1
|
Takeda K, Hashimoto A, Liu S, Rong A. A basket trial design based on constrained hierarchical Bayesian model for latent subgroups. J Biopharm Stat 2025; 35:271-282. [PMID: 38369872 DOI: 10.1080/10543406.2024.2311851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/24/2024] [Indexed: 02/20/2024]
Abstract
It is well known a basket trial consisting of multiple cancer types has the potential of borrowing strength across the baskets defined by the cancer types, leading to an efficient design in terms of sample size and trial duration. The treatment effects in those baskets are often heterogeneous and categorized by the cancer types being sensitive or insensitive to the treatment. Hence, the assumption of exchangeability in many existing basket trials may be violated, and there is a need to design trials without this assumption. In this paper, we simplify the constrained hierarchical Bayesian model for latent subgroups (CHBM-LS) for two classifiers to deal with the potential heterogeneity of treatment effects due to the single classifier of the cancer type. Different baskets are aggregated into subgroups using a latent subgroup modeling approach. The treatment effects are similar and exchangeable to facilitate information borrowing within each latent subgroup. Applying the simplified CHBM-LS approach to the real basket trials where baskets defined by only cancer types shows better performance than other available approaches. Further simulation study also demonstrates this CHBM-LS approach outperforms other approaches with higher statistical power and better-controlled type I error rates under various scenarios.
Collapse
Affiliation(s)
- Kentaro Takeda
- Data Science, Astellas Pharma Global Development Inc, Northbrook, Illinois, USA
| | | | - Shufang Liu
- Oncology Biostatistics, Gilead Sciences Inc, Foster City, California, USA
| | - Alan Rong
- Oncology Biostatistics, Gilead Sciences Inc, Foster City, California, USA
| |
Collapse
|
2
|
Suo Y, Song Y, Wang Y, Liu Q, Rodriguez H, Zhou H. Advancements in proteogenomics for preclinical targeted cancer therapy research. BIOPHYSICS REPORTS 2025; 11:56-76. [PMID: 40070661 PMCID: PMC11891078 DOI: 10.52601/bpr.2024.240053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 12/03/2024] [Indexed: 03/14/2025] Open
Abstract
Advancements in molecular characterization technologies have accelerated targeted cancer therapy research at unprecedented resolution and dimensionality. Integrating comprehensive multi-omic molecular profiling of a tumor, proteogenomics, marks a transformative milestone for preclinical cancer research. In this paper, we initially provided an overview of proteogenomics in cancer research, spanning genomics, transcriptomics, and proteomics. Subsequently, the applications were introduced and examined from different perspectives, including but not limited to genetic alterations, molecular quantifications, single-cell patterns, different post-translational modification levels, subtype signatures, and immune landscape. We also paid attention to the combined multi-omics data analysis and pan-cancer analysis. This paper highlights the crucial role of proteogenomics in preclinical targeted cancer therapy research, including but not limited to elucidating the mechanisms of tumorigenesis, discovering effective therapeutic targets and promising biomarkers, and developing subtype-specific therapies.
Collapse
Affiliation(s)
- Yuying Suo
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuanli Song
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yuqiu Wang
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Department of Otolaryngology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
| | - Qian Liu
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Henry Rodriguez
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, National Institutes of Health, Rockville, MD 20850, USA
| | - Hu Zhou
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
3
|
Wen W, Zhao S, Jiang Y, Ou C, Guo C, Jia Z, Li J, Huang Y, Xu H, Pu P, Shang T, Cong L, Wang X, Wu N, Liu J. Genome sequencing enhances the diagnostic yield and expands the genetic landscape of male breast cancer. GENETICS IN MEDICINE OPEN 2024; 3:101899. [PMID: 39981113 PMCID: PMC11840214 DOI: 10.1016/j.gimo.2024.101899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 02/22/2025]
Abstract
Purpose To understand the broader genetic landscape of male breast cancer (MBC), focusing on the utility of genome sequencing (GS) beyond BRCA1/2 (HGNC: 1100, 1101) variants. Methods Twenty-four patients with MBC underwent a multistep genetic analysis. Initial screening targeted BRCA1/2 variants followed by GS to identify pathogenic/likely pathogenic germline variants through a 3-tiered classification. Polygenic risk score analysis was further incorporated using a model for female breast cancer with 2666 noncancer controls. Exome sequencing was used to transition from germline to somatic investigations, assessing second-hit variant and mutational signatures. Results The GS analysis unveiled previously unrecognized pathogenic/likely pathogenic germline variants in BARD1, ATR, BRIP1, and CHEK2 (HGNC: 952, 882, 20473, 16627) among 21 BRCA1/2-negative patients with MBC, elevating the diagnostic yield from 12.5% to 33.0% in all MBC. Elevated average polygenic risk score was noted compared with controls, with a significant correlation to early-onset MBC when combined with high-penetrance germline pathogenic variants (P = 1.10 × 10-4). Exome sequencing analysis further identified significant somatic oncogenic drivers and revealed a dominant mutational signature SBS3 across BRCA1/2-negative samples, reinforcing the contribution of omologous recombination deficiency underlying the MBC development. Conclusion Our findings extended the MBC genetic spectrum beyond BRCA1/2 and highlighted the intricate interplay of monogenic and polygenic predispositions, presenting a comprehensive MBC genomic profile.
Collapse
Affiliation(s)
- Wen Wen
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- School of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Sen Zhao
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | - Yiwen Jiang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- School of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chengzhu Ou
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- School of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Changyuan Guo
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ziqi Jia
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiayi Li
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- School of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yansong Huang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- School of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hengyi Xu
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- School of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Pengming Pu
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- School of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tongxuan Shang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- School of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lin Cong
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- School of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiang Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nan Wu
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing, China
| | - Jiaqi Liu
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
4
|
Shams A. Leveraging State-of-the-Art AI Algorithms in Personalized Oncology: From Transcriptomics to Treatment. Diagnostics (Basel) 2024; 14:2174. [PMID: 39410578 PMCID: PMC11476216 DOI: 10.3390/diagnostics14192174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND Continuous breakthroughs in computational algorithms have positioned AI-based models as some of the most sophisticated technologies in the healthcare system. AI shows dynamic contributions in advancing various medical fields involving data interpretation and monitoring, imaging screening and diagnosis, and treatment response and survival prediction. Despite advances in clinical oncology, more effort must be employed to tailor therapeutic plans based on each patient's unique transcriptomic profile within the precision/personalized oncology frame. Furthermore, the standard analysis method is not compatible with the comprehensive deciphering of significant data streams, thus precluding the prediction of accurate treatment options. METHODOLOGY We proposed a novel approach that includes obtaining different tumour tissues and preparing RNA samples for comprehensive transcriptomic interpretation using specifically trained, programmed, and optimized AI-based models for extracting large data volumes, refining, and analyzing them. Next, the transcriptomic results will be scanned against an expansive drug library to predict the response of each target to the tested drugs. The obtained target-drug combination/s will be then validated using in vitro and in vivo experimental models. Finally, the best treatment combination option/s will be introduced to the patient. We also provided a comprehensive review discussing AI models' recent innovations and implementations to aid in molecular diagnosis and treatment planning. RESULTS The expected transcriptomic analysis generated by the AI-based algorithms will provide an inclusive genomic profile for each patient, containing statistical and bioinformatics analyses, identification of the dysregulated pathways, detection of the targeted genes, and recognition of molecular biomarkers. Subjecting these results to the prediction and pairing AI-based processes will result in statistical graphs presenting each target's likely response rate to various treatment options. Different in vitro and in vivo investigations will further validate the selection of the target drug/s pairs. CONCLUSIONS Leveraging AI models will provide more rigorous manipulation of large-scale datasets on specific cancer care paths. Such a strategy would shape treatment according to each patient's demand, thus fortifying the avenue of personalized/precision medicine. Undoubtedly, this will assist in improving the oncology domain and alleviate the burden of clinicians in the coming decade.
Collapse
Affiliation(s)
- Anwar Shams
- Department of Pharmacology, College of Medicine, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia; or ; Tel.: +00966-548638099
- Research Center for Health Sciences, Deanship of Graduate Studies and Scientific Research, Taif University, Taif 26432, Saudi Arabia
- High Altitude Research Center, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| |
Collapse
|
5
|
El-Sayed MM, Bianco JR, Li Y, Fabian Z. Tumor-Agnostic Therapy-The Final Step Forward in the Cure for Human Neoplasms? Cells 2024; 13:1071. [PMID: 38920700 PMCID: PMC11201516 DOI: 10.3390/cells13121071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 06/27/2024] Open
Abstract
Cancer accounted for 10 million deaths in 2020, nearly one in every six deaths annually. Despite advancements, the contemporary clinical management of human neoplasms faces a number of challenges. Surgical removal of tumor tissues is often not possible technically, while radiation and chemotherapy pose the risk of damaging healthy cells, tissues, and organs, presenting complex clinical challenges. These require a paradigm shift in developing new therapeutic modalities moving towards a more personalized and targeted approach. The tumor-agnostic philosophy, one of these new modalities, focuses on characteristic molecular signatures of transformed cells independently of their traditional histopathological classification. These include commonly occurring DNA aberrations in cancer cells, shared metabolic features of their homeostasis or immune evasion measures of the tumor tissues. The first dedicated, FDA-approved tumor-agnostic agent's profound progression-free survival of 78% in mismatch repair-deficient colorectal cancer paved the way for the accelerated FDA approvals of novel tumor-agnostic therapeutic compounds. Here, we review the historical background, current status, and future perspectives of this new era of clinical oncology.
Collapse
Affiliation(s)
| | | | | | - Zsolt Fabian
- School of Medicine and Dentistry, Faculty of Clinical and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UK; (M.M.E.-S.); (J.R.B.); (Y.L.)
| |
Collapse
|
6
|
Murciano-Goroff YR, Uppal M, Chen M, Harada G, Schram AM. Basket Trials: Past, Present, and Future. ANNUAL REVIEW OF CANCER BIOLOGY 2024; 8:59-80. [PMID: 38938274 PMCID: PMC11210107 DOI: 10.1146/annurev-cancerbio-061421-012927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
Abstract
Large-scale tumor molecular profiling has revealed that diverse cancer histologies are driven by common pathways with unifying biomarkers that can be exploited therapeutically. Disease-agnostic basket trials have been increasingly utilized to test biomarker-driven therapies across cancer types. These trials have led to drug approvals and improved the lives of patients while simultaneously advancing our understanding of cancer biology. This review focuses on the practicalities of implementing basket trials, with an emphasis on molecularly targeted trials. We examine the biologic subtleties of genomic biomarker and patient selection, discuss previous successes in drug development facilitated by basket trials, describe certain novel targets and drugs, and emphasize practical considerations for participant recruitment and study design. This review also highlights strategies for aiding patient access to basket trials. As basket trials become more common, steps to ensure equitable implementation of these studies will be critical for molecularly targeted drug development.
Collapse
Affiliation(s)
| | - Manik Uppal
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Monica Chen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Guilherme Harada
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alison M Schram
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
7
|
Shahjahan, Dey JK, Dey SK. Translational bioinformatics approach to combat cardiovascular disease and cancers. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 139:221-261. [PMID: 38448136 DOI: 10.1016/bs.apcsb.2023.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
Bioinformatics is an interconnected subject of science dealing with diverse fields including biology, chemistry, physics, statistics, mathematics, and computer science as the key fields to answer complicated physiological problems. Key intention of bioinformatics is to store, analyze, organize, and retrieve essential information about genome, proteome, transcriptome, metabolome, as well as organisms to investigate the biological system along with its dynamics, if any. The outcome of bioinformatics depends on the type, quantity, and quality of the raw data provided and the algorithm employed to analyze the same. Despite several approved medicines available, cardiovascular disorders (CVDs) and cancers comprises of the two leading causes of human deaths. Understanding the unknown facts of both these non-communicable disorders is inevitable to discover new pathways, find new drug targets, and eventually newer drugs to combat them successfully. Since, all these goals involve complex investigation and handling of various types of macro- and small- molecules of the human body, bioinformatics plays a key role in such processes. Results from such investigation has direct human application and thus we call this filed as translational bioinformatics. Current book chapter thus deals with diverse scope and applications of this translational bioinformatics to find cure, diagnosis, and understanding the mechanisms of CVDs and cancers. Developing complex yet small or long algorithms to address such problems is very common in translational bioinformatics. Structure-based drug discovery or AI-guided invention of novel antibodies that too with super-high accuracy, speed, and involvement of considerably low amount of investment are some of the astonishing features of the translational bioinformatics and its applications in the fields of CVDs and cancers.
