1
|
Guo Y, Li T, Gong B, Hu Y, Wang S, Yang L, Zheng C. From Images to Genes: Radiogenomics Based on Artificial Intelligence to Achieve Non-Invasive Precision Medicine in Cancer Patients. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2408069. [PMID: 39535476 DOI: 10.1002/advs.202408069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/19/2024] [Indexed: 11/16/2024]
Abstract
With the increasing demand for precision medicine in cancer patients, radiogenomics emerges as a promising frontier. Radiogenomics is originally defined as a methodology for associating gene expression information from high-throughput technologies with imaging phenotypes. However, with advancements in medical imaging, high-throughput omics technologies, and artificial intelligence, both the concept and application of radiogenomics have significantly broadened. In this review, the history of radiogenomics is enumerated, related omics technologies, the five basic workflows and their applications across tumors, the role of AI in radiogenomics, the opportunities and challenges from tumor heterogeneity, and the applications of radiogenomics in tumor immune microenvironment. The application of radiogenomics in positron emission tomography and the role of radiogenomics in multi-omics studies is also discussed. Finally, the challenges faced by clinical transformation, along with future trends in this field is discussed.
Collapse
Affiliation(s)
- Yusheng Guo
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Tianxiang Li
- Department of Ultrasound, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical. Sciences, Peking Union Medical College, Beijing, 100730, China
| | - Bingxin Gong
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yan Hu
- Research Institute of Trustworthy Autonomous Systems and Department of Computer Science and Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Sichen Wang
- School of Life Science and Technology, Computational Biology Research Center, Harbin Institute of Technology, Harbin, 150001, China
| | - Lian Yang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| |
Collapse
|
2
|
Chhabra R. Molecular and modular intricacies of precision oncology. Front Immunol 2024; 15:1476494. [PMID: 39507541 PMCID: PMC11537923 DOI: 10.3389/fimmu.2024.1476494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 09/30/2024] [Indexed: 11/08/2024] Open
Abstract
Precision medicine is revolutionizing the world in combating different disease modalities, including cancer. The concept of personalized treatments is not new, but modeling it into a reality has faced various limitations. The last decade has seen significant improvements in incorporating several novel tools, scientific innovations and governmental support in precision oncology. However, the socio-economic factors and risk-benefit analyses are important considerations. This mini review includes a summary of some commendable milestones, which are not just a series of successes, but also a cautious outlook to the challenges and practical implications of the advancing techno-medical era.
Collapse
Affiliation(s)
- Ravneet Chhabra
- Business Department, Biocytogen Boston Corporation, Waltham, MA, United States
| |
Collapse
|
3
|
Farah E, Kenney M, Warkentin MT, Cheung WY, Brenner DR. Examining external control arms in oncology: A scoping review of applications to date. Cancer Med 2024; 13:e7447. [PMID: 38984669 PMCID: PMC11234289 DOI: 10.1002/cam4.7447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 06/11/2024] [Accepted: 06/24/2024] [Indexed: 07/11/2024] Open
Abstract
OBJECTIVES Randomized controlled trials (RCTs) are the gold standard for evaluating the comparative efficacy and safety of new cancer therapies. However, enrolling patients in control arms of clinical trials can be challenging for rare cancers, particularly in the context of precision oncology and targeted therapies. External Control Arms (ECAs) are a potential solution to address these challenges in clinical research design. We conducted a scoping review to explore the use of ECAs in oncology. METHODS We systematically searched four databases, namely MEDLINE, EMBASE, Web of Science, and Scopus. We screened titles, abstracts, and full texts for eligible articles focusing on patients undergoing therapy for cancer, employing ECAs, and reporting clinical outcomes. RESULTS Of the 629 articles screened, 23 were included in this review. The earliest included studies were published in 1996, while most studies were published in the past 5 years. 44% (10/23) of ECAs were employed in blood-related cancer studies. Geographically, 30% (7/23) of studies were conducted in the United States, 22% (5/23) in Japan, and 9% (2/23) in South Korea. The primary data sources used to construct the ECAs involved pooled data from previous trials (35%, 8/23), administrative health databases (17%, 4/23) and electronic medical records (17%, 4/23). While 52% (12/23) of the studies employed methods to align treatment and ECAs characteristics, 48% (11/23) lacked explicit strategies. CONCLUSION ECAs offer a valuable approach in oncology research, particularly when alternative designs are not feasible. However, careful methodological planning and detailed reporting are essential for meaningful and reliable results.
