinary molecular tumor conferences established in Europe. The role of the MTB is to recommend personalized therapy for patients with cancer beyond standard-of-care treatment. METHODS This retrospective case series includes 198 patients discussed from March 2015 through February 2017. The MTB guided individual molecular diagnostics, assessed evidence of actionability of molecular alterations, and provided therapy recommendations, including approved and off-label treatments as well as available matched clinical trials. RESULTS The majority of patients had metastatic solid tumors (73.7%), mostly progressive (77.3%) after a mean of 2.0 lines of standard treatment. Diagnostic recommendations resulted in 867 molecular diagnostic tests for 172 patients (five per case), including exome analysis in 36 cases (18.2%). With a median turnaround time of 28 days, treatment recommendations were given to 104 patients (52.5%). These included single-agent targeted therapies (42.3%), checkpoint inhibitors (37.5%), and combination therapies (18.3%). Treatment recommendations were implemented in 33 of 104 patients (31.7%), of whom 19 (57.6%) showed stable disease or partial response, including 14 patients (7.1% of the entire population) receiving off-label treatments. CONCLUSION Personalized extended molecular-guided patient care is effective for a small but clinically meaningful proportion of patients in challenging clinical situations. Limited access to targeted drugs, lack of trials, and submission at late disease stage prevents broader applicability, whereas genome-wide analyses are not a strict requirement for predictive molecular testing.
Collapse
Affiliation(s)
- Rouven Hoefflin
- All authors: University of Freiburg, Freiburg; Ralph Fritsch, Julius Wehrle, Cornelius Miething, Christoph Peters, Martin Werner, Justus Duyster, Tilman Brummer, Melanie Boerries, Silke Lassmann, and Nikolas von Bubnoff, German Cancer Consortium, partner site Freiburg, and German Cancer Research Center, Heidelberg; Rainer Claus, Augsburg Medical Center, Augsburg; and Hauke Busch, University of Lübeck, Lübeck, Germany
| | - Anna-Lena Geißler
- All authors: University of Freiburg, Freiburg; Ralph Fritsch, Julius Wehrle, Cornelius Miething, Christoph Peters, Martin Werner, Justus Duyster, Tilman Brummer, Melanie Boerries, Silke Lassmann, and Nikolas von Bubnoff, German Cancer Consortium, partner site Freiburg, and German Cancer Research Center, Heidelberg; Rainer Claus, Augsburg Medical Center, Augsburg; and Hauke Busch, University of Lübeck, Lübeck, Germany
| | - Ralph Fritsch
- All authors: University of Freiburg, Freiburg; Ralph Fritsch, Julius Wehrle, Cornelius Miething, Christoph Peters, Martin Werner, Justus Duyster, Tilman Brummer, Melanie Boerries, Silke Lassmann, and Nikolas von Bubnoff, German Cancer Consortium, partner site Freiburg, and German Cancer Research Center, Heidelberg; Rainer Claus, Augsburg Medical Center, Augsburg; and Hauke Busch, University of Lübeck, Lübeck, Germany
| | - Rainer Claus
- All authors: University of Freiburg, Freiburg; Ralph Fritsch, Julius Wehrle, Cornelius Miething, Christoph Peters, Martin Werner, Justus Duyster, Tilman Brummer, Melanie Boerries, Silke Lassmann, and Nikolas von Bubnoff, German Cancer Consortium, partner site Freiburg, and German Cancer Research Center, Heidelberg; Rainer Claus, Augsburg Medical Center, Augsburg; and Hauke Busch, University of Lübeck, Lübeck, Germany
| | - Julius Wehrle
- All authors: University of Freiburg, Freiburg; Ralph Fritsch, Julius Wehrle, Cornelius Miething, Christoph Peters, Martin Werner, Justus Duyster, Tilman Brummer, Melanie Boerries, Silke Lassmann, and Nikolas von Bubnoff, German Cancer Consortium, partner site Freiburg, and German Cancer Research Center, Heidelberg; Rainer Claus, Augsburg Medical Center, Augsburg; and Hauke Busch, University of Lübeck, Lübeck, Germany
| | - Patrick Metzger