Collapse
Affiliation(s)
- Shahjahan
- Laboratory for Structural Biology of Membrane Proteins, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, India
| | - Joy Kumar Dey
- Central Council for Research in Homoeopathy, Ministry of Ayush, Govt. of India, New Delhi, Delhi, India
| | - Sanjay Kumar Dey
- Laboratory for Structural Biology of Membrane Proteins, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, India.
| |
Collapse
|
8
|
Pandey P, Garg A, Singh V, Rai G, Mishra N. Clinical Trials and Future Prospects of Autophagy and ROS in Cancer. CANCER DRUG DISCOVERY AND DEVELOPMENT 2024:337-369. [DOI: 10.1007/978-3-031-66421-2_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
9
|
Pankiw M, Brezden-Masley C, Charames GS. Comprehensive genomic profiling for oncological advancements by precision medicine. Med Oncol 2023; 41:1. [PMID: 37993657 DOI: 10.1007/s12032-023-02228-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 10/25/2023] [Indexed: 11/24/2023]
Abstract
Considerable advancements in next generation sequencing (NGS) techniques have sparked the use of comprehensive genomic profiling (CGP) as a guiding tool for precision-centered oncological treatments. The past two decades have seen the completion of the human genome project, and the consequential invention of NGS. High-throughput sequencing technologies support the discovery and commonplace use of individualized cancer treatments, specifically immune-centered checkpoint inhibitor therapies, and oncogene and tumor suppressor gene targeted therapies. Nevertheless, CGP is not commonly used in all clinical settings. This review investigates the clinically relevant applications of CGP. Studies published between the years 2000-2023 have shown substantial evidence of the benefits of integrating CGP into routine care practice, while also making important comparisons to current-standard oncological treatment strategies. Findings of a comprehensive genomic profile includes predictive, prognostic, and diagnostic biomarkers, together with somatic mutation identification which can indicate the efficacy of immunotherapies and molecularly guided therapies. This review highlights the importance of CGP in identifying driver mutations in tumors that subsequently can be effectively targeted with molecular therapeutics and lead to drug discovery, allowing for increased precision in treating tumors selectively based on their specific genetic mutations, thereby improving patient outcomes.
Collapse
Affiliation(s)
- Maya Pankiw
- Department of Medicine, Mount Sinai Hospital, Toronto, ON, Canada
- Department of Pathology and Lab Medicine, Mount Sinai Hospital, Toronto, ON, Canada
| | - Christine Brezden-Masley
- Department of Medicine, Mount Sinai Hospital, Toronto, ON, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - George S Charames
- Department of Pathology and Lab Medicine, Mount Sinai Hospital, Toronto, ON, Canada.
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada.
- Department of Lab Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
- Mount Sinai Services, Toronto, ON, Canada.
| |
Collapse
|
10
|
Tateo V, Marchese PV, Mollica V, Massari F, Kurzrock R, Adashek JJ. Agnostic Approvals in Oncology: Getting the Right Drug to the Right Patient with the Right Genomics. Pharmaceuticals (Basel) 2023; 16:ph16040614. [PMID: 37111371 PMCID: PMC10144220 DOI: 10.3390/ph16040614] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/15/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
(1) Background: The oncology field has drastically changed with the advent of precision medicine, led by the discovery of druggable genes or immune targets assessed through next-generation sequencing. Biomarker-based treatments are increasingly emerging, and currently, six tissue-agnostic therapies are FDA-approved. (2) Methods: We performed a review of the literature and reported the trials that led to the approval of tissue-agnostic treatments and ongoing clinical trials currently investigating novel biomarker-based approaches. (3) Results: We discussed the approval of agnostic treatments: pembrolizumab and dostarlimab for MMRd/MSI-H, pembrolizumab for TMB-H, larotrectinib and entrectinib for NTRK-fusions, dabrafenib plus trametinib for BRAF V600E mutation, and selpercatinib for RET fusions. In addition, we reported novel clinical trials of biomarker-based approaches, including ALK, HER2, FGFR, and NRG1. (4) Conclusions: Precision medicine is constantly evolving, and with the improvement of diagnostic tools that allow a wider genomic definition of the tumor, tissue-agnostic targeted therapies are a promising treatment strategy tailored to the specific tumor genomic profile, leading to improved survival outcomes.
Collapse
Affiliation(s)
- Valentina Tateo
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Paola Valeria Marchese
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Veronica Mollica
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Francesco Massari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40127 Bologna, Italy
| | - Razelle Kurzrock
- MCW Cancer Center, Milwaukee, WI 53226, USA
- WIN Consortium, San Diego, CA 92093, USA
- Department of Oncology, University of Nebraska, Omaha, NE 68198, USA
| | - Jacob J Adashek
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, MD 21287, USA
| |
Collapse
|
11
|
Wang Z, Mehmood A, Yao J, Zhang H, Wang L, Al-Shehri M, Kaushik AC, Wei DQ. Combination of furosemide, gold, and dopamine as a potential therapy for breast cancer. Funct Integr Genomics 2023; 23:94. [PMID: 36943579 DOI: 10.1007/s10142-023-01007-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 02/28/2023] [Accepted: 03/01/2023] [Indexed: 03/23/2023]
Abstract
Breast cancer is one of the leading causes of death in women worldwide. Initially, it develops in the epithelium of the ducts or lobules of the breast glandular tissues with limited growth and the potential to metastasize. It is a highly heterogeneous malignancy; however, the common molecular mechanisms could help identify new targeted drugs for treating its subtypes. This study uses computational drug repositioning approaches to explore fresh drug candidates for breast cancer treatment. We also implemented reversal gene expression and gene expression-based signatures to explore novel drug candidates computationally. The drug activity profiles and related gene expression changes were acquired from the DrugBank, PubChem, and LINCS databases, and then in silico drug screening, molecular dynamics (MD) simulation, replica exchange MD simulations, and simulated annealing molecular dynamics (SAMD) simulations were conducted to discover and verify the valid drug candidates. We have found that compounds like furosemide, gold, and dopamine showed significant outcomes. Furthermore, the expression of genes related to breast cancer was observed to be reversed by these shortlisted drugs. Therefore, we postulate that combining furosemide, gold, and dopamine would be a potential combination therapy measurement for breast cancer patients.
Collapse
Affiliation(s)
- Zhen Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Aamir Mehmood
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Jia Yao
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Hui Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Li Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Mohammed Al-Shehri
- Department of Biology, Faculty of Science, King Khalid University, Abha, Saudi Arabia
| | | | - Dong-Qing Wei
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China.
- Zhongjing Research and Industrialization Institute of Chinese Medicine, Zhongguancun Scientific Park, Nanyang, Henan, China.
- Peng Cheng Laboratory, Nanshan District, Shenzhen, Guangdong, China.
| |
Collapse
|
12
|
Hegde M, Girisa S, Kunnumakkara AB. A compilation of bioinformatic approaches to identify novel downstream targets for the detection and prophylaxis of cancer. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 134:75-113. [PMID: 36858743 DOI: 10.1016/bs.apcsb.2022.11.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
The paradigm of cancer genomics has been radically changed by the development in next-generation sequencing (NGS) technologies making it possible to envisage individualized treatment based on tumor and stromal cells genome in a clinical setting within a short timeframe. The abundance of data has led to new avenues for studying coordinated alterations that impair biological processes, which in turn has increased the demand for bioinformatic tools for pathway analysis. While most of this work has been concentrated on optimizing certain algorithms to obtain quicker and more accurate results. Large volumes of these existing algorithm-based data are difficult for the biologists and clinicians to access, download and reanalyze them. In the present study, we have listed the bioinformatics algorithms and user-friendly graphical user interface (GUI) tools that enable code-independent analysis of big data without compromising the quality and time. We have also described the advantages and drawbacks of each of these platforms. Additionally, we emphasize the importance of creating new, more user-friendly solutions to provide better access to open data and talk about relevant problems like data sharing and patient privacy.
Collapse
Affiliation(s)
- Mangala Hegde
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Guwahati, Guwahati, Assam, India
| | - Sosmitha Girisa
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Guwahati, Guwahati, Assam, India
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Guwahati, Guwahati, Assam, India.
| |
Collapse
|
13
|
Establishment of four head and neck squamous cell carcinoma cell lines: importance of reference DNA for accurate genomic characterisation. J Laryngol Otol 2023; 137:301-307. [PMID: 35317874 PMCID: PMC9975763 DOI: 10.1017/s0022215122000846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
OBJECTIVE There is significant interest in developing early passage cell lines with matched normal reference DNA to facilitate a precision medicine approach in assessing drug response. This study aimed to establish early passage cell lines, and perform whole exome sequencing and short tandem repeat profiling on matched normal reference DNA, primary tumour and corresponding cell lines. METHODS A cell culture based, in vitro study was conducted of patients with primary human papillomavirus positive and human papillomavirus negative tumours. RESULTS Four early passage cell lines were established. Two cell lines were human papillomavirus positive, confirmed by sequencing and p16 immunoblotting. Short tandem repeat profiling confirmed that all cell lines were established from their index tumours. Whole exome sequencing revealed that the matched normal reference DNA was critical for accurate mutational analysis: a high rate of false positive mutation calls were excluded (87.6 per cent). CONCLUSION Early passage cell lines were successfully established. Patient-matched reference DNA is important for accurate cell line mutational calls.
Collapse
|
14
|
Cancer Biomarkers: Status and Its Future Direction. Indian J Surg 2023. [DOI: 10.1007/s12262-023-03723-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023] Open
|
15
|
Migliozzi S, Oh YT, Hasanain M, Garofano L, D'Angelo F, Najac RD, Picca A, Bielle F, Di Stefano AL, Lerond J, Sarkaria JN, Ceccarelli M, Sanson M, Lasorella A, Iavarone A. Integrative multi-omics networks identify PKCδ and DNA-PK as master kinases of glioblastoma subtypes and guide targeted cancer therapy. NATURE CANCER 2023; 4:181-202. [PMID: 36732634 PMCID: PMC9970878 DOI: 10.1038/s43018-022-00510-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 12/21/2022] [Indexed: 02/04/2023]
Abstract
Despite producing a panoply of potential cancer-specific targets, the proteogenomic characterization of human tumors has yet to demonstrate value for precision cancer medicine. Integrative multi-omics using a machine-learning network identified master kinases responsible for effecting phenotypic hallmarks of functional glioblastoma subtypes. In subtype-matched patient-derived models, we validated PKCδ and DNA-PK as master kinases of glycolytic/plurimetabolic and proliferative/progenitor subtypes, respectively, and qualified the kinases as potent and actionable glioblastoma subtype-specific therapeutic targets. Glioblastoma subtypes were associated with clinical and radiomics features, orthogonally validated by proteomics, phospho-proteomics, metabolomics, lipidomics and acetylomics analyses, and recapitulated in pediatric glioma, breast and lung squamous cell carcinoma, including subtype specificity of PKCδ and DNA-PK activity. We developed a probabilistic classification tool that performs optimally with RNA from frozen and paraffin-embedded tissues, which can be used to evaluate the association of therapeutic response with glioblastoma subtypes and to inform patient selection in prospective clinical trials.