Collapse
Affiliation(s)
- Eliya Farah
- Department of Oncology, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
- Department of Community Health Sciences, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
| | - Matthew Kenney
- Department of Oncology, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
- Department of Community Health Sciences, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
| | - Matthew T. Warkentin
- Department of Oncology, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
- Department of Community Health Sciences, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
| | - Winson Y. Cheung
- Department of Oncology, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
- Department of Community Health Sciences, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
| | - Darren R. Brenner
- Department of Oncology, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
- Department of Community Health Sciences, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
| |
Collapse
|
4
|
Dhruba SR, Sahni S, Wang B, Wu D, Rajagopal PS, Schmidt Y, Shulman ED, Sinha S, Sammut SJ, Caldas C, Wang K, Ruppin E. The expression patterns of different cell types and their interactions in the tumor microenvironment are predictive of breast cancer patient response to neoadjuvant chemotherapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.14.598770. [PMID: 39372749 PMCID: PMC11451622 DOI: 10.1101/2024.06.14.598770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
The tumor microenvironment (TME) is a complex ecosystem of diverse cell types whose interactions govern tumor growth and clinical outcome. While the TME's impact on immunotherapy has been extensively studied, its role in chemotherapy response remains less explored. To address this, we developed DECODEM (DEcoupling Cell-type-specific Outcomes using DEconvolution and Machine learning), a generic computational framework leveraging cellular deconvolution of bulk transcriptomics to associate the gene expression of individual cell types in the TME with clinical response. Employing DECODEM to analyze the gene expression of breast cancer (BC) patients treated with neoadjuvant chemotherapy, we find that the gene expression of specific immune cells (myeloid, plasmablasts, B-cells) and stromal cells (endothelial, normal epithelial, CAFs) are highly predictive of chemotherapy response, going beyond that of the malignant cells. These findings are further tested and validated in a single-cell cohort of triple negative breast cancer. To investigate the possible role of immune cell-cell interactions (CCIs) in mediating chemotherapy response, we extended DECODEM to DECODEMi to identify such CCIs, validated in single-cell data. Our findings highlight the importance of active pre-treatment immune infiltration for chemotherapy success. The tools developed here are made publicly available and are applicable for studying the role of the TME in mediating response from readily available bulk tumor expression in a wide range of cancer treatments and indications.
Collapse
Affiliation(s)
- Saugato Rahman Dhruba
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sahil Sahni
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Binbin Wang
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Di Wu
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Padma Sheila Rajagopal
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Women’s Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yael Schmidt
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Eldad D. Shulman
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sanju Sinha
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, San Diego, CA, USA
| | - Stephen-John Sammut
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital NHS Foundation Trust, London, UK
| | - Carlos Caldas
- Institute of Metabolic Science, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Department of Clinical Biochemistry, University of Cambridge, Cambridge, UK
| | - Kun Wang
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Eytan Ruppin
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
5
|
Sinha S, Vegesna R, Mukherjee S, Kammula AV, Dhruba SR, Wu W, Kerr DL, Nair NU, Jones MG, Yosef N, Stroganov OV, Grishagin I, Aldape KD, Blakely CM, Jiang P, Thomas CJ, Benes CH, Bivona TG, Schäffer AA, Ruppin E. PERCEPTION predicts patient response and resistance to treatment using single-cell transcriptomics of their tumors. NATURE CANCER 2024; 5:938-952. [PMID: 38637658 DOI: 10.1038/s43018-024-00756-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 03/08/2024] [Indexed: 04/20/2024]
Abstract
Tailoring optimal treatment for individual cancer patients remains a significant challenge. To address this issue, we developed PERCEPTION (PERsonalized Single-Cell Expression-Based Planning for Treatments In ONcology), a precision oncology computational pipeline. Our approach uses publicly available matched bulk and single-cell (sc) expression profiles from large-scale cell-line drug screens. These profiles help build treatment response models based on patients' sc-tumor transcriptomics. PERCEPTION demonstrates success in predicting responses to targeted therapies in cultured and patient-tumor-derived primary cells, as well as in two clinical trials for multiple myeloma and breast cancer. It also captures the resistance development in patients with lung cancer treated with tyrosine kinase inhibitors. PERCEPTION outperforms published state-of-the-art sc-based and bulk-based predictors in all clinical cohorts. PERCEPTION is accessible at https://github.com/ruppinlab/PERCEPTION . Our work, showcasing patient stratification using sc-expression profiles of their tumors, will encourage the adoption of sc-omics profiling in clinical settings, enhancing precision oncology tools based on sc-omics.
Collapse
Affiliation(s)
- Sanju Sinha
- Cancer Data Science Laboratory, National Cancer Institute, Bethesda, MD, USA.
- NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, San Diego, CA, USA.