- All authors: University of Freiburg, Freiburg; Ralph Fritsch, Julius Wehrle, Cornelius Miething, Christoph Peters, Martin Werner, Justus Duyster, Tilman Brummer, Melanie Boerries, Silke Lassmann, and Nikolas von Bubnoff, German Cancer Consortium, partner site Freiburg, and German Cancer Research Center, Heidelberg; Rainer Claus, Augsburg Medical Center, Augsburg; and Hauke Busch, University of Lübeck, Lübeck, Germany
| | - Meike Reiser
- All authors: University of Freiburg, Freiburg; Ralph Fritsch, Julius Wehrle, Cornelius Miething, Christoph Peters, Martin Werner, Justus Duyster, Tilman Brummer, Melanie Boerries, Silke Lassmann, and Nikolas von Bubnoff, German Cancer Consortium, partner site Freiburg, and German Cancer Research Center, Heidelberg; Rainer Claus, Augsburg Medical Center, Augsburg; and Hauke Busch, University of Lübeck, Lübeck, Germany
| | - Leman Mehmed
- All authors: University of Freiburg, Freiburg; Ralph Fritsch, Julius Wehrle, Cornelius Miething, Christoph Peters, Martin Werner, Justus Duyster, Tilman Brummer, Melanie Boerries, Silke Lassmann, and Nikolas von Bubnoff, German Cancer Consortium, partner site Freiburg, and German Cancer Research Center, Heidelberg; Rainer Claus, Augsburg Medical Center, Augsburg; and Hauke Busch, University of Lübeck, Lübeck, Germany
| | - Lisa Fauth
- All authors: University of Freiburg, Freiburg; Ralph Fritsch, Julius Wehrle, Cornelius Miething, Christoph Peters, Martin Werner, Justus Duyster, Tilman Brummer, Melanie Boerries, Silke Lassmann, and Nikolas von Bubnoff, German Cancer Consortium, partner site Freiburg, and German Cancer Research Center, Heidelberg; Rainer Claus, Augsburg Medical Center, Augsburg; and Hauke Busch, University of Lübeck, Lübeck, Germany
| | - Dieter Henrik Heiland
- All authors: University of Freiburg, Freiburg; Ralph Fritsch, Julius Wehrle, Cornelius Miething, Christoph Peters, Martin Werner, Justus Duyster, Tilman Brummer, Melanie Boerries, Silke Lassmann, and Nikolas von Bubnoff, German Cancer Consortium, partner site Freiburg, and German Cancer Research Center, Heidelberg; Rainer Claus, Augsburg Medical Center, Augsburg; and Hauke Busch, University of Lübeck, Lübeck, Germany
| | - Thalia Erbes
- All authors: University of Freiburg, Freiburg; Ralph Fritsch, Julius Wehrle, Cornelius Miething, Christoph Peters, Martin Werner, Justus Duyster, Tilman Brummer, Melanie Boerries, Silke Lassmann, and Nikolas von Bubnoff, German Cancer Consortium, partner site Freiburg, and German Cancer Research Center, Heidelberg; Rainer Claus, Augsburg Medical Center, Augsburg; and Hauke Busch, University of Lübeck, Lübeck, Germany
| | - Friedrich Stock
- All authors: University of Freiburg, Freiburg; Ralph Fritsch, Julius Wehrle, Cornelius Miething, Christoph Peters, Martin Werner, Justus Duyster, Tilman Brummer, Melanie Boerries, Silke Lassmann, and Nikolas von Bubnoff, German Cancer Consortium, partner site Freiburg, and German Cancer Research Center, Heidelberg; Rainer Claus, Augsburg Medical Center, Augsburg; and Hauke Busch, University of Lübeck, Lübeck, Germany
| | - Agnes Csanadi
- All authors: University of Freiburg, Freiburg; Ralph Fritsch, Julius Wehrle, Cornelius Miething, Christoph Peters, Martin Werner, Justus Duyster, Tilman Brummer, Melanie Boerries, Silke Lassmann, and Nikolas von Bubnoff, German Cancer Consortium, partner site Freiburg, and German Cancer Research Center, Heidelberg; Rainer Claus, Augsburg Medical Center, Augsburg; and Hauke Busch, University of Lübeck, Lübeck, Germany
| | - Cornelius Miething
- All authors: University of Freiburg, Freiburg; Ralph Fritsch, Julius Wehrle, Cornelius Miething, Christoph Peters, Martin Werner, Justus Duyster, Tilman Brummer, Melanie Boerries, Silke Lassmann, and Nikolas von Bubnoff, German Cancer Consortium, partner site Freiburg, and German Cancer Research Center, Heidelberg; Rainer Claus, Augsburg Medical Center, Augsburg; and Hauke Busch, University of Lübeck, Lübeck, Germany
| | - Britta Weddeling