Collapse
Affiliation(s)
- Simona Migliozzi
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY, USA.,Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Young Taek Oh
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY, USA.,Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Mohammad Hasanain
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY, USA.,Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Luciano Garofano
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY, USA.,Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Fulvio D'Angelo
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY, USA.,Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Ryan D Najac
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY, USA
| | - Alberto Picca
- AP-HP, Hôpital de la Pitié-Salpêtrière, Service de Neurologie 2, Paris, France.,Sorbonne Université, INSERM Unité 1127, CNRS UMR 7225, Paris Brain Institute, Equipe labellissée LNCC, Paris, France
| | - Franck Bielle
- Sorbonne Université, INSERM Unité 1127, CNRS UMR 7225, Paris Brain Institute, Equipe labellissée LNCC, Paris, France.,Department of Neuropathology, Pitié-Salpêtrière-Charles Foix, AP-HP, Paris, France
| | - Anna Luisa Di Stefano
- Sorbonne Université, INSERM Unité 1127, CNRS UMR 7225, Paris Brain Institute, Equipe labellissée LNCC, Paris, France.,Department of Neurology, Foch Hospital, Suresnes, Paris, France.,Neurosurgery Unit, Spedali Riuniti, Livorno, Italy
| | - Julie Lerond
- Sorbonne Université, INSERM Unité 1127, CNRS UMR 7225, Paris Brain Institute, Equipe labellissée LNCC, Paris, France
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - Michele Ceccarelli
- Department of Electrical Engineering and Information Technology (DIETI), University of Naples Federico II, Napoli, Italy.,BIOGEM Institute of Molecular Biology and Genetics, Via Camporeale, Ariano Irpino, Italy
| | - Marc Sanson
- AP-HP, Hôpital de la Pitié-Salpêtrière, Service de Neurologie 2, Paris, France.,Sorbonne Université, INSERM Unité 1127, CNRS UMR 7225, Paris Brain Institute, Equipe labellissée LNCC, Paris, France.,Onconeurotek Tumor Bank, Paris Brain Institute ICM, Paris, France
| | - Anna Lasorella
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY, USA. .,Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, FL, USA. .,Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA. .,Department of Pediatrics, Columbia University Medical Center, New York, NY, USA. .,Department of Biochemistry and Molecular Biology, University of Miami, Miller School of Medicine, Miami, FL, USA.
| | - Antonio Iavarone
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY, USA. .,Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, FL, USA. .,Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA. .,Department of Neurology, Columbia University Medical Center, New York, NY, USA. .,Department of Neurological Surgery, University of Miami, Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
16
|
Liu S, Takeda K, Rong A. An adaptive biomarker basket design in phase II oncology trials. Pharm Stat 2023; 22:128-142. [PMID: 36163614 DOI: 10.1002/pst.2264] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 07/30/2022] [Accepted: 09/09/2022] [Indexed: 02/01/2023]
Abstract
The phase II basket trial in oncology is a novel design that enables the simultaneous assessment of treatment effects of one anti-cancer targeted agent in multiple cancer types. Biomarkers could potentially associate with the clinical outcomes and re-define clinically meaningful treatment effects. It is therefore natural to develop a biomarker-based basket design to allow the prospective enrichment of the trials with the adaptive selection of the biomarker-positive (BM+) subjects who are most sensitive to the experimental treatment. We propose a two-stage phase II adaptive biomarker basket (ABB) design based on a potential predictive biomarker measured on a continuous scale. At Stage 1, the design incorporates a biomarker cutoff estimation procedure via a hierarchical Bayesian model with biomarker as a covariate (HBMbc). At Stage 2, the design enrolls only BM+ subjects, defined as those with the biomarker values exceeding the biomarker cutoff within each cancer type, and subsequently assesses the early efficacy and/or futility stopping through the pre-defined interim analyses. At the end of the trial, the response rate of all BM+ subjects for each cancer type can guide drug development, while the data from all subjects can be used to further model the relationship between the biomarker value and the clinical outcome for potential future research. The extensive simulation studies show that the ABB design could produce a good estimate of the biomarker cutoff to select BM+ subjects with high accuracy and could outperform the existing phase II basket biomarker cutoff design under various scenarios.
Collapse
Affiliation(s)
- Shufang Liu
- Data Science, Astellas Pharma Inc., Northbrook, Illinois, USA
| | - Kentaro Takeda
- Data Science, Astellas Pharma Inc., Northbrook, Illinois, USA
| | - Alan Rong
- Data Science, Astellas Pharma Inc., Northbrook, Illinois, USA
| |
Collapse
|
17
|
Molecular Characteristics and Prognostic Role of MFAP2 in Stomach Adenocarcinoma. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:1417238. [PMID: 35356627 PMCID: PMC8959993 DOI: 10.1155/2022/1417238] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 11/19/2021] [Indexed: 11/26/2022]
Abstract
Molecular characteristics and prognostic role of MFAP2 were by no means stated. The MFAP2 expression and prognostic prices in this study, with Cox analysis, was employed to develop a predictive fee for MFAP2. To know about coexpression and practical networks associated with MFAP2, LinkedOmics and GEPIA2 have been used. MFAP2 expression has been increased and verified in many unbiased coalitions in TCGA-STAD tumor tissues. In addition, in each TCGA and various cohorts, increased MFAP2 was linked with lower survival. Evaluation by Cox revealed the unbiased danger to average survival, disease-specific survival, and progression-free survival of STAD used to be due to the elevated expression of MFAP2. Active community assessed the MFAP2, through which more than a few cancer-associated kinases and E2F household pathways are regulated, which shows that MFAP2 affects RNA transportation, oocyte meiosis, spliceosome, and ribosome biogenesis. MFAP2 can predict and is linked to the prediction of STAD independently. The closure of the MFAP2 link to the macrophage marker genes is, in particular, the achievable core of immune response.
Collapse
|
18
|
Chiang CC, Lin GL, Yang SY, Tu CW, Huang WL, Wei CF, Wang FC, Lin PJ, Huang WH, Chuang YM, Lee YT, Yeh CC, Chan M, Hsu YC. PCDHB15 as a potential tumor suppressor and epigenetic biomarker for breast cancer. Oncol Lett 2022; 23:117. [PMID: 35261631 PMCID: PMC8855166 DOI: 10.3892/ol.2022.13237] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 01/07/2022] [Indexed: 11/05/2022] Open
Abstract
Breast cancer is among the most frequently diagnosed cancer types and the leading cause of cancer-related death in women. The mortality rate of patients with breast cancer is currently increasing, perhaps due to a lack of early screening tools. In the present study, using The Cancer Genome Atlas (TCGA) breast cancer dataset (n=883), it was determined that methylation of the protocadherin β15 (PCDHB15) promoter was higher in breast cancer samples than that in normal tissues. A negative association between promoter methylation and expression of PCDHB15 was observed in the TCGA dataset and breast cancer cell lines. In TCGA cohort, lower PCDHB15 expression was associated with shorter relapse-free survival times. Treatment with the DNA methyltransferase inhibitor restored PCDHB15 expression in a breast cancer cell line; however, overexpression of PCDHB15 was shown to suppress colony formation. PCDHB15 methylation detected in circulating cell-free DNA (cfDNA) isolated from serum samples was higher in patients with breast cancer (40.8%) compared with that in patients with benign tumors (22.4%). PCDHB15 methylation was not correlated with any clinical parameters. Taken together, PCDHB15 is a potential tumor suppressor in cases of breast cancer, which can be epigenetically silenced via promoter methylation. PCDHB15 methylation using cfDNA is a novel minimally invasive epigenetic biomarker for the diagnosis and prognosis of breast cancer.
Collapse
Affiliation(s)
- Ching-Chung Chiang
- Department of Surgery, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi 60002, Taiwan, R.O.C
| | - Guan-Ling Lin
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi 62101, Taiwan, R.O.C
| | - Shu-Yi Yang
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi 62101, Taiwan, R.O.C
| | - Chi-Wen Tu
- Department of Surgery, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi 60002, Taiwan, R.O.C
| | - Wen-Long Huang
- Department of Chinese Medicine, Dalin Tzuchi Hospital, The Buddhist Tzuchi Medical Foundation, Chiayi 62247, Taiwan, R.O.C
| | - Chun-Feng Wei
- Department of Surgery, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi 60002, Taiwan, R.O.C
| | - Feng-Chi Wang
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi 62101, Taiwan, R.O.C
| | - Pin-Ju Lin
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi 62101, Taiwan, R.O.C
| | - Wan-Hong Huang
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi 62101, Taiwan, R.O.C
| | - Yu-Ming Chuang
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi 62101, Taiwan, R.O.C
| | - Yu-Ting Lee
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi 62101, Taiwan, R.O.C
| | - Chia-Chou Yeh
- Department of Chinese Medicine, Dalin Tzuchi Hospital, The Buddhist Tzuchi Medical Foundation, Chiayi 62247, Taiwan, R.O.C
| | - Michael Chan
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi 62101, Taiwan, R.O.C
| | - Yu-Chen Hsu
- Department of Surgery, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi 60002, Taiwan, R.O.C
| |
Collapse
|
19
|
Prabahar A. Integration of Transcriptomics Data and Metabolomic Data Using Biomedical Literature Mining and Pathway Analysis. Methods Mol Biol 2022; 2496:301-316. [PMID: 35713871 DOI: 10.1007/978-1-0716-2305-3_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Recent progress in omics technologies such as transcriptomics and metabolomics offers an unprecedented opportunity to understand the disease mechanisms and determines the associated biomedical entities using biomedical literature mining. Tremendous data available in the biomedical literature helps in addressing complex biomedical problems. Advancements in genomics and transcriptomics helps in decoding the genetic information obtained from various high throughput techniques for its use in personalized medicine and therapeutics. Integration of data from biomedical literature and data from large-scale genomic studies aids in the determination of the etiology of a disease and drug targets. This chapter addresses the perspectives of transcriptomics and metabolomics in biomedical literature mining and gives an overview of state-of-the-art techniques in this field.
Collapse
Affiliation(s)
- Archana Prabahar
- R&D Division, Eriks-Precision Components India Pvt Ltd, Mohali, Punjab, India.
| |
Collapse
|
20
|
Takeda K, Liu S, Rong A. Constrained hierarchical Bayesian model for latent subgroups in basket trials with two classifiers. Stat Med 2021; 41:298-309. [PMID: 34697822 DOI: 10.1002/sim.9237] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 10/10/2021] [Accepted: 10/12/2021] [Indexed: 02/01/2023]
Abstract
The basket trial in oncology is a novel clinical trial design that enables the simultaneous assessment of one treatment in multiple cancer types. In addition to the usual basket classifier of the cancer types, many recent basket trials further contain other classifiers like biomarkers that potentially affect the clinical outcomes. In other words, the treatment effects in those baskets are often categorized by not only the cancer types but also the levels of other classifiers. Therefore, the assumption of exchangeability is often violated when some baskets are more sensitive to the targeted treatment, whereas others are less. In this article, we propose a constrained hierarchical Bayesian model for latent subgroups (CHBM-LS) to deal with potential heterogeneity of treatment effects due to both the cancer type (first classifier) and another classifier (second classifier) in basket trials. Different baskets defined by multiple cancer types and multiple levels of the second classifier are aggregated into subgroups using a latent subgroup modeling approach. Within each latent subgroup, the treatment effects are similar and approximately exchangeable to borrow information. The CHBM-LS approach evaluates the treatment effect for each basket while allowing adaptive information borrowing across the baskets by identifying latent subgroups. The simulation study shows that the CHBM-LS approach outperforms other approaches with higher statistical power and better-controlled type I error rates under various scenarios with heterogeneous treatment effects across baskets.
Collapse
Affiliation(s)
- Kentaro Takeda
- Data Science, Astellas Pharma Global Development, Inc., Northbrook, Illinois, USA
| | - Shufang Liu
- Data Science, Astellas Pharma Global Development, Inc., Northbrook, Illinois, USA
| | - Alan Rong
- Data Science, Astellas Pharma Global Development, Inc., Northbrook, Illinois, USA
| |
Collapse
|
21
|
Thirunavukarasu D, Cheng LY, Song P, Chen SX, Borad MJ, Kwong L, James P, Turner DJ, Zhang DY. Oncogene Concatenated Enriched Amplicon Nanopore Sequencing for rapid, accurate, and affordable somatic mutation detection. Genome Biol 2021; 22:227. [PMID: 34482832 PMCID: PMC8419911 DOI: 10.1186/s13059-021-02449-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 08/02/2021] [Indexed: 12/13/2022] Open
Abstract
We develop the Oncogene Concatenated Enriched Amplicon Nanopore Sequencing (OCEANS) method, in which variants with low variant allele frequency (VAFs) are amplified and subsequently concatenated for Nanopore Sequencing. OCEANS allows accurate detection of somatic mutations with VAF limits of detection between 0.05 and 1%. We construct 4 distinct multi-gene OCEANS panels targeting recurrent mutations in acute myeloid leukemia, melanoma, non-small- cell lung cancer, and hepatocellular carcinoma and validate them on clinical samples. By demonstrating detection of low VAF single nucleotide variant mutations using Nanopore Sequencing, OCEANS is poised to enable same-day clinical sequencing panels.