| | - Rahulsimham Vegesna
- Cancer Data Science Laboratory, National Cancer Institute, Bethesda, MD, USA
| | - Sumit Mukherjee
- Cancer Data Science Laboratory, National Cancer Institute, Bethesda, MD, USA
| | - Ashwin V Kammula
- Cancer Data Science Laboratory, National Cancer Institute, Bethesda, MD, USA
- University of Maryland, College Park, MD, USA
| | | | - Wei Wu
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - D Lucas Kerr
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Nishanth Ulhas Nair
- Cancer Data Science Laboratory, National Cancer Institute, Bethesda, MD, USA
| | - Matthew G Jones
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA, USA
- Department of Electrical Engineering and Computer Science, University of California, Berkeley, Berkeley, CA, USA
- Integrative Program in Quantitative Biology, University of California, San Francisco, San Francisco, CA, USA
- Whitehead Institute, Cambridge, MA, USA
| | - Nir Yosef
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA, USA
- Department of Electrical Engineering and Computer Science, University of California, Berkeley, Berkeley, CA, USA
| | | | - Ivan Grishagin
- Rancho BioSciences, San Diego, CA, USA
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Kenneth D Aldape
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Collin M Blakely
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Peng Jiang
- Cancer Data Science Laboratory, National Cancer Institute, Bethesda, MD, USA
| | - Craig J Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Cyril H Benes
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Trever G Bivona
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub Investigator, San Francisco, CA, USA
| | | | - Eytan Ruppin
- Cancer Data Science Laboratory, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
6
|
Moaveni AK, Amiri M, Shademan B, Farhadi A, Behroozi J, Nourazarian A. Advances and challenges in gene therapy strategies for pediatric cancer: a comprehensive update. Front Mol Biosci 2024; 11:1382190. [PMID: 38836106 PMCID: PMC11149429 DOI: 10.3389/fmolb.2024.1382190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 03/27/2024] [Indexed: 06/06/2024] Open
Abstract
Pediatric cancers represent a tragic but also promising area for gene therapy. Although conventional treatments have improved survival rates, there is still a need for targeted and less toxic interventions. This article critically analyzes recent advances in gene therapy for pediatric malignancies and discusses the challenges that remain. We explore the innovative vectors and delivery systems that have emerged, such as adeno-associated viruses and non-viral platforms, which show promise in addressing the unique pathophysiology of pediatric tumors. Specifically, we examine the field of chimeric antigen receptor (CAR) T-cell therapies and their adaptation for solid tumors, which historically have been more challenging to treat than hematologic malignancies. We also discuss the genetic and epigenetic complexities inherent to pediatric cancers, such as tumor heterogeneity and the dynamic tumor microenvironment, which pose significant hurdles for gene therapy. Ethical considerations specific to pediatric populations, including consent and long-term follow-up, are also analyzed. Additionally, we scrutinize the translation of research from preclinical models that often fail to mimic pediatric cancer biology to the regulatory landscapes that can either support or hinder innovation. In summary, this article provides an up-to-date overview of gene therapy in pediatric oncology, highlighting both the rapid scientific progress and the substantial obstacles that need to be addressed. Through this lens, we propose a roadmap for future research that prioritizes the safety, efficacy, and complex ethical considerations involved in treating pediatric patients. Our ultimate goal is to move from incremental advancements to transformative therapies.
Collapse
Affiliation(s)
- Amir Kian Moaveni
- Pediatric Urology and Regenerative Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Amiri
- Pediatric Urology and Regenerative Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Behrouz Shademan
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Arezoo Farhadi
- Department of Genetics and Molecular Medicine, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Javad Behroozi
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Alireza Nourazarian
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran
| |
Collapse
|
7
|
Slootbeek PHJ, Tolmeijer SH, Mehra N, Schalken JA. Therapeutic biomarkers in metastatic castration-resistant prostate cancer: does the state matter? Crit Rev Clin Lab Sci 2024; 61:178-204. [PMID: 37882463 DOI: 10.1080/10408363.2023.2266482] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 09/28/2023] [Indexed: 10/27/2023]
Abstract
The treatment of metastatic castration-resistant prostate cancer (mCRPC) has been fundamentally transformed by our greater understanding of its complex biological mechanisms and its entrance into the era of precision oncology. A broad aim is to use the extreme heterogeneity of mCRPC by matching already approved or new targeted therapies to the correct tumor genotype. To achieve this, tumor DNA must be obtained, sequenced, and correctly interpreted, with individual aberrations explored for their druggability, taking into account the hierarchy of driving molecular pathways. Although tumor tissue sequencing is the gold standard, tumor tissue can be challenging to obtain, and a biopsy from one metastatic site or primary tumor may not provide an accurate representation of the current genetic underpinning. Sequencing of circulating tumor DNA (ctDNA) might catalyze precision oncology in mCRPC, as it enables real-time observation of genomic changes in tumors and allows for monitoring of treatment response and identification of resistance mechanisms. Moreover, ctDNA can be used to identify mutations that may not be detected in solitary metastatic lesions and can provide a more in-depth understanding of inter- and intra-tumor heterogeneity. Finally, ctDNA abundance can serve as a prognostic biomarker in patients with mCRPC.The androgen receptor (AR)-axis is a well-established therapeutical target for prostate cancer, and through ctDNA sequencing, insights have been obtained in (temporal) resistance mechanisms that develop through castration resistance. New third-generation AR-axis inhibitors are being developed to overcome some of these resistance mechanisms. The druggability of defects in the DNA damage repair machinery has impacted the treatment landscape of mCRPC in recent years. For patients with deleterious gene aberrations in genes linked to homologous recombination, particularly BRCA1 or BRCA2, PARP inhibitors have shown efficacy compared to the standard of care armamentarium, but platinum-based chemotherapy may be equally effective. A hierarchy exists in genes associated with homologous recombination, where, besides the canonical genes in this pathway, not every other gene aberration predicts the same likelihood of response. Moreover, evidence is emerging on cross-resistance between therapies such as PARP inhibitors, platinum-based chemotherapy and even radioligand therapy that target this genotype. Mismatch repair-deficient patients can experience a beneficial response to immune checkpoint inhibitors. Activation of other cellular signaling pathways such as PI3K, cell cycle, and MAPK have shown limited success with monotherapy, but there is potential in co-targeting these pathways with combination therapy, either already witnessed or anticipated. This review outlines precision medicine in mCRPC, zooming in on the role of ctDNA, to identify genomic biomarkers that may be used to tailor molecularly targeted therapies. The most common druggable pathways and outcomes of therapies matched to these pathways are discussed.