- All authors: University of Freiburg, Freiburg; Ralph Fritsch, Julius Wehrle, Cornelius Miething, Christoph Peters, Martin Werner, Justus Duyster, Tilman Brummer, Melanie Boerries, Silke Lassmann, and Nikolas von Bubnoff, German Cancer Consortium, partner site Freiburg, and German Cancer Research Center, Heidelberg; Rainer Claus, Augsburg Medical Center, Augsburg; and Hauke Busch, University of Lübeck, Lübeck, Germany
| | - Frank Meiss
- All authors: University of Freiburg, Freiburg; Ralph Fritsch, Julius Wehrle, Cornelius Miething, Christoph Peters, Martin Werner, Justus Duyster, Tilman Brummer, Melanie Boerries, Silke Lassmann, and Nikolas von Bubnoff, German Cancer Consortium, partner site Freiburg, and German Cancer Research Center, Heidelberg; Rainer Claus, Augsburg Medical Center, Augsburg; and Hauke Busch, University of Lübeck, Lübeck, Germany
| | - Dagmar von Bubnoff
- All authors: University of Freiburg, Freiburg; Ralph Fritsch, Julius Wehrle, Cornelius Miething, Christoph Peters, Martin Werner, Justus Duyster, Tilman Brummer, Melanie Boerries, Silke Lassmann, and Nikolas von Bubnoff, German Cancer Consortium, partner site Freiburg, and German Cancer Research Center, Heidelberg; Rainer Claus, Augsburg Medical Center, Augsburg; and Hauke Busch, University of Lübeck, Lübeck, Germany
| | - Christine Dierks
- All authors: University of Freiburg, Freiburg; Ralph Fritsch, Julius Wehrle, Cornelius Miething, Christoph Peters, Martin Werner, Justus Duyster, Tilman Brummer, Melanie Boerries, Silke Lassmann, and Nikolas von Bubnoff, German Cancer Consortium, partner site Freiburg, and German Cancer Research Center, Heidelberg; Rainer Claus, Augsburg Medical Center, Augsburg; and Hauke Busch, University of Lübeck, Lübeck, Germany
| | - Isabell Ge
- All authors: University of Freiburg, Freiburg; Ralph Fritsch, Julius Wehrle, Cornelius Miething, Christoph Peters, Martin Werner, Justus Duyster, Tilman Brummer, Melanie Boerries, Silke Lassmann, and Nikolas von Bubnoff, German Cancer Consortium, partner site Freiburg, and German Cancer Research Center, Heidelberg; Rainer Claus, Augsburg Medical Center, Augsburg; and Hauke Busch, University of Lübeck, Lübeck, Germany
| | - Volker Brass
- All authors: University of Freiburg, Freiburg; Ralph Fritsch, Julius Wehrle, Cornelius Miething, Christoph Peters, Martin Werner, Justus Duyster, Tilman Brummer, Melanie Boerries, Silke Lassmann, and Nikolas von Bubnoff, German Cancer Consortium, partner site Freiburg, and German Cancer Research Center, Heidelberg; Rainer Claus, Augsburg Medical Center, Augsburg; and Hauke Busch, University of Lübeck, Lübeck, Germany
| | - Steffen Heeg
- All authors: University of Freiburg, Freiburg; Ralph Fritsch, Julius Wehrle, Cornelius Miething, Christoph Peters, Martin Werner, Justus Duyster, Tilman Brummer, Melanie Boerries, Silke Lassmann, and Nikolas von Bubnoff, German Cancer Consortium, partner site Freiburg, and German Cancer Research Center, Heidelberg; Rainer Claus, Augsburg Medical Center, Augsburg; and Hauke Busch, University of Lübeck, Lübeck, Germany
| | - Henning Schäfer
- All authors: University of Freiburg, Freiburg; Ralph Fritsch, Julius Wehrle, Cornelius Miething, Christoph Peters, Martin Werner, Justus Duyster, Tilman Brummer, Melanie Boerries, Silke Lassmann, and Nikolas von Bubnoff, German Cancer Consortium, partner site Freiburg, and German Cancer Research Center, Heidelberg; Rainer Claus, Augsburg Medical Center, Augsburg; and Hauke Busch, University of Lübeck, Lübeck, Germany
| | - Martin Boeker
- All authors: University of Freiburg, Freiburg; Ralph Fritsch, Julius Wehrle, Cornelius Miething, Christoph Peters, Martin Werner, Justus Duyster, Tilman Brummer, Melanie Boerries, Silke Lassmann, and Nikolas von Bubnoff, German Cancer Consortium, partner site Freiburg, and German Cancer Research Center, Heidelberg; Rainer Claus, Augsburg Medical Center, Augsburg; and Hauke Busch, University of Lübeck, Lübeck, Germany
| | - Justyna Rawluk