Collapse
Affiliation(s)
| | - Lauren Y Cheng
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Ping Song
- Department of Bioengineering, Rice University, Houston, TX, USA
| | | | | | - Lawrence Kwong
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - David Yu Zhang
- NuProbe USA Inc, Houston, TX, USA.
- Department of Bioengineering, Rice University, Houston, TX, USA.
| |
Collapse
|
22
|
Rapid single-molecule digital detection of protein biomarkers for continuous monitoring of systemic immune disorders. Blood 2021; 137:1591-1602. [PMID: 33275650 DOI: 10.1182/blood.2019004399] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 11/24/2020] [Indexed: 12/12/2022] Open
Abstract
Digital protein assays have great potential to advance immunodiagnostics because of their single-molecule sensitivity, high precision, and robust measurements. However, translating digital protein assays to acute clinical care has been challenging because it requires deployment of these assays with a rapid turnaround. Herein, we present a technology platform for ultrafast digital protein biomarker detection by using single-molecule counting of immune-complex formation events at an early, pre-equilibrium state. This method, which we term "pre-equilibrium digital enzyme-linked immunosorbent assay" (PEdELISA), can quantify a multiplexed panel of protein biomarkers in 10 µL of serum within an unprecedented assay incubation time of 15 to 300 seconds over a 104 dynamic range. PEdELISA allowed us to perform rapid monitoring of protein biomarkers in patients manifesting post-chimeric antigen receptor T-cell therapy cytokine release syndrome, with ∼30-minute sample-to-answer time and a sub-picograms per mL limit of detection. The rapid, sensitive, and low-input volume biomarker quantification enabled by PEdELISA is broadly applicable to timely monitoring of acute disease, potentially enabling more personalized treatment.
Collapse
|
23
|
Noor F, Noor A, Ishaq AR, Farzeen I, Saleem MH, Ghaffar K, Aslam MF, Aslam S, Chen JT. Recent Advances in Diagnostic and Therapeutic Approaches for Breast Cancer: A Comprehensive Review. Curr Pharm Des 2021; 27:2344-2365. [PMID: 33655849 DOI: 10.2174/1381612827666210303141416] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 01/22/2021] [Indexed: 11/22/2022]
Abstract
A silent monster, breast cancer, is a challenging medical task for researchers. Breast cancer is a leading cause of death in women with respect to other cancers. A case of breast cancer is diagnosed among women every 19 seconds, and every 74 seconds, a woman dies of breast cancer somewhere in the world. Several risk factors, such as genetic and environmental factors, favor breast cancer development. This review tends to provide deep insights regarding the genetics of breast cancer along with multiple diagnostic and therapeutic approaches as problem-solving negotiators to prevent the progression of breast cancer. This assembled data mainly aims to discuss omics-based approaches to provide enthralling diagnostic biomarkers and emerging novel therapies to combat breast cancer. This review article intends to pave a new path for the discovery of effective treatment options.
Collapse
Affiliation(s)
- Fatima Noor
- Department of Bioinformatics and Biotechnology, Government College University Allama Iqbal Road, 38000 Faisalabad, Pakistan
| | - Ayesha Noor
- Department of Zoology, Government College University Allama Iqbal Road, 38000 Faisalabad, Pakistan
| | - Ali Raza Ishaq
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Environmental Microbial Technology Center of Hubei Province, College of Life Science, Hubei University, Wuhan 430062, China
| | - Iqra Farzeen
- Department of Zoology, Government College University Allama Iqbal Road, 38000 Faisalabad, Pakistan
| | - Muhammad Hamzah Saleem
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Environmental Microbial Technology Center of Hubei Province, College of Life Science, Hubei University, Wuhan 430062, China
| | - Kanwal Ghaffar
- Department of Zoology, Government College University Allama Iqbal Road, 38000 Faisalabad, Pakistan
| | - Muhammad Farhan Aslam
- MOA Key Laboratory of Crop Ecophysiology and Farming System in the Middle Reaches of the Yangtze River, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Sidra Aslam
- Department of Bioinformatics and Biotechnology, Government College University Allama Iqbal Road, 38000 Faisalabad, Pakistan
| | - Jen-Tsung Chen
- Department of Life Sciences, National University of Kaohsiung, Kaohsiung 811, China
| |
Collapse
|
24
|
Di Capua D, Bracken-Clarke D, Ronan K, Baird AM, Finn S. The Liquid Biopsy for Lung Cancer: State of the Art, Limitations and Future Developments. Cancers (Basel) 2021; 13:cancers13163923. [PMID: 34439082 PMCID: PMC8391249 DOI: 10.3390/cancers13163923] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary During the development and progression of lung tumors, processes such as necrosis and vascular invasion shed tumor cells or cellular components into various fluid compartments. Liquid biopsies consist of obtaining a bodily fluid, typically peripheral blood, in order to isolate and investigate these shed tumor constituents. Circulating tumor cells (CTCs) are one such constituent, which can be isolated from blood and can act as a diagnostic aid and provide valuable prognostic information. Liquid-based biopsies may also have a potential future role in lung cancer screening. Circulating tumor DNA (ctDNA) is found in small quantities in blood and, with the recent development of sensitive molecular and sequencing technologies, can be used to directly detect actionable genetic alterations or monitor for resistance mutations and guide clinical management. While potential benefits of liquid biopsies are promising, they are not without limitations. In this review, we summarize the current state and limitations of CTCs and ctDNA and possible future directions. Abstract Lung cancer is a leading cause of cancer-related deaths, contributing to 18.4% of cancer deaths globally. Treatment of non-small cell lung carcinoma has seen rapid progression with targeted therapies tailored to specific genetic drivers. However, identifying genetic alterations can be difficult due to lack of tissue, inaccessible tumors and the risk of complications for the patient with serial tissue sampling. The liquid biopsy provides a minimally invasive method which can obtain circulating biomarkers shed from the tumor and could be a safer alternative to tissue biopsy. While tissue biopsy remains the gold standard, liquid biopsies could be very beneficial where serial sampling is required, such as monitoring disease progression or development of resistance mutations to current targeted therapies. Liquid biopsies also have a potential role in identifying patients at risk of relapse post treatment and as a component of future lung cancer screening protocols. Rapid developments have led to multiple platforms for isolating circulating tumor cells (CTCs) and detecting circulating tumor DNA (ctDNA); however, standardization is lacking, especially in lung carcinoma. Additionally, clonal hematopoiesis of uncertain clinical significance must be taken into consideration in genetic sequencing, as it introduces the potential for false positives. Various biomarkers have been investigated in liquid biopsies; however, in this review, we will concentrate on the current use of ctDNA and CTCs, focusing on the clinical relevance, current and possible future applications and limitations of each.
Collapse
Affiliation(s)
- Daniel Di Capua
- Department of Histopathology, St. James’s Hospital, D08NHY1 Dublin, Ireland;
| | - Dara Bracken-Clarke
- Department of Medical Oncology, St. James’ Hospital, D08NHY1 Dublin, Ireland;
| | - Karine Ronan
- Faculty of Medicine, University College Dublin, D04V1W8 Dublin, Ireland;
| | - Anne-Marie Baird
- School of Medicine, Trinity Translational Medicine Institute, Trinity College, D02PN40 Dublin, Ireland;
| | - Stephen Finn
- Department of Histopathology, St. James’s Hospital, D08NHY1 Dublin, Ireland;
- Correspondence:
| |
Collapse
|
25
|
Kordbacheh F, Farah CS. Molecular Pathways and Druggable Targets in Head and Neck Squamous Cell Carcinoma. Cancers (Basel) 2021; 13:3453. [PMID: 34298667 PMCID: PMC8307423 DOI: 10.3390/cancers13143453] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/02/2021] [Accepted: 07/08/2021] [Indexed: 12/30/2022] Open
Abstract
Head and neck cancers are a heterogeneous group of neoplasms, affecting an ever increasing global population. Despite advances in diagnostic technology and surgical approaches to manage these conditions, survival rates have only marginally improved and this has occurred mainly in developed countries. Some improvements in survival, however, have been a result of new management and treatment approaches made possible because of our ever-increasing understanding of the molecular pathways triggered in head and neck oncogenesis, and the growing understanding of the abundant heterogeneity of this group of cancers. Some important pathways are common to other solid tumours, but their impact on reducing the burden of head and neck disease has been less than impressive. Other less known and little-explored pathways may hold the key to the development of potential druggable targets. The extensive work carried out over the last decade, mostly utilising next generation sequencing has opened up the development of many novel approaches to head and neck cancer treatment. This paper explores our current understanding of the molecular pathways of this group of tumours and outlines associated druggable targets which are deployed as therapeutic approaches in head and neck oncology with the ultimate aim of improving patient outcomes and controlling the personal and economic burden of head and neck cancer.
Collapse
Affiliation(s)
- Farzaneh Kordbacheh
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA;
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, Australian National University, Canberra, ACT 0200, Australia
| | - Camile S. Farah
- The Australian Centre for Oral Oncology Research & Education, Perth, WA 6009, Australia
- Genomics for Life, Brisbane, QLD 4064, Australia
- Anatomical Pathology, Australian Clinical Labs, Subiaco, WA 6008, Australia
- Peter MacCallum Cancer Centre, Head and Neck Cancer Signalling Laboratory, Melbourne, VIC 3000, Australia
| |
Collapse
|
26
|
Shiri I, Maleki H, Hajianfar G, Abdollahi H, Ashrafinia S, Hatt M, Zaidi H, Oveisi M, Rahmim A. Next-Generation Radiogenomics Sequencing for Prediction of EGFR and KRAS Mutation Status in NSCLC Patients Using Multimodal Imaging and Machine Learning Algorithms. Mol Imaging Biol 2021; 22:1132-1148. [PMID: 32185618 DOI: 10.1007/s11307-020-01487-8] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE Considerable progress has been made in the assessment and management of non-small cell lung cancer (NSCLC) patients based on mutation status in the epidermal growth factor receptor (EGFR) and Kirsten rat sarcoma viral oncogene (KRAS). At the same time, NSCLC management through KRAS and EGFR mutation profiling faces challenges. In the present work, we aimed to evaluate a comprehensive radiomics framework that enabled prediction of EGFR and KRAS mutation status in NSCLC patients based on radiomic features from low-dose computed tomography (CT), contrast-enhanced diagnostic quality CT (CTD), and positron emission tomography (PET) imaging modalities and use of machine learning algorithms. METHODS Our study involved NSCLC patients including 150 PET, low-dose CT, and CTD images. Radiomic features from original and preprocessed (including 64 bin discretizing, Laplacian-of-Gaussian (LOG), and Wavelet) images were extracted. Conventional clinically used standard uptake value (SUV) parameters and metabolic tumor volume (MTV) were also obtained from PET images. Highly correlated features were pre-eliminated, and false discovery rate (FDR) correction was performed with the resulting q-values reported for univariate analysis. Six feature selection methods and 12 classifiers were then used for multivariate prediction of gene mutation status (provided by polymerase chain reaction (PCR)) in patients. We performed 10-fold cross-validation for model tuning to improve robustness, and our developed models were assessed on an independent validation set with 68 patients (common in all three imaging modalities). The average area under the receiver operator characteristic curve (AUC) was utilized for performance evaluation. RESULTS The best predictive power for conventional PET parameters was achieved by SUVpeak (AUC 0.69, p value = 0.0002) and MTV (AUC 0.55, p value = 0.0011) for EGFR and KRAS, respectively. Univariate analysis of extracted radiomics features improved AUC performance to 0.75 (q-value 0.003, Short-Run Emphasis feature of GLRLM from LOG preprocessed image of PET with sigma value 1.5) and 0.71 (q-value 0.00005, Large Dependence Low Gray-Level Emphasis feature of GLDM in LOG preprocessed image of CTD with sigma value 5) for EGFR and KRAS, respectively. Furthermore, multivariate machine learning-based AUC performances were significantly improved to 0.82 for EGFR (LOG preprocessed image of PET with sigma 3 with variance threshold (VT) feature selector and stochastic gradient descent (SGD) classifier (q-value = 4.86E-05) and 0.83 for KRAS (LOG preprocessed image of CT with sigma 3.5 with select model (SM) feature selector and SGD classifier (q-value = 2.81E-09). CONCLUSION Our work demonstrated that non-invasive and reliable radiomics analysis can be successfully used to predict EGFR and KRAS mutation status in NSCLC patients. We demonstrated that radiomic features extracted from different image-feature sets could be used for EGFR and KRAS mutation status prediction in NSCLC patients and showed improved predictive power relative to conventional image-derived metrics.