Collapse
Affiliation(s)
- Peter H J Slootbeek
- Department of Medical Oncology, Radboud university medical center, Nijmegen, The Netherland
| | - Sofie H Tolmeijer
- Department of Medical Oncology, Radboud university medical center, Nijmegen, The Netherland
| | - Niven Mehra
- Department of Medical Oncology, Radboud university medical center, Nijmegen, The Netherland
| | - Jack A Schalken
- Department of Experimental Urology, Research Institute of Medical Innovation, Radboud university medical center, Nijmegen, The Netherlands
| |
Collapse
|
8
|
Wang S, Lee D. Community cohesion looseness in gene networks reveals individualized drug targets and resistance. Brief Bioinform 2024; 25:bbae175. [PMID: 38622359 PMCID: PMC11018546 DOI: 10.1093/bib/bbae175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/19/2024] [Accepted: 04/02/2024] [Indexed: 04/17/2024] Open
Abstract
Community cohesion plays a critical role in the determination of an individual's health in social science. Intriguingly, a community structure of gene networks indicates that the concept of community cohesion could be applied between the genes as well to overcome the limitations of single gene-based biomarkers for precision oncology. Here, we develop community cohesion scores which precisely quantify the community ability to retain the interactions between the genes and their cellular functions in each individualized gene network. Using breast cancer as a proof-of-concept study, we measure the community cohesion score profiles of 950 case samples and predict the individualized therapeutic targets in 2-fold. First, we prioritize them by finding druggable genes present in the community with the most and relatively decreased scores in each individual. Then, we pinpoint more individualized therapeutic targets by discovering the genes which greatly contribute to the community cohesion looseness in each individualized gene network. Compared with the previous approaches, the community cohesion scores show at least four times higher performance in predicting effective individualized chemotherapy targets based on drug sensitivity data. Furthermore, the community cohesion scores successfully discover the known breast cancer subtypes and we suggest new targeted therapy targets for triple negative breast cancer (e.g. KIT and GABRP). Lastly, we demonstrate that the community cohesion scores can predict tamoxifen responses in ER+ breast cancer and suggest potential combination therapies (e.g. NAMPT and RXRA inhibitors) to reduce endocrine therapy resistance based on individualized characteristics. Our method opens new perspectives for the biomarker development in precision oncology.
Collapse
Affiliation(s)
- Seunghyun Wang
- Department of Bio and Brain Engineering, KAIST, Daejeon, Republic of Korea
| | - Doheon Lee
- Department of Bio and Brain Engineering, KAIST, Daejeon, Republic of Korea
| |
Collapse
|
9
|
Wu M, Zhang X, Tu Y, Cheng W, Zeng Y. Culture and expansion of murine proximal airway basal stem cells. Stem Cell Res Ther 2024; 15:26. [PMID: 38287366 PMCID: PMC10826159 DOI: 10.1186/s13287-024-03642-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 01/21/2024] [Indexed: 01/31/2024] Open
Abstract
BACKGROUND The stem cell characteristic makes basal cells desirable for ex vivo modeling of airway diseases. However, to date, approaches allowing them extensively in vitro serial expansion and maintaining bona fide stem cell property are still awaiting to be established. This study aims to develop a feeder-free culture system of mouse airway basal stem cells (ABSCs) that sustain their stem cell potential in vitro, providing an experimental basis for further in-depth research and mechanism exploration. METHODS We used ROCK inhibitor Y-27632-containing 3T3-CM, MEF-CM, and RbEF-CM to determine the proper feeder-free culture system that could maintain in vitro stem cell morphology of mouse ABSCs. Immunocytofluorescence was used to identify the basal cell markers of obtained cells. Serial propagation was carried out to observe whether the stem cell morphology and basal cell markers could be preserved in this cultivation system. Next, we examined the in vitro expansion and self-renewal ability by evaluating population doubling time and colony-forming efficiency. Moreover, the differentiation potential was detected by an in vitro differentiation culture and a 3D tracheosphere assay. RESULTS When the mouse ABSCs were cultured using 3T3-CM containing ROCK inhibitor Y-27632 in combination with Matrigel-coated culture dishes, they could stably expand and maintain stem cell-like clones. We confirmed that the obtained clones comprised p63/Krt5 double-positive ABSCs. In continuous passage and maintenance culture, we found that it could be subculture to at least 15 passages in vitro, stably maintaining its stem cell morphology, basal cell markers, and in vitro expansion and self-renewal capabilities. Meanwhile, through in vitro differentiation culture and 3D tracheosphere culture, we found that in addition to maintaining self-renewal, mouse ABSCs could differentiate into other airway epithelial cells such as acetylated tubulin (Act-Tub) + ciliated and MUC5AC + mucus-secreting cells. However, they failed to differentiate into alveoli epithelial cells, including alveolar type I and alveolar type II. CONCLUSION We established an in vitro feeder-free culture system that allows mouse ABSCs to maintain their stem cell characteristics, including self-renewal and airway epithelium differentiation potential, while keeping up in vitro expansion stability.