- All authors: University of Freiburg, Freiburg; Ralph Fritsch, Julius Wehrle, Cornelius Miething, Christoph Peters, Martin Werner, Justus Duyster, Tilman Brummer, Melanie Boerries, Silke Lassmann, and Nikolas von Bubnoff, German Cancer Consortium, partner site Freiburg, and German Cancer Research Center, Heidelberg; Rainer Claus, Augsburg Medical Center, Augsburg; and Hauke Busch, University of Lübeck, Lübeck, Germany
| | - Elke Maria Botzenhart
- All authors: University of Freiburg, Freiburg; Ralph Fritsch, Julius Wehrle, Cornelius Miething, Christoph Peters, Martin Werner, Justus Duyster, Tilman Brummer, Melanie Boerries, Silke Lassmann, and Nikolas von Bubnoff, German Cancer Consortium, partner site Freiburg, and German Cancer Research Center, Heidelberg; Rainer Claus, Augsburg Medical Center, Augsburg; and Hauke Busch, University of Lübeck, Lübeck, Germany
| | - Gian Kayser
- All authors: University of Freiburg, Freiburg; Ralph Fritsch, Julius Wehrle, Cornelius Miething, Christoph Peters, Martin Werner, Justus Duyster, Tilman Brummer, Melanie Boerries, Silke Lassmann, and Nikolas von Bubnoff, German Cancer Consortium, partner site Freiburg, and German Cancer Research Center, Heidelberg; Rainer Claus, Augsburg Medical Center, Augsburg; and Hauke Busch, University of Lübeck, Lübeck, Germany
| | - Simone Hettmer
- All authors: University of Freiburg, Freiburg; Ralph Fritsch, Julius Wehrle, Cornelius Miething, Christoph Peters, Martin Werner, Justus Duyster, Tilman Brummer, Melanie Boerries, Silke Lassmann, and Nikolas von Bubnoff, German Cancer Consortium, partner site Freiburg, and German Cancer Research Center, Heidelberg; Rainer Claus, Augsburg Medical Center, Augsburg; and Hauke Busch, University of Lübeck, Lübeck, Germany
| | - Hauke Busch
- All authors: University of Freiburg, Freiburg; Ralph Fritsch, Julius Wehrle, Cornelius Miething, Christoph Peters, Martin Werner, Justus Duyster, Tilman Brummer, Melanie Boerries, Silke Lassmann, and Nikolas von Bubnoff, German Cancer Consortium, partner site Freiburg, and German Cancer Research Center, Heidelberg; Rainer Claus, Augsburg Medical Center, Augsburg; and Hauke Busch, University of Lübeck, Lübeck, Germany
| | - Christoph Peters
- All authors: University of Freiburg, Freiburg; Ralph Fritsch, Julius Wehrle, Cornelius Miething, Christoph Peters, Martin Werner, Justus Duyster, Tilman Brummer, Melanie Boerries, Silke Lassmann, and Nikolas von Bubnoff, German Cancer Consortium, partner site Freiburg, and German Cancer Research Center, Heidelberg; Rainer Claus, Augsburg Medical Center, Augsburg; and Hauke Busch, University of Lübeck, Lübeck, Germany
| | - Martin Werner
- All authors: University of Freiburg, Freiburg; Ralph Fritsch, Julius Wehrle, Cornelius Miething, Christoph Peters, Martin Werner, Justus Duyster, Tilman Brummer, Melanie Boerries, Silke Lassmann, and Nikolas von Bubnoff, German Cancer Consortium, partner site Freiburg, and German Cancer Research Center, Heidelberg; Rainer Claus, Augsburg Medical Center, Augsburg; and Hauke Busch, University of Lübeck, Lübeck, Germany
| | - Justus Duyster
- All authors: University of Freiburg, Freiburg; Ralph Fritsch, Julius Wehrle, Cornelius Miething, Christoph Peters, Martin Werner, Justus Duyster, Tilman Brummer, Melanie Boerries, Silke Lassmann, and Nikolas von Bubnoff, German Cancer Consortium, partner site Freiburg, and German Cancer Research Center, Heidelberg; Rainer Claus, Augsburg Medical Center, Augsburg; and Hauke Busch, University of Lübeck, Lübeck, Germany
| | - Tilman Brummer
- All authors: University of Freiburg, Freiburg; Ralph Fritsch, Julius Wehrle, Cornelius Miething, Christoph Peters, Martin Werner, Justus Duyster, Tilman Brummer, Melanie Boerries, Silke Lassmann, and Nikolas von Bubnoff, German Cancer Consortium, partner site Freiburg, and German Cancer Research Center, Heidelberg; Rainer Claus, Augsburg Medical Center, Augsburg; and Hauke Busch, University of Lübeck, Lübeck, Germany
| | - Melanie Boerries