Collapse
Affiliation(s)
- Isaac Shiri
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211, Geneva 4, Switzerland
| | - Hasan Maleki
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Science, Tehran, Iran.,Department of Computer Science and Engineering, Shahid Beheshti University, Tehran, Iran
| | - Ghasem Hajianfar
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211, Geneva 4, Switzerland
| | - Hamid Abdollahi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211, Geneva 4, Switzerland.,Department of Radiologic Sciences and Medical Physics, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Saeed Ashrafinia
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, USA.,Department of Electrical and Computer Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Mathieu Hatt
- LaTIM, INSERM, UMR 1101, Univ Brest, Brest, France
| | - Habib Zaidi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211, Geneva 4, Switzerland.,Geneva University Neurocenter, Geneva University, Geneva, Switzerland.,Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, Netherlands.,Department of Nuclear Medicine, University of Southern Denmark, Odense, Denmark
| | - Mehrdad Oveisi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Science, Tehran, Iran.,Department of Computer Science, University of British Columbia, Vancouver, BC, Canada
| | - Arman Rahmim
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, USA. .,Departments of Radiology and Physics, University of British Columbia, Vancouver, BC, Canada. .,Department of Integrative Oncology, BC Cancer Research Centre, 675 West 10th Ave, Vancouver, BC, V5Z 1L3, Canada.
| |
Collapse
|
27
|
D’Abbronzo G, Franco R. The changing role of the pathologist in the era of targeted therapy in personalized medicine. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2021. [DOI: 10.1080/23808993.2021.1923400] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Giuseppe D’Abbronzo
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Renato Franco
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| |
Collapse
|
28
|
Scopetti D, Piobbico D, Brunacci C, Pieroni S, Bellezza G, Castelli M, Ludovini V, Tofanetti FR, Cagini L, Sidoni A, Puxeddu E, Della-Fazia MA, Servillo G. INSL4 as prognostic marker for proliferation and invasiveness in Non-Small-Cell Lung Cancer. J Cancer 2021; 12:3781-3795. [PMID: 34093787 PMCID: PMC8176261 DOI: 10.7150/jca.51332] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 02/25/2021] [Indexed: 12/24/2022] Open
Abstract
Non-small-cell-lung cancer accounts for 80-85% of all forms of lung cancer as leading cause of cancer-related death in human. Despite remarkable advances in the diagnosis and therapy of lung cancer, no significant improvements have thus far been achieved in terms of patients' prognosis. Here, we investigated the role of INSL4 - a member of the relaxin-family - in NSCLC. We overexpressed INSL4 in NSCLC cells to analyse in vitro the growth rate and the tumourigenic features. We investigated the signalling pathways engaged in INSL4 overexpressing cells and the tumour growth ability by studying the tumour development in a patient derived tumour xenograft mouse model. We found an INSL4 cell growth promoting effect in vitro in H1299 cells and in vivo in NOD/SCID mice. Surprisingly, in NSCLC-A549 cells, INSL4 overexpression has not similar effect, despite huge basal INSL4-mRNA expression respect to H1299. The INSL4-mRNA analysis of eight different NSCLC-derived cell lines, revealed highly difference in the INSL4-mRNA amount. Transfection of NSCLC lines with INSL4-Myc showed huge level of INSL4-mRNA with a very low amount of protein expressed. Notably, similar discrepancy has been observed in NSCLC patients. However, in a cohort of NSCLC patients analysing a database, we found a significant inverse correlation between INSL4 expression and Overall Survival. By combining the in vitro and in vivo results, suggest that in patients whose NSCLC adenocarcinoma spontaneously expressed high levels of INSL4 post-transcriptional modifications affecting INSL4 do not allow to assess precision therapy in selected patients without consider protein INSL4 amount.
Collapse
Affiliation(s)
- Damiano Scopetti
- Department of Medicine and Surgery, Section of General Pathology, University of Perugia, Perugia- Italy
| | - Danilo Piobbico
- Department of Medicine and Surgery, Section of General Pathology, University of Perugia, Perugia- Italy
| | - Cinzia Brunacci
- Department of Medicine and Surgery, Section of General Pathology, University of Perugia, Perugia- Italy
| | - Stefania Pieroni
- Department of Medicine and Surgery, Section of General Pathology, University of Perugia, Perugia- Italy
| | - Guido Bellezza
- Department of Medicine and Surgery, Section of Anatomic Pathology and Histology, University of Perugia, Perugia- Italy
| | - Marilena Castelli
- Department of Medicine and Surgery, Section of General Pathology, University of Perugia, Perugia- Italy
| | - Vienna Ludovini
- Medical Oncology, S. Maria Della Misericordia Hospital, Perugia, Italy
| | | | - Lucio Cagini
- Department of Medicine and Surgery, Section of internal medicine, angiology and atherosclerosis diseases
| | - Angelo Sidoni
- Department of Medicine and Surgery, Section of Anatomic Pathology and Histology, University of Perugia, Perugia- Italy
| | - Efisio Puxeddu
- Department of Medicine and Surgery, Section of internal medicine and endocrine and metabolic sciences
| | - Maria Agnese Della-Fazia
- Department of Medicine and Surgery, Section of General Pathology, University of Perugia, Perugia- Italy
| | - Giuseppe Servillo
- Department of Medicine and Surgery, Section of General Pathology, University of Perugia, Perugia- Italy
| |
Collapse
|
29
|
PharmaKU: A Web-Based Tool Aimed at Improving Outreach and Clinical Utility of Pharmacogenomics. J Pers Med 2021; 11:jpm11030210. [PMID: 33809530 PMCID: PMC7998233 DOI: 10.3390/jpm11030210] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/03/2021] [Accepted: 03/05/2021] [Indexed: 12/15/2022] Open
Abstract
With the tremendous advancements in genome sequencing technology in the field of pharmacogenomics, data have to be made accessible to be more efficiently utilized by broader clinical disciplines. Physicians who require the drug–genome interactome information, have been challenged by the complicated pharmacogenomic star-based classification system. We present here an end-to-end web-based pharmacogenomics tool, PharmaKU, which has a comprehensive easy-to-use interface. PharmaKU can help to overcome several hurdles posed by previous pharmacogenomics tools, including input in hg38 format only, while hg19/GRCh37 is now the most popular reference genome assembly among clinicians and geneticists, as well as the lack of clinical recommendations and other pertinent dosage-related information. This tool extracts genetic variants from nine well-annotated pharmacogenes (for which diplotype to phenotype information is available) from whole genome variant files and uses Stargazer software to assign diplotypes and apply prescribing recommendations from pharmacogenomic resources. The tool is wrapped with a user-friendly web interface, which allows for choosing hg19 or hg38 as the reference genome version and reports results as a comprehensive PDF document. PharmaKU is anticipated to enable bench to bedside implementation of pharmacogenomics knowledge by bringing precision medicine closer to a clinical reality.
Collapse
|
30
|
Batalha S, Ferreira S, Brito C. The Peripheral Immune Landscape of Breast Cancer: Clinical Findings and In Vitro Models for Biomarker Discovery. Cancers (Basel) 2021; 13:1305. [PMID: 33804027 PMCID: PMC8001103 DOI: 10.3390/cancers13061305] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/09/2021] [Accepted: 03/12/2021] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is the deadliest female malignancy worldwide and, while much is known about phenotype and function of infiltrating immune cells, the same attention has not been paid to the peripheral immune compartment of breast cancer patients. To obtain faster, cheaper, and more precise monitoring of patients' status, it is crucial to define and analyze circulating immune profiles. This review compiles and summarizes the disperse knowledge on the peripheral immune profile of breast cancer patients, how it departs from healthy individuals and how it changes with disease progression. We propose this data to be used as a starting point for validation of clinically relevant biomarkers of disease progression and therapy response, which warrants more thorough investigation in patient cohorts of specific breast cancer subtypes. Relevant clinical findings may also be explored experimentally using advanced 3D cellular models of human cancer-immune system interactions, which are under intensive development. We review the latest findings and discuss the strengths and limitations of such models, as well as the future perspectives. Together, the scientific advancement of peripheral biomarker discovery and cancer-immune crosstalk in breast cancer will be instrumental to uncover molecular mechanisms and putative biomarkers and drug targets in an all-human setting.
Collapse
Affiliation(s)
- Sofia Batalha
- Instituto de Biologia Experimental e Tecnológica (iBET), Apartado 12, 2781-901 Oeiras, Portugal;
- Instituto de Tecnologia Química e Biológica António Xavier, University Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal
| | - Sofia Ferreira
- Instituto Português de Oncologia de Lisboa Francisco Gentil, Rua Prof Lima Basto, 1099-023 Lisboa, Portugal;
| | - Catarina Brito
- Instituto de Biologia Experimental e Tecnológica (iBET), Apartado 12, 2781-901 Oeiras, Portugal;
- Instituto de Tecnologia Química e Biológica António Xavier, University Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal
| |
Collapse
|
31
|
Lin R, Thall PF, Yuan Y. A Phase I-II Basket Trial Design to Optimize Dose-Schedule Regimes Based on Delayed Outcomes. BAYESIAN ANALYSIS 2021; 16:179-202. [PMID: 34267857 PMCID: PMC8277108 DOI: 10.1214/20-ba1205] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
This paper proposes a Bayesian adaptive basket trial design to optimize the dose-schedule regimes of an experimental agent within disease subtypes, called "baskets", for phase I-II clinical trials based on late-onset efficacy and toxicity. To characterize the association among the baskets and regimes, a Bayesian hierarchical model is assumed that includes a heterogeneity parameter, adaptively updated during the trial, that quantifies information shared across baskets. To account for late-onset outcomes when doing sequential decision making, unobserved outcomes are treated as missing values and imputed by exploiting early biomarker and low-grade toxicity information. Elicited joint utilities of efficacy and toxicity are used for decision making. Patients are randomized adaptively to regimes while accounting for baskets, with randomization probabilities proportional to the posterior probability of achieving maximum utility. Simulations are presented to assess the design's robustness and ability to identify optimal dose-schedule regimes within disease subtypes, and to compare it to a simplified design that treats the subtypes independently.
Collapse
Affiliation(s)
- Ruitao Lin
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, U.S.A
| | - Peter F Thall
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, U.S.A
| | - Ying Yuan
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, U.S.A
| |
Collapse
|
32
|
Fan X, Luo G, Huang YS. Accucopy: accurate and fast inference of allele-specific copy number alterations from low-coverage low-purity tumor sequencing data. BMC Bioinformatics 2021; 22:23. [PMID: 33451280 PMCID: PMC7811225 DOI: 10.1186/s12859-020-03924-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 12/07/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Copy number alterations (CNAs), due to their large impact on the genome, have been an important contributing factor to oncogenesis and metastasis. Detecting genomic alterations from the shallow-sequencing data of a low-purity tumor sample remains a challenging task. RESULTS We introduce Accucopy, a method to infer total copy numbers (TCNs) and allele-specific copy numbers (ASCNs) from challenging low-purity and low-coverage tumor samples. Accucopy adopts many robust statistical techniques such as kernel smoothing of coverage differentiation information to discern signals from noise and combines ideas from time-series analysis and the signal-processing field to derive a range of estimates for the period in a histogram of coverage differentiation information. Statistical learning models such as the tiered Gaussian mixture model, the expectation-maximization algorithm, and sparse Bayesian learning were customized and built into the model. Accucopy is implemented in C++ /Rust, packaged in a docker image, and supports non-human samples, more at http://www.yfish.org/software/ . CONCLUSIONS We describe Accucopy, a method that can predict both TCNs and ASCNs from low-coverage low-purity tumor sequencing data. Through comparative analyses in both simulated and real-sequencing samples, we demonstrate that Accucopy is more accurate than Sclust, ABSOLUTE, and Sequenza.