Collapse
Affiliation(s)
- Meirong Wu
- Department of Pulmonary and Critical Care Medicine, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, People's Republic of China
- Fujian Key Laboratory of Lung Stem Cells, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, People's Republic of China
| | - Xiaojing Zhang
- Department of Pulmonary and Critical Care Medicine, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, People's Republic of China
- Fujian Key Laboratory of Lung Stem Cells, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, People's Republic of China
| | - Yanjuan Tu
- Department of Pathology, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, People's Republic of China
| | - Wenzhao Cheng
- Fujian Key Laboratory of Lung Stem Cells, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, People's Republic of China
| | - Yiming Zeng
- Department of Pulmonary and Critical Care Medicine, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, People's Republic of China.
- Fujian Key Laboratory of Lung Stem Cells, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, People's Republic of China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong Province, People's Republic of China.
| |
Collapse
|
10
|
Lee CL, Cremona M, Farrelly A, Workman JA, Kennedy S, Aslam R, Carr A, Madden S, O’Neill B, Hennessy BT, Toomey S. Preclinical evaluation of the CDK4/6 inhibitor palbociclib in combination with a PI3K or MEK inhibitor in colorectal cancer. Cancer Biol Ther 2023; 24:2223388. [PMID: 37326340 PMCID: PMC10281467 DOI: 10.1080/15384047.2023.2223388] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 03/28/2023] [Accepted: 06/06/2023] [Indexed: 06/17/2023] Open
Abstract
BACKGROUND Studies have demonstrated the efficacy of Palbociclib (CDK 4/6 inhibitor), Gedatolisib (PI3K/mTOR dual inhibitor) and PD0325901 (MEK1/2 inhibitor) in colorectal cancer (CRC), however single agent therapeutics are often limited by the development of resistance. METHODS We compared the anti-proliferative effects of the combination of Gedatolisib and Palbociclib and Gedatolisib and PD0325901 in five CRC cell lines with varying mutational background and tested their combinations on total and phosphoprotein levels of signaling pathway proteins. RESULTS The combination of Palbociclib and Gedatolisib was superior to the combination of Palbociclib and PD0325901. The combination of Palbociclib and Gedatolisib had synergistic anti-proliferative effects in all cell lines tested [CI range: 0.11-0.69] and resulted in the suppression of S6rp (S240/244), without AKT reactivation. The combination of Palbociclib and Gedatolisib increased BAX and Bcl-2 levels in PIK3CA mutated cell lines. The combination of Palbociclib and Gedatolisib caused MAPK/ERK reactivation, as seen by an increase in expression of total EGFR, regardless of the mutational status of the cells. CONCLUSION This study shows that the combination of Palbociclib and Gedatolisib has synergistic anti-proliferative effects in both wild-type and mutated CRC cell lines. Separately, the phosphorylation of S6rp may be a promising biomarker of responsiveness to this combination.
Collapse
Affiliation(s)
- Cha Len Lee
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Mattia Cremona
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Angela Farrelly
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Julie A. Workman
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Sean Kennedy
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Razia Aslam
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Aoife Carr
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Stephen Madden
- Data Science Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Brian O’Neill
- Department of Radiation Oncology, St. Luke’s Radiation Oncology Centre, Beaumont Hospital, Dublin 9, Ireland
| | - Bryan T. Hennessy
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Sinead Toomey
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
11
|
Biswas A, Kumari A, Gaikwad DS, Pandey DK. Revolutionizing Biological Science: The Synergy of Genomics in Health, Bioinformatics, Agriculture, and Artificial Intelligence. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2023; 27:550-569. [PMID: 38100404 DOI: 10.1089/omi.2023.0197] [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: 12/17/2023]
Abstract
With climate emergency, COVID-19, and the rise of planetary health scholarship, the binary of human and ecosystem health has been deeply challenged. The interdependence of human and nonhuman animal health is increasingly acknowledged and paving the way for new frontiers in integrative biology. The convergence of genomics in health, bioinformatics, agriculture, and artificial intelligence (AI) has ushered in a new era of possibilities and applications. However, the sheer volume of genomic/multiomics big data generated also presents formidable sociotechnical challenges in extracting meaningful biological, planetary health and ecological insights. Over the past few years, AI-guided bioinformatics has emerged as a powerful tool for managing, analyzing, and interpreting complex biological datasets. The advances in AI, particularly in machine learning and deep learning, have been transforming the fields of genomics, planetary health, and agriculture. This article aims to unpack and explore the formidable range of possibilities and challenges that result from such transdisciplinary integration, and emphasizes its radically transformative potential for human and ecosystem health. The integration of these disciplines is also driving significant advancements in precision medicine and personalized health care. This presents an unprecedented opportunity to deepen our understanding of complex biological systems and advance the well-being of all life in planetary ecosystems. Notwithstanding in mind its sociotechnical, ethical, and critical policy challenges, the integration of genomics, multiomics, planetary health, and agriculture with AI-guided bioinformatics opens up vast opportunities for transnational collaborative efforts, data sharing, analysis, valorization, and interdisciplinary innovations in life sciences and integrative biology.