- All authors: University of Freiburg, Freiburg; Ralph Fritsch, Julius Wehrle, Cornelius Miething, Christoph Peters, Martin Werner, Justus Duyster, Tilman Brummer, Melanie Boerries, Silke Lassmann, and Nikolas von Bubnoff, German Cancer Consortium, partner site Freiburg, and German Cancer Research Center, Heidelberg; Rainer Claus, Augsburg Medical Center, Augsburg; and Hauke Busch, University of Lübeck, Lübeck, Germany
| | - Silke Lassmann
- All authors: University of Freiburg, Freiburg; Ralph Fritsch, Julius Wehrle, Cornelius Miething, Christoph Peters, Martin Werner, Justus Duyster, Tilman Brummer, Melanie Boerries, Silke Lassmann, and Nikolas von Bubnoff, German Cancer Consortium, partner site Freiburg, and German Cancer Research Center, Heidelberg; Rainer Claus, Augsburg Medical Center, Augsburg; and Hauke Busch, University of Lübeck, Lübeck, Germany
| | - Nikolas von Bubnoff
- All authors: University of Freiburg, Freiburg; Ralph Fritsch, Julius Wehrle, Cornelius Miething, Christoph Peters, Martin Werner, Justus Duyster, Tilman Brummer, Melanie Boerries, Silke Lassmann, and Nikolas von Bubnoff, German Cancer Consortium, partner site Freiburg, and German Cancer Research Center, Heidelberg; Rainer Claus, Augsburg Medical Center, Augsburg; and Hauke Busch, University of Lübeck, Lübeck, Germany
| |
Collapse
|
39
|
Pantziarka P, Bouche G, André N. "Hard" Drug Repurposing for Precision Oncology: The Missing Link? Front Pharmacol 2018; 9:637. [PMID: 29962954 PMCID: PMC6010551 DOI: 10.3389/fphar.2018.00637] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 05/29/2018] [Indexed: 12/18/2022] Open
Affiliation(s)
- Pan Pantziarka
- Anticancer Fund, Brussels, Belgium.,The George Pantziarka TP53 Trust, London, United Kingdom
| | | | - Nicolas André
- Department of Pediatric Hematology-Oncology, Assistance Publique Hôpitaux de Marseille, La Timone Hospital, Marseille, France.,Centre de Recherche en Oncologie Biologique et en Oncopharmacologie, Institut National de la Santé et de la Recherche Médicale-UMR 79 911, Aix-Marseille University, Marseille, France.,Metronomics Global Health Initiative, Marseille, France
| |
Collapse
|
40
|
Sun Y, Sun X, Liu S, Liu L, Chen J. The overlap between regeneration and fibrosis in injured skeletal muscle is regulated by phosphatidylinositol 3-kinase/Akt signaling pathway - A bioinformatic analysis based on lncRNA microarray. Gene 2018; 672:79-87. [PMID: 29870770 DOI: 10.1016/j.gene.2018.06.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 05/05/2018] [Accepted: 06/01/2018] [Indexed: 02/05/2023]
Abstract
Injured skeletal muscle would go through a sequence of the pathological phases of degeneration, myogenesis and fibrosis. Growing evidence indicated that fibrotic and myogenic phases might overlap within the injured skeletal muscle in the early time after injury. However, the mechanism underlying this overlapping remains unclear. Here, we performed an lncRNA microarray to identify the activated pathways in mice muscle seven days after contusion. KEGG analysis indicated that phosphatidylinositol 3-kinase/Akt (PI3K/Akt) signaling cascade was predicted to be activated by lncRNAs. The top genes targeted by lncRNAs in PI3K/Akt signaling were subunits of laminin, collagen 5, and collagen 6, which participated in either myogenic or fibrotic process. Reverse transcriptase-polymerase chain reaction analysis and immunohistochemical stain further confirmed the prediction in silico. These results suggested that the overlap might be related to an activated PI3K/Akt pathway by lncRNA regulation.
Collapse
Affiliation(s)
- Yaying Sun
- Department of Sports Medicine, Huashan Hospital, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xiaomeng Sun
- Institute of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Shaohua Liu
- Department of Sports Medicine, Huashan Hospital, Shanghai Medical College of Fudan University, Shanghai, China
| | - Lei Liu
- Institute of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Jiwu Chen
- Department of Sports Medicine, Huashan Hospital, Shanghai Medical College of Fudan University, Shanghai, China.
| |
Collapse
|
41
|
|