Collapse
Affiliation(s)
- Xinping Fan
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Guanghao Luo
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Yu S Huang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
33
|
Yoon AJ, Santella RM, Wang S, Kutler DI, Carvajal RD, Philipone E, Wang T, Peters SM, Stewart CR, Momen-Heravi F, Troob S, Levin M, AkhavanAghdam Z, Shackelford AJ, Canterbury CR, Shimonosono M, Hernandez BY, McDowell BD, Nakagawa H. MicroRNA-Based Cancer Mortality Risk Scoring System and hTERT Expression in Early-Stage Oral Squamous Cell Carcinoma. JOURNAL OF ONCOLOGY 2021; 2021:8292453. [PMID: 33510789 PMCID: PMC7822680 DOI: 10.1155/2021/8292453] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 12/01/2020] [Accepted: 12/14/2020] [Indexed: 11/17/2022]
Abstract
We have previously constructed a novel microRNA (miRNA)-based prognostic model and cancer-specific mortality risk score formula to predict survival outcome in oral squamous cell carcinoma (OSCC) patients who are already categorized into "early-stage" by the TNM staging system. A total of 836 early-stage OSCC patients were assigned the mortality risk scores. We evaluated the efficacy of various treatment regimens in terms of survival benefit compared to surgery only in patients stratified into high (risk score ≥0) versus low (risk score <0) mortality risk categories. For the high-risk group, surgery with neck dissection significantly improved the 5-year survival to 75% from 46% with surgery only (p < 0.001); a Cox proportional hazard model on time-to-death demonstrated a hazard ratio of 0.37 for surgery with neck dissection (95% CI: 0.2-0.6; p=0.0005). For the low-risk group, surgery only was the treatment of choice associated with 5-year survival benefit. Regardless of treatment selected, those with risk score ≥2 may benefit from additional therapy to prevent cancer relapse. We also identified hTERT (human telomerase reverse transcriptase) as a gene target common to the prognostic miRNAs. There was 22-fold increase in the hTERT expression level in patients with risk score ≥2 compared to healthy controls (p < 0.0005). Overexpression of hTERT was also observed in the patient-derived OSCC organoid compared to that of normal organoid. The DNA cancer vaccine that targets hTERT-expressing cells currently undergoing rigorous clinical evaluation for other tumors can be repurposed to prevent cancer recurrence in these high-risk early-stage oral cancer patients.
Collapse
Affiliation(s)
- Angela J. Yoon
- Columbia University Irving Medical Center, New York, NY, USA
| | | | - Shuang Wang
- Columbia University Mailman School of Public Health, New York, NY, USA
| | | | | | | | - Tian Wang
- Columbia University Mailman School of Public Health, New York, NY, USA
| | - Scott M. Peters
- Columbia University Irving Medical Center, New York, NY, USA
| | | | | | - Scott Troob
- Columbia University Irving Medical Center, New York, NY, USA
| | | | | | | | | | | | - Brenda Y. Hernandez
- Hawaii Tumor Registry, University of Hawaii Cancer Center, Honolulu, HI, USA
| | | | | |
Collapse
|
34
|
Li C, Kasinski AL. InVivo Cancer-Based Functional Genomics. Trends Cancer 2020; 6:1002-1017. [PMID: 32828714 DOI: 10.1016/j.trecan.2020.07.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 07/13/2020] [Accepted: 07/20/2020] [Indexed: 12/18/2022]
Abstract
Pinpointing the underlying mechanisms that drive tumorigenesis in human patients is a prerequisite for identifying suitable therapeutic targets for precision medicine. In contrast to cell culture systems, mouse models are highly favored for evaluating tumor progression and therapeutic response in a more realistic in vivo context. The past decade has witnessed a dramatic increase in the number of functional genomic studies using diverse mouse models, including in vivo clustered regularly interspaced short palindromic repeats (CRISPR) and RNA interference (RNAi) screens, and these have provided a wealth of knowledge addressing multiple essential questions in translational cancer research. We compare the multiple mouse systems and genomic tools that are commonly used for in vivo screens to illustrate their strengths and limitations. Crucial components of screen design and data analysis are also discussed.
Collapse
Affiliation(s)
- Chennan Li
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA; Bindley Biosciences Center, Purdue University, West Lafayette, IN 47907, USA
| | - Andrea L Kasinski
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA; Bindley Biosciences Center, Purdue University, West Lafayette, IN 47907, USA; Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
35
|
Schork NJ, Goetz LH, Lowey J, Trent J. Strategies for Testing Intervention Matching Schemes in Cancer. Clin Pharmacol Ther 2020; 108:542-552. [PMID: 32535886 DOI: 10.1002/cpt.1947] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 06/04/2020] [Indexed: 01/02/2023]
Abstract
Personalized medicine, or the tailoring of health interventions to an individual's nuanced and often unique genetic, biochemical, physiological, behavioral, and/or exposure profile, is seen by many as a biological necessity given the great heterogeneity of pathogenic processes underlying most diseases. However, testing and ultimately proving the benefit of strategies or algorithms connecting the mechanisms of action of specific interventions to patient pathophysiological profiles (referred to here as "intervention matching schemes" (IMS)) is complex for many reasons. We argue that IMS are likely to be pervasive, if not ubiquitous, in future health care, but raise important questions about their broad deployment and the contexts within which their utility can be proven. For example, one could question the need to, the efficiency associated with, and the reliability of, strategies for comparing competing or perhaps complementary IMS. We briefly summarize some of the more salient issues surrounding the vetting of IMS in cancer contexts and argue that IMS are at the foundation of many modern clinical trials and intervention strategies, such as basket, umbrella, and adaptive trials. In addition, IMS are at the heart of proposed "rapid learning systems" in hospitals, and implicit in cell replacement strategies, such as cytotoxic T-cell therapies targeting patient-specific neo-antigen profiles. We also consider the need for sensitivity to issues surrounding the deployment of IMS and comment on directions for future research.
Collapse
Affiliation(s)
- Nicholas J Schork
- The Translational Genomics Research Institute (TGen), Phoenix, Arizona, USA.,Department of Population Sciences, The City of Hope National Medical Center, Duarte, California, USA.,Department of Molecular and Cell Biology, The City of Hope National Medical Center, Duarte, California, USA
| | - Laura H Goetz
- The Translational Genomics Research Institute (TGen), Phoenix, Arizona, USA.,Department of Medical Oncology, The City of Hope National Medical Center, Duarte, California, USA
| | - James Lowey
- The Translational Genomics Research Institute (TGen), Phoenix, Arizona, USA
| | - Jeffrey Trent
- The Translational Genomics Research Institute (TGen), Phoenix, Arizona, USA.,Department of Medical Oncology, The City of Hope National Medical Center, Duarte, California, USA
| |
Collapse
|
36
|
Pillonel V, Juskevicius D, Bihl M, Stenner F, Halter JP, Dirnhofer S, Tzankov A. Routine next generation sequencing of lymphoid malignancies: clinical utility and challenges from a 3-Year practical experience. Leuk Lymphoma 2020; 61:2568-2583. [PMID: 32623938 DOI: 10.1080/10428194.2020.1786560] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Since 2016, a next-generation sequencing (NGS) panel targeting 68 genes frequently mutated in lymphoid malignancies is an accredited part of routine diagnostics at the Institute of Pathology in Basel, Switzerland. Here, we retrospectively evaluate the feasibility and utility of integrating this NGS platform into routine practice on 80 diagnostic cases of lymphoid proliferations. NGS analysis was useful in most instances, yielding a diagnostically, predictively and/or prognostically meaningful result. In 35 out of the 50 cases, in which conventional histopathological evaluation remained indecisive, molecular subtyping with the NGS panel was helpful to either confirm or support the favored diagnosis, enable a differential diagnosis, or seriously question a suspected diagnosis. A total of 61 actionable or potentially actionable mutations in 34 out of 80 cases that might have enabled patient selection for targeted therapies was detected. NGS panel analysis had implications for prognosis in all 15 cases interrogated for risk assessment.
Collapse
Affiliation(s)
- Vincent Pillonel
- Institute of Pathology and Medical Genetics, University Hospital Basel, Basel, Switzerland.,Department of Medical Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Darius Juskevicius
- Institute of Pathology and Medical Genetics, University Hospital Basel, Basel, Switzerland
| | - Michel Bihl
- Institute of Pathology and Medical Genetics, University Hospital Basel, Basel, Switzerland
| | - Frank Stenner
- Department of Medical Oncology, University Hospital Basel, Basel, Switzerland
| | - Jörg P Halter
- Department of Medicine, Division of Hematology, University Hospital Basel, Basel, Switzerland
| | - Stefan Dirnhofer
- Institute of Pathology and Medical Genetics, University Hospital Basel, Basel, Switzerland
| | - Alexandar Tzankov
- Institute of Pathology and Medical Genetics, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
37
|
Lowder CY, Dhir T, Goetz AB, Thomsett HL, Bender J, Tatarian T, Madhavan S, Petricoin EF, Blais E, Lavu H, Winter JM, Posey J, Brody JR, Pishvaian MJ, Yeo CJ. A step towards personalizing next line therapy for resected pancreatic and related cancer patients: A single institution's experience. Surg Oncol 2020; 33:118-125. [PMID: 32561076 PMCID: PMC7498307 DOI: 10.1016/j.suronc.2020.02.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 12/09/2019] [Accepted: 02/06/2020] [Indexed: 12/17/2022]
Abstract
Background: There is a lack of precision medicine in pancreatic ductal adenocarcinoma (PDA) and related cancers, and outcomes for patients with this diagnosis remain poor despite decades of research investigating this disease. Therefore, it is necessary to explore novel therapeutic options for these patients who may benefit from personalized therapies. Objective: Molecular profiling of hepatopancreaticobiliary malignancies at our institution, including but not limited to PDA, was initiated to assess the feasibility of incorporating molecular profiling results into patient oncological therapy planning. Methods: All eligible patients from Thomas Jefferson University (TJU) with hepatopancreaticobiliary tumors including PDA, who agreed to molecular testing profiling, were prospectively enrolled in a registry study from December 2014 to September 2017 and their tumor samples were tested to identify molecular markers that can be used to guide therapy options in the future. Next generation sequencing (NGS) and protein expression in tumor samples were tested at CLIA-certified laboratories. Prospective clinicopathologic data were extracted from medical records and compiled in a de-identified fashion. Results: Seventy eight (78) patients were enrolled in the study, which included 65/78 patients with PDA (local and metastatic) and out of that subset, 52/65 patients had surgically resected PDA. Therapy recommendations were generated based on molecular and clinicopathologic data for all enrolled patients. NGS uncovered actionable alterations in 25/52 surgically resected PDAs (48%) which could be used to guide therapy options in the future. High expression of three proteins, TS (p ¼ 0.005), ERCC1 (p = 0.001), and PD-1 (p = 0.04), was associated with reduced recurrence-free survival (RFS), while TP53 mutations were correlated with longer RFS (p = 0.01). Conclusions: The goal of this study was to implement a stepwise strategy to identify and profile resected PDAs at our institution. Consistent with previous studies, approximately half of patients with resected PDA harbor actionable mutations with possible targeted therapeutic implications. Ongoing studies will determine the clinical value of identifying these mutations in patients with resected PDA.