Collapse
Affiliation(s)
- Aakanksha Biswas
- Amity Institute of Biotechnology, Amity University Jharkhand, Ranchi, India
| | - Aditi Kumari
- Amity Institute of Biotechnology, Amity University Jharkhand, Ranchi, India
| | - D S Gaikwad
- Amity Institute of Organic Agriculture, Amity University, Noida, India
| | - Dhananjay K Pandey
- Amity Institute of Biotechnology, Amity University Jharkhand, Ranchi, India
| |
Collapse
|
12
|
Liu S, Yu CY, Wei H. Spherical nucleic acids-based nanoplatforms for tumor precision medicine and immunotherapy. Mater Today Bio 2023; 22:100750. [PMID: 37545568 PMCID: PMC10400933 DOI: 10.1016/j.mtbio.2023.100750] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/23/2023] [Accepted: 07/26/2023] [Indexed: 08/08/2023] Open
Abstract
Precise diagnosis and treatment of tumors currently still face considerable challenges due to the development of highly degreed heterogeneity in the dynamic evolution of tumors. With the rapid development of genomics, personalized diagnosis and treatment using specific genes may be a robust strategy to break through the bottleneck of traditional tumor treatment. Nevertheless, efficient in vivo gene delivery has been frequently hampered by the inherent defects of vectors and various biological barriers. Encouragingly, spherical nucleic acids (SNAs) with good modularity and programmability are excellent candidates capable of addressing traditional gene transfer-associated issues, which enables SNAs a precision nanoplatform with great potential for diverse biomedical applications. In this regard, there have been detailed reviews of SNA in drug delivery, gene regulation, and dermatology treatment. Still, to the best of our knowledge, there is no published systematic review summarizing the use of SNAs in oncology precision medicine and immunotherapy, which are considered new guidelines for oncology treatment. To this end, we summarized the notable advances in SNAs-based precision therapy and immunotherapy for tumors following a classification standard of different types of precise spatiotemporal control on active species by SNAs. Specifically, we focus on the structural diversity and programmability of SNAs. Finally, the challenges and possible solutions were discussed in the concluding remarks. This review will promote the rational design and development of SNAs for tumor-precise medicine and immunotherapy.
Collapse
Affiliation(s)
- Songbin Liu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Cui-Yun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| |
Collapse
|
13
|
Bottet B, Piton N, Selim J, Sarsam M, Guisier F, Baste JM. Beyond the Frontline: A Triple-Line Approach of Thoracic Surgeons in Lung Cancer Management-State of the Art. Cancers (Basel) 2023; 15:4039. [PMID: 37627067 PMCID: PMC10452134 DOI: 10.3390/cancers15164039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 07/26/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is now described as an extremely heterogeneous disease in its clinical presentation, histology, molecular characteristics, and patient conditions. Over the past 20 years, the management of lung cancer has evolved with positive results. Immune checkpoint inhibitors have revolutionized the treatment landscape for NSCLC in both metastatic and locally advanced stages. The identification of molecular alterations in NSCLC has also allowed the development of targeted therapies, which provide better outcomes than chemotherapy in selected patients. However, patients usually develop acquired resistance to these treatments. On the other hand, thoracic surgery has progressed thanks to minimally invasive procedures, pre-habilitation and enhanced recovery after surgery. Moreover, within thoracic surgery, precision surgery considers the patient and his/her disease in their entirety to offer the best oncologic strategy. Surgeons support patients from pre-operative rehabilitation to surgery and beyond. They are involved in post-treatment follow-up and lung cancer recurrence. When conventional therapies are no longer effective, salvage surgery can be performed on selected patients.
Collapse
Affiliation(s)
- Benjamin Bottet
- Department of General and Thoracic Surgery, Hospital Center University De Rouen, 1 Rue de Germont, F-76000 Rouen, France; (B.B.); (M.S.)
| | - Nicolas Piton
- Department of Pathology, UNIROUEN, INSERM U1245, CHU Rouen, Normandy University, F-76000 Rouen, France;
| | - Jean Selim
- Department of Anaesthesiology and Critical Care, CHU Rouen, F-76000 Rouen, France;
- INSERM EnVI UMR 1096, University of Rouen Normandy, F-76000 Rouen, France
| | - Matthieu Sarsam
- Department of General and Thoracic Surgery, Hospital Center University De Rouen, 1 Rue de Germont, F-76000 Rouen, France; (B.B.); (M.S.)
| | - Florian Guisier
- Department of Pneumology, CHU Rouen, 1 Rue de Germont, F-76000 Rouen, France;
- Clinical Investigation Center, Rouen University Hospital, CIC INSERM 1404, 1 Rue de Germont, F-76000 Rouen, France
| | - Jean-Marc Baste
- Department of General and Thoracic Surgery, Hospital Center University De Rouen, 1 Rue de Germont, F-76000 Rouen, France; (B.B.); (M.S.)