Collapse
Affiliation(s)
- Cinthya Y Lowder
- Department of Surgery, Albert Einstein Medical Center, Philadelphia, PA, USA
| | - Teena Dhir
- The Jefferson Pancreatic, Biliary, and Related Cancer Center, Department of Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Austin B Goetz
- Department of Surgery, Albert Einstein Medical Center, Philadelphia, PA, USA
| | - Henry L Thomsett
- The Jefferson Pancreatic, Biliary, and Related Cancer Center, Department of Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Talar Tatarian
- The Jefferson Pancreatic, Biliary, and Related Cancer Center, Department of Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Subha Madhavan
- Perthera, Inc, McLean, VA, USA; The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Harish Lavu
- The Jefferson Pancreatic, Biliary, and Related Cancer Center, Department of Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Jordan M Winter
- University Hospital Seidman Cancer Center, Cleveland, OH, USA; University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - James Posey
- The Jefferson Pancreatic, Biliary, and Related Cancer Center, Department of Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Jonathan R Brody
- The Jefferson Pancreatic, Biliary, and Related Cancer Center, Department of Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Michael J Pishvaian
- Perthera, Inc, McLean, VA, USA; The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Charles J Yeo
- The Jefferson Pancreatic, Biliary, and Related Cancer Center, Department of Surgery, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
38
|
Park P, Shin SY, Park SY, Yun J, Shin C, Jung J, Choi KS, Cha HS. Next-Generation Sequencing-Based Cancer Panel Data Conversion Using International Standards to Implement a Clinical Next-Generation Sequencing Research System: Single-Institution Study. JMIR Med Inform 2020; 8:e14710. [PMID: 32329738 PMCID: PMC7210491 DOI: 10.2196/14710] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 11/19/2019] [Accepted: 02/07/2020] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND The analytical capacity and speed of next-generation sequencing (NGS) technology have been improved. Many genetic variants associated with various diseases have been discovered using NGS. Therefore, applying NGS to clinical practice results in precision or personalized medicine. However, as clinical sequencing reports in electronic health records (EHRs) are not structured according to recommended standards, clinical decision support systems have not been fully utilized. In addition, integrating genomic data with clinical data for translational research remains a great challenge. OBJECTIVE To apply international standards to clinical sequencing reports and to develop a clinical research information system to integrate standardized genomic data with clinical data. METHODS We applied the recently published ISO/TS 20428 standard to 367 clinical sequencing reports generated by panel (91 genes) sequencing in EHRs and implemented a clinical NGS research system by extending the clinical data warehouse to integrate the necessary clinical data for each patient. We also developed a user interface with a clinical research portal and an NGS result viewer. RESULTS A single clinical sequencing report with 28 items was restructured into four database tables and 49 entities. As a result, 367 patients' clinical sequencing data were connected with clinical data in EHRs, such as diagnosis, surgery, and death information. This system can support the development of cohort or case-control datasets as well. CONCLUSIONS The standardized clinical sequencing data are not only for clinical practice and could be further applied to translational research.
Collapse
Affiliation(s)
- Phillip Park
- Cancer Data Center, National Cancer Center, Goyang, Republic of Korea
| | - Soo-Yong Shin
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, Republic of Korea
- Big Data Research Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Seog Yun Park
- Department of Pathology, National Cancer Center, Goyang, Republic of Korea
| | - Jeonghee Yun
- Department of Pathology, National Cancer Center, Goyang, Republic of Korea
| | - Chulmin Shin
- Cancer Data Center, National Cancer Center, Goyang, Republic of Korea
| | - Jipmin Jung
- Cancer Data Center, National Cancer Center, Goyang, Republic of Korea
| | - Kui Son Choi
- Cancer Data Center, National Cancer Center, Goyang, Republic of Korea
| | - Hyo Soung Cha
- Cancer Data Center, National Cancer Center, Goyang, Republic of Korea
| |
Collapse
|
39
|
Affiliation(s)
- Xiwei Tang
- Department of Statistics, University of Virginia , Charlottesville , VA
| | - Fei Xue
- Department of Biostatistics and Epidemiology, University of Pennsylvania , Philadelphia , PA
| | - Annie Qu
- Department of Statistics, University of Illinois at Irvine , Irvine , CA
| |
Collapse
|
40
|
Aggarwal V, Sak K, Arora M, Iqubal A, Kumar A, Srivastava S, Pandey A, Kaur S, Tuli HS. History of Oncotherapies in Cancer Biology. DRUG TARGETS IN CELLULAR PROCESSES OF CANCER: FROM NONCLINICAL TO PRECLINICAL MODELS 2020:1-13. [DOI: 10.1007/978-981-15-7586-0_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/26/2024]
|
41
|
Sun T, Wang D, Ping Y, Sang Y, Dai Y, Wang Y, Liu Z, Duan X, Tao Z, Liu W. Integrated profiling identifies SLC5A6 and MFAP2 as novel diagnostic and prognostic biomarkers in gastric cancer patients. Int J Oncol 2019; 56:460-469. [PMID: 31894266 PMCID: PMC6959404 DOI: 10.3892/ijo.2019.4944] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 09/02/2019] [Indexed: 12/13/2022] Open
Abstract
Gastric cancer (GC) is one of the leading causes of malignancy‑associated mortality worldwide. However, the underlying molecular mechanisms of GC are unclear and the prognosis of GC is poor. Therefore, it is important and urgent to explore the underlying mechanisms and screen for novel diagnostic and prognostic biomarkers, as well as therapeutic targets. In the current study, scale‑free gene co‑expression networks were constructed using weighted gene co‑expression network analysis, the potential associations between gene sets and clinical features were investigated, and the hub genes were identified. The gene expression profiles of GSE38749 were downloaded from the Gene Expression Omnibus database. RNA‑seq and clinical data for GC from The Cancer Genome Atlas were utilized for verification. Furthermore, the expression of candidate biomarkers in gastric tissues was investigated. Survival analysis was performed using Kaplan‑Meier and log‑rank test. The predictive role of candidate biomarkers in GC was evaluated using a receiver operator characteristic (ROC) curve. Gene Ontology, gene set enrichment analysis and gene set variation analysis methods were used to interpret the function of candidate biomarkers in GC. A total of 29 modules were identified via the average linkage hierarchical clustering. A significant module consisting of 48 genes associated with clinical traits was found; three genes with high connectivity in the clinical significant module were identified as hub genes. Among them, SLC5A6 and microfibril‑associated protein 2 (MFAP2) were negatively associated with the overall survival, and their expression was elevated in GC compared with non‑tumor tissues. Additionally, ROC curves indicated that SLC5A6 and MFAP2 showed a good diagnostic power in discriminating cancerous from normal tissues. SLC5A6 and MFAP2 were identified as novel diagnostic and prognostic biomarkers in GC patients; both of these genes were first reported here in connection with GC and deserved further research.
Collapse
Affiliation(s)
- Tao Sun
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Danhua Wang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Ying Ping
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Yiwen Sang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Yibei Dai
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Yiyun Wang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Zhenping Liu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Xiuzhi Duan
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Zhihua Tao
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Weiwei Liu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| |
Collapse
|
42
|
Zhang R, Yuan F, Shu Y, Tian Y, Zhou B, Yi L, Zhang X, Ding Z, Xu H, Yang L. Personalized neoantigen-pulsed dendritic cell vaccines show superior immunogenicity to neoantigen-adjuvant vaccines in mouse tumor models. Cancer Immunol Immunother 2019; 69:135-145. [PMID: 31807878 PMCID: PMC6949210 DOI: 10.1007/s00262-019-02448-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 11/28/2019] [Indexed: 02/05/2023]
Abstract
Development of personalized cancer vaccines based on neoantigens has become a new direction in cancer immunotherapy. Two forms of cancer vaccines have been widely studied: tumor-associated antigen (including proteins, peptides, or tumor lysates)-pulsed dendritic cell (DC) vaccines and protein- or peptide-adjuvant vaccines. However, different immune modalities may produce different therapeutic effects and immune responses when the same antigen is used. Therefore, it is necessary to choose a more effective neoantigen vaccination method. In this study, we compared the differences in immune and anti-tumor effects between neoantigen-pulsed DC vaccines and neoantigen-adjuvant vaccines using murine lung carcinoma (LL2) candidate neoantigens. The enzyme-linked immunospot (ELISPOT) assay showed that 4/6 of the neoantigen-adjuvant vaccines and 6/6 of the neoantigen-pulsed DC vaccines induced strong T-cell immune responses. Also, 2/6 of the neoantigen-adjuvant vaccines and 5/6 of the neoantigen-pulsed DC vaccines exhibited potent anti-tumor effects. The results indicated that the neoantigen-pulsed DC vaccines were superior to the neoantigen-adjuvant vaccines in both activating immune responses and inhibiting tumor growth. Our fundings provide an experimental basis for the selection of immune modalities for the use of neoantigens in individualized tumor immunotherapies.
Collapse
Affiliation(s)
- Rui Zhang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, No. 17, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, Sichuan, China
| | - Fengjiao Yuan
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, No. 17, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, Sichuan, China
| | - Yang Shu
- State Key Laboratory of Biotherapy, and Precision Medicine Key Laboratory of Sichuan Province, Precision Medicine Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Yaomei Tian
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, No. 17, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, Sichuan, China
| | - Bailing Zhou
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, No. 17, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, Sichuan, China
| | - Linglu Yi
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, No. 17, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, Sichuan, China
| | - Xueyan Zhang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, No. 17, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, Sichuan, China
| | - Zhenyu Ding
- State Key Laboratory of Biotherapy, West China Medical School, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Heng Xu
- State Key Laboratory of Biotherapy, and Precision Medicine Key Laboratory of Sichuan Province, Precision Medicine Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Li Yang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, No. 17, Section 3, South Renmin Road, Wuhou District, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
43
|
Zhou C, Zhong X, Song Y, Shi J, Wu Z, Guo Z, Sun J, Wang Z. Prognostic Biomarkers for Gastric Cancer: An Umbrella Review of the Evidence. Front Oncol 2019; 9:1321. [PMID: 31850212 PMCID: PMC6895018 DOI: 10.3389/fonc.2019.01321] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 11/12/2019] [Indexed: 12/14/2022] Open
Abstract
Introduction: Biomarkers are biological molecules entirely or partially participating in cancerous processes that function as measurable indicators of abnormal changes in the human body microenvironment. Aiming to provide an overview of associations between prognostic biomarkers and gastric cancer (GC), we performed this umbrella review analyzing currently available meta-analyses and grading the evidence depending on the credibility of their associations. Methods: A systematic literature search was conducted by two independent investigators of the PubMed, Embase, Web of Science, and Cochrane Databases to identify meta-analyses investigating associations between prognostic biomarkers and GC. The strength of evidence for prognostic biomarkers for GC were categorized into four grades: strong, highly suggestive, suggestive, and weak. Results: Among 120 associations between prognostic biomarkers and GC survival outcomes, only one association, namely the association between platelet count and GC OS, was supported by strong evidence. Associations between FITC, CEA, NLR, foxp3+ Treg lymphocytes (both 1- and 3-year OS), CA 19-9, or VEGF and GC OS were supported by highly suggestive evidence. Four associations were considered suggestive and the remaining 108 associations were supported by weak or not suggestive evidence. Discussion: The association between platelet count and GC OS was supported by strong evidence. Associations between FITC, CEA, NLR, foxp3+ Treg lymphocytes (both 1- and 3-year OS), CA 19-9, or VEGF and GC OS were supported by highly suggestive evidence, however, the results should be interpreted cautiously due to inadequate methodological quality as deemed by AMSTAR 2.0.
Collapse
Affiliation(s)
- Cen Zhou
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xi Zhong
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yongxi Song
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Jinxin Shi
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Zhonghua Wu
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Zhexu Guo
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Jie Sun
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Zhenning Wang
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
44
|
Paananen J, Fortino V. An omics perspective on drug target discovery platforms. Brief Bioinform 2019; 21:1937-1953. [PMID: 31774113 PMCID: PMC7711264 DOI: 10.1093/bib/bbz122] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/23/2019] [Accepted: 07/27/2019] [Indexed: 01/28/2023] Open
Abstract
The drug discovery process starts with identification of a disease-modifying target. This critical step traditionally begins with manual investigation of scientific literature and biomedical databases to gather evidence linking molecular target to disease, and to evaluate the efficacy, safety and commercial potential of the target. The high-throughput and affordability of current omics technologies, allowing quantitative measurements of many putative targets (e.g. DNA, RNA, protein, metabolite), has exponentially increased the volume of scientific data available for this arduous task. Therefore, computational platforms identifying and ranking disease-relevant targets from existing biomedical data sources, including omics databases, are needed. To date, more than 30 drug target discovery (DTD) platforms exist. They provide information-rich databases and graphical user interfaces to help scientists identify putative targets and pre-evaluate their therapeutic efficacy and potential side effects. Here we survey and compare a set of popular DTD platforms that utilize multiple data sources and omics-driven knowledge bases (either directly or indirectly) for identifying drug targets. We also provide a description of omics technologies and related data repositories which are important for DTD tasks.