| |
Collapse
|
14
|
Kim SC, Cho YE, Shin YK, Yu HJ, Chowdhury T, Kim S, Yi KS, Choi CH, Cha SH, Park CK, Ku JL. Patient-derived glioblastoma cell lines with conserved genome profiles of the original tissue. Sci Data 2023; 10:448. [PMID: 37438387 DOI: 10.1038/s41597-023-02365-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 06/29/2023] [Indexed: 07/14/2023] Open
Abstract
Glioblastoma (GBM) is the most lethal intracranial tumor. Sequencing technologies have supported personalized therapy for precise diagnosis and optimal treatment of GBM by revealing clinically actionable molecular characteristics. Although accumulating sequence data from brain tumors and matched normal tissues have facilitated a comprehensive understanding of genomic features of GBM, these in silico evaluations could gain more biological credibility when they are verified with in vitro and in vivo models. From this perspective, GBM cell lines with whole exome sequencing (WES) datasets of matched tumor tissues and normal blood are suitable biological platforms to not only investigate molecular markers of GBM but also validate the applicability of druggable targets. Here, we provide a complete WES dataset of 26 GBM patient-derived cell lines along with their matched tumor tissues and blood to demonstrate that cell lines can mostly recapitulate genomic profiles of original tumors such as mutational signatures and copy number alterations.
Collapse
Grants
- 20009660 Ministry of Trade, Industry and Energy (Ministry of Trade, Industry and Energy, Korea)
- 20009660 Ministry of Trade, Industry and Energy (Ministry of Trade, Industry and Energy, Korea)
- 20009660 Ministry of Trade, Industry and Energy (Ministry of Trade, Industry and Energy, Korea)
- 20009660 Ministry of Trade, Industry and Energy (Ministry of Trade, Industry and Energy, Korea)
- 20009660 Ministry of Trade, Industry and Energy (Ministry of Trade, Industry and Energy, Korea)
- 20009660 Ministry of Trade, Industry and Energy (Ministry of Trade, Industry and Energy, Korea)
- 20009660 Ministry of Trade, Industry and Energy (Ministry of Trade, Industry and Energy, Korea)
- 20009660 Ministry of Trade, Industry and Energy (Ministry of Trade, Industry and Energy, Korea)
- 20009660 Ministry of Trade, Industry and Energy (Ministry of Trade, Industry and Energy, Korea)
- 20009660 Ministry of Trade, Industry and Energy (Ministry of Trade, Industry and Energy, Korea)
- 2020M3A9I6A02036061, 2021M3H9A1030151, NRF2022R1A5A102641311 National Research Foundation of Korea (NRF)
- 2020M3A9I6A02036061, 2021M3H9A1030151, NRF2022R1A5A102641311 National Research Foundation of Korea (NRF)
- 2020M3A9I6A02036061, 2021M3H9A1030151, NRF2022R1A5A102641311 National Research Foundation of Korea (NRF)
Collapse
Affiliation(s)
- Soon-Chan Kim
- Korean Cell Line Bank, Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Cancer Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Young-Eun Cho
- Korean Cell Line Bank, Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Cancer Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
| | - Young-Kyoung Shin
- Korean Cell Line Bank, Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Cancer Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Hyeon Jong Yu
- Department of Neurosurgery, Seoul National University Hospital and Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Tamrin Chowdhury
- Department of Neurosurgery, Seoul National University Hospital and Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Sojin Kim
- Department of Neurosurgery, Seoul National University Hospital and Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Kyung Sik Yi
- Department of Radiology, Chungbuk National University Hospital and Chungbuk National University College of Medicine, Cheongju, Chung Buk, 28644, Republic of Korea
| | - Chi-Hoon Choi
- Department of Radiology, Chungbuk National University Hospital and Chungbuk National University College of Medicine, Cheongju, Chung Buk, 28644, Republic of Korea
- Chungbuk National University College of Medicine, Cheongju, Chung Buk, 28644, Republic of Korea
| | - Sang-Hoon Cha
- Department of Radiology, Chungbuk National University Hospital and Chungbuk National University College of Medicine, Cheongju, Chung Buk, 28644, Republic of Korea.
- Chungbuk National University College of Medicine, Cheongju, Chung Buk, 28644, Republic of Korea.
| | - Chul-Kee Park
- Department of Neurosurgery, Seoul National University Hospital and Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
| | - Ja-Lok Ku
- Korean Cell Line Bank, Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Cancer Research Institute, Seoul National University, Seoul, 03080, Republic of Korea.
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea.
| |
Collapse
|
15
|
Tebon PJ, Wang B, Markowitz AL, Davarifar A, Tsai BL, Krawczuk P, Gonzalez AE, Sartini S, Murray GF, Nguyen HTL, Tavanaie N, Nguyen TL, Boutros PC, Teitell MA, Soragni A. Drug screening at single-organoid resolution via bioprinting and interferometry. Nat Commun 2023; 14:3168. [PMID: 37280220 PMCID: PMC10244450 DOI: 10.1038/s41467-023-38832-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 05/17/2023] [Indexed: 06/08/2023] Open
Abstract
High throughput drug screening is an established approach to investigate tumor biology and identify therapeutic leads. Traditional platforms use two-dimensional cultures which do not accurately reflect the biology of human tumors. More clinically relevant model systems such as three-dimensional tumor organoids can be difficult to scale and screen. Manually seeded organoids coupled to destructive endpoint assays allow for the characterization of treatment response, but do not capture transitory changes and intra-sample heterogeneity underlying clinically observed resistance to therapy. We present a pipeline to generate bioprinted tumor organoids linked to label-free, time-resolved imaging via high-speed live cell interferometry (HSLCI) and machine learning-based quantitation of individual organoids. Bioprinting cells gives rise to 3D structures with unaltered tumor histology and gene expression profiles. HSLCI imaging in tandem with machine learning-based segmentation and classification tools enables accurate, label-free parallel mass measurements for thousands of organoids. We demonstrate that this strategy identifies organoids transiently or persistently sensitive or resistant to specific therapies, information that could be used to guide rapid therapy selection.