Collapse
Affiliation(s)
- Jussi Paananen
- Institute of Biomedicine, University of Eastern Finland, Finland.,Blueprint Genetics Ltd, Finland
| | - Vittorio Fortino
- Institute of Biomedicine, University of Eastern Finland, Finland
| |
Collapse
|
45
|
Schoenborn P, Osborne R, Toms N, Johnstone K, Milsom C, Muneer R, Jarvis MA, Belshaw R. OncoSim and OncoWiki: an authentic learning approach to teaching cancer genomics. BMC MEDICAL EDUCATION 2019; 19:407. [PMID: 31699073 PMCID: PMC6836658 DOI: 10.1186/s12909-019-1812-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 09/20/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Personalised medicine is rapidly changing the clinical environment, especially in regard to the management of cancer. However, for the large part, methods used to educate undergraduate students as future biomedical scientists and medical doctors have not reflected these changes. In order to make effective use of advances in cancer genomic knowledge, there is a need to expose students to the challenges of genomic medicine and to do so in a manner that makes this complex information accessible. METHODS The teaching method developed, OncoSim, is a scaffolded 'Personal Research' module option for final year biomedical undergraduate students. It uses an authentic learning approach to teach cancer genomics via simulated cancer patient case studies that have identifiable potential therapeutic targets with associated drug therapies (so-called targeted therapy/precision oncology). In addition, these simulated case studies can be uploaded to a dedicated learning website (OncoWiki) where they can be freely downloaded and used to teach medical students the principles of targeted therapy. A preliminary evaluation of OncoSim was carried out using 3 research tools: (1) online questionnaires; (2) semi-structured interviews; and (3) analysis of whole cohort mark ranges. Thematic analysis was used to code and categorise interview data. RESULTS The teaching materials for OncoSim and the OncoWiki site are freely accessible at https://www.oncowiki.co.uk. Questionnaire data and comparison of whole cohort marks showed OncoSim was at least as effective as alternative choices, and suggested OncoSim provided a valued alternative to traditional laboratory-based projects. No barriers to receptiveness were found. Interview analysis provided 5 broad themes (authentic learning experience; individual challenges; interest in cancer; positive learning experience; supportive structure) supporting the authentic learning aspect of the project, the strong scaffolding provided and the overall effectiveness of the approach. CONCLUSIONS Our preliminary, proof-of-concept, evaluation suggests that OncoSim will be effective in supporting the teaching of genomic medicine to undergraduate students. We plan and hope our study will encourage further formal evaluation in a larger cohort of students, including a control group. The OncoWiki site has the capacity to grow independently as future students create and upload simulated case studies for other students to then download and analyse.
Collapse
Affiliation(s)
| | | | - Nick Toms
- Peninsula Medical School, Faculty of Health: Medicine, Dentistry and Human Sciences, University of Plymouth, Plymouth, UK
| | - Karen Johnstone
- Peninsula Medical School, Faculty of Health: Medicine, Dentistry and Human Sciences, University of Plymouth, Plymouth, UK
| | - Chlöe Milsom
- Peninsula Medical School, Faculty of Health: Medicine, Dentistry and Human Sciences, University of Plymouth, Plymouth, UK
| | - Reema Muneer
- Educational Development, University of Plymouth, Plymouth, UK
| | - Michael A Jarvis
- School of Biomedical Sciences, Faculty of Health: Medicine, Dentistry and Human Sciences, University of Plymouth, Plymouth, UK
| | - Robert Belshaw
- School of Biomedical Sciences, Faculty of Health: Medicine, Dentistry and Human Sciences, University of Plymouth, Plymouth, UK.
| |
Collapse
|
46
|
Ziats CA, Rennert OM, Ziats MN. Toward a Pathway-Driven Clinical-Molecular Framework for Classifying Autism Spectrum Disorders. Pediatr Neurol 2019; 98:46-52. [PMID: 31272785 DOI: 10.1016/j.pediatrneurol.2019.05.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 04/30/2019] [Accepted: 05/06/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND The current classification system of neurodevelopmental disorders is based on clinical criteria; however, this method alone fails to incorporate what is now known about genomic similarities and differences between closely related clinical neurodevelopmental disorders. Here we present an alternative clinical molecular classification system of neurodevelopmental disorders based on shared molecular and cellular pathways, using syndromes with autistic features as examples. METHODS Using the Online Mendelian Inheritance in Man database, we identified 83 syndromes that had "autism" as a feature of disease, which in combination were associated with 69 autism disease-causing genes. Using annotation terms generated from the DAVID annotation tool, we grouped each gene and its associated autism syndrome into three biological pathways: ion transport, cellular synaptic function, and transcriptional regulation. RESULTS The majority of the autism syndromes we analyzed (54 of 83) enriched for processes related to transcriptional regulation and were associated with more non-neurologic symptoms and co-morbid psychiatric disease when compared with the other two pathways studied. Disorders with disrupted cellular synaptic function had significantly more motor-related symptoms when compared with the other groups of disorders. CONCLUSION Our pathway-based classification system identified unique clinical characteristics within each group that may help guide clinical diagnosis, prognosis, and treatment. These results suggest that shifting current clinical classification of autism disorders toward molecularly driven, pathway-related diagnostic groups such as this may more precisely guide clinical decision making and may be informative for future clinical trial and drug development approaches.
Collapse
Affiliation(s)
- Catherine A Ziats
- National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland.
| | - Owen M Rennert
- National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Mark N Ziats
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
47
|
Huang CC, Du M, Wang L. Bioinformatics Analysis for Circulating Cell-Free DNA in Cancer. Cancers (Basel) 2019; 11:cancers11060805. [PMID: 31212602 PMCID: PMC6627444 DOI: 10.3390/cancers11060805] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 06/03/2019] [Accepted: 06/06/2019] [Indexed: 12/28/2022] Open
Abstract
Molecular analysis of cell-free DNA (cfDNA) that circulates in plasma and other body fluids represents a "liquid biopsy" approach for non-invasive cancer screening or monitoring. The rapid development of sequencing technologies has made cfDNA a promising source to study cancer development and progression. Specific genetic and epigenetic alterations have been found in plasma, serum, and urine cfDNA and could potentially be used as diagnostic or prognostic biomarkers in various cancer types. In this review, we will discuss the molecular characteristics of cancer cfDNA and major bioinformatics approaches involved in the analysis of cfDNA sequencing data for detecting genetic mutation, copy number alteration, methylation change, and nucleosome positioning variation. We highlight specific challenges in sensitivity to detect genetic aberrations and robustness of statistical analysis. Finally, we provide perspectives regarding the standard and continuing development of bioinformatics analysis to move this promising screening tool into clinical practice.
Collapse
Affiliation(s)
- Chiang-Ching Huang
- Zilber School of Public Health, University of Wisconsin, Milwaukee, WI 53205, USA.
| | - Meijun Du
- Department of Pathology and MCW Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Liang Wang
- Department of Pathology and MCW Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
48
|
O'Cathail SM, Buffa FM. Science in Focus: Bioinformatics Part 1 - Lost in Translation. Clin Oncol (R Coll Radiol) 2019; 31:337-340. [PMID: 30975523 DOI: 10.1016/j.clon.2019.03.043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 03/02/2019] [Accepted: 03/04/2019] [Indexed: 02/07/2023]
Affiliation(s)
- S M O'Cathail
- CRUK/MRC Oxford Institute of Radiation Oncology, University of Oxford, Oxford, UK.
| | - F M Buffa
- Department of Oncology, University of Oxford, Oxford, UK
| |
Collapse
|
49
|
Ankney JA, Xie L, Wrobel JA, Wang L, Chen X. Novel secretome-to-transcriptome integrated or secreto-transcriptomic approach to reveal liquid biopsy biomarkers for predicting individualized prognosis of breast cancer patients. BMC Med Genomics 2019; 12:78. [PMID: 31146747 PMCID: PMC6543675 DOI: 10.1186/s12920-019-0530-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 05/13/2019] [Indexed: 02/08/2023] Open
Abstract
Background Presently, a 50-gene expression model (PAM50) serves as a breast cancer (BC) subtype classifier that is insufficient to distinguish, within each single PAM50-classified subtype, patient subpopulations having different prognosis. There is a pressing need for inexpensive and minimally invasive biomarker tests to easily and accurately predict individuals’ clinical outcomes and response to treatments. Although quantitative proteomic approaches have been developed to identify/profile proteins secreted (secretome) from various cancer cell lines in vitro, missing are the clinicopathological relevance and the associated prognostic value of these secretomic identifications. Methods To discover biomarkers to predict individualized prognosis we introduce a new multi-omics (secreto-transcriptomics) method that identifies, in their oncogenically secreted states, candidate markers of BC subtypes whose genes bear patient-specific mRNA expression alterations of prognostic significance. First, we used label-free quantitative (LFQ) proteomics to identify the proteins showing BC-subtypic secretion from a series of BC cell lines representing major BC-subtypes. To determine and externally validate the prognostic value of these secreted proteins, we developed a secreto-transcriptomic approach that discovered a PAM50-subtypic Secretion-Correlated mRNA Expression Pattern (SeCEP) wherein the PAM50-subtypic secretion of select proteins statistically correlated with cis-mRNA expression of their encoding genes in patients of the corresponding PAM50-subtypes. Kaplan-Meier analysis of SeCEP genes was used to identify new liquid biopsy biomarkers for predicting individualized prognosis. Results The mRNA expression-to-secretion correlation (SeCEP) pinpointed multiple genes that are fully translated into the oncogenically active secretome in a PAM50-subtypic manner. Further, multiple SeCEP genes in distinct combinations or panels of multiple SeCEP genes were identified as ‘systems prognostic markers’ that showed mRNA co-overexpression patterns in the distinct subpopulations of PAM50-subtypic patients with poor prognosis or high-risk of relapse. Thus, our secreto-transcriptomic approach statistically linked BC subtypic secretome genes with patient-specific information about their mRNA expression alterations and significantly improved the sensitivity and specificity in patient stratification in the context of clinical outcomes or prognosis. Conclusions By combining LFQ secretome screening with proteo-transcriptomic retrospective analysis of patient data our integrated multi-omics approach bypasses costly, tedious, genome-wide fishing and predictive modeling that are commonly required to distinguish a few prognostically altered genes from thousands of other non-BC related genes in a genome. Electronic supplementary material The online version of this article (10.1186/s12920-019-0530-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- J Astor Ankney
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Ling Xie
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - John A Wrobel
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Li Wang
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Xian Chen
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA. .,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
50
|
Dressler LG, Bell GC, Schuetze DP, Steciuk MR, Binns OA, Raab RE, Abernathy PM, Wilson CM, Kunutsor SE, Loveless MC, Ahearne PM, Messino MJ. Implementing a personalized medicine cancer program in a community cancer system. Per Med 2019; 16:221-232. [DOI: 10.2217/pme-2018-0112] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Lynn G Dressler
- Independent Consultant, LGD consulting, Fairview, NC 287303, USA
- Independent Consultant, Asheville, NC 288014, USA
- Duke University Research Institute, Durham, NC 28777, USA
- Mission Health Cancer Center, Hospital Drive, Asheville, NC 28801, USA
| | - Gillian C Bell
- Mission Health Cancer Center, Hospital Drive, Asheville, NC 28801, USA
| | - David P Schuetze
- Mission Health Cancer Center, Hospital Drive, Asheville, NC 28801, USA
| | - Mark R Steciuk
- Mission Health Cancer Center, Hospital Drive, Asheville, NC 28801, USA
| | - Oliver A Binns
- Mission Health Cancer Center, Hospital Drive, Asheville, NC 28801, USA
| | - Rachel E Raab
- Mission Health Cancer Center, Hospital Drive, Asheville, NC 28801, USA
- Independent Scholar, Asheville, NC 2880, USA
| | - Pearl M Abernathy
- Mission Health Cancer Center, Hospital Drive, Asheville, NC 28801, USA
| | - Carolyn M Wilson
- Mission Health Cancer Center, Hospital Drive, Asheville, NC 28801, USA
| | - Sedope E Kunutsor
- Mission Health Cancer Center, Hospital Drive, Asheville, NC 28801, USA
- Texas Cancer Registry, Austin, TX 73301
| | - Marika C Loveless
- Duke University Research Institute, Durham, NC 28777, USA
- Mission Health Cancer Center, Hospital Drive, Asheville, NC 28801, USA
| | - Paul M Ahearne
- Mission Health Cancer Center, Hospital Drive, Asheville, NC 28801, USA
| | - Michael J Messino
- Mission Health Cancer Center, Hospital Drive, Asheville, NC 28801, USA
| |
Collapse
|