Collapse
Affiliation(s)
- Peyton J Tebon
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA
| | - Bowen Wang
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
- Department of Pathology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Alexander L Markowitz
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, University of California Los Angeles, Los Angeles, CA, USA
- Institute for Precision Health, University of California Los Angeles, Los Angeles, CA, USA
| | - Ardalan Davarifar
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, University of California Los Angeles, Los Angeles, CA, USA
| | - Brandon L Tsai
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, University of California Los Angeles, Los Angeles, CA, USA
- Institute for Precision Health, University of California Los Angeles, Los Angeles, CA, USA
| | - Patrycja Krawczuk
- Information Sciences Institute, University of Southern California, Marina Del Rey, CA, USA
| | - Alfredo E Gonzalez
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, University of California Los Angeles, Los Angeles, CA, USA
- Institute for Precision Health, University of California Los Angeles, Los Angeles, CA, USA
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA, USA
| | - Sara Sartini
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Graeme F Murray
- Department of Physics, Virginia Commonwealth University, Richmond, VA, USA
| | - Huyen Thi Lam Nguyen
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Nasrin Tavanaie
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Thang L Nguyen
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
- Department of Pathology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Paul C Boutros
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, University of California Los Angeles, Los Angeles, CA, USA
- Institute for Precision Health, University of California Los Angeles, Los Angeles, CA, USA
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA, USA
- California NanoSystems Institute, University of California Los Angeles, Los Angeles, CA, USA
- Department of Urology, University of California Los Angeles, Los Angeles, CA, USA
| | - Michael A Teitell
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA.
- Department of Pathology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA, USA.
- California NanoSystems Institute, University of California Los Angeles, Los Angeles, CA, USA.
- Department of Pediatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| | - Alice Soragni
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA, USA.
- California NanoSystems Institute, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
16
|
Zhao B, Lv Y. Suspension state and shear stress enhance breast tumor cells EMT through YAP by microRNA-29b. Cell Biol Toxicol 2023; 39:1037-1052. [PMID: 34618275 DOI: 10.1007/s10565-021-09661-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 09/24/2021] [Indexed: 12/11/2022]
Abstract
Except for biochemical effects, suspension state (Sus) is proved to induce epithelial-mesenchymal transition (EMT) of circulating tumor cells (CTCs) mechanically. However, the difference between the effects of the mechanical microenvironment in capillaries (simplified as shear stress (SS) and Sus) and single Sus on EMT is unclear, nor the underlying mechanism. Here, breast tumor cells (BTCs) were loaded with Sus and SS to mimic the situation of CTCs stimulated by these two kinds of mechanics. It was demonstrated that the EMT of BTCs was enhanced by Sus and SS and the mechanotransductor yes-associated protein (YAP) was partially cytoplasmic stored with microRNA (miR)-29b decreased, which was detected by miR sequencing. Though it couldn't possess a feedback regulation, YAP promoted miR-29b expression and posttranscriptionally regulated BTCs EMT through miR-29b, where transforming growth factor β involved. Analysis of clinical database showed that high miR-29b expression was beneficial to high survival rate stabilizing its role of tumor suppressor. This study discovers the mechanism that Sus and SS promote BTCs EMT by YAP through miR-29b posttranscriptionally and highlight the potential of YAP and miR-29b in tumor therapy. The combination of suspension state and shear stress promotes transforming growth factor β involved epithelial-mesenchymal transition by yes-associated protein through microRNA-29b.
Collapse
Affiliation(s)
- Boyuan Zhao
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing, 400044, People's Republic of China
| | - Yonggang Lv
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing, 400044, People's Republic of China.
| |
Collapse
|
17
|
Santhanam B, Oikonomou P, Tavazoie S. Systematic assessment of prognostic molecular features across cancers. CELL GENOMICS 2023; 3:100262. [PMID: 36950380 PMCID: PMC10025453 DOI: 10.1016/j.xgen.2023.100262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 09/29/2022] [Accepted: 01/12/2023] [Indexed: 02/05/2023]
Abstract
Precision oncology promises accurate prediction of disease trajectories by utilizing molecular features of tumors. We present a systematic analysis of the prognostic potential of diverse molecular features across large cancer cohorts. We find that the mRNA expression of biologically coherent sets of genes (modules) is substantially more predictive of patient survival than single-locus genomic and transcriptomic aberrations. Extending our analysis beyond existing curated gene modules, we find a large novel class of highly prognostic DNA/RNA cis-regulatory modules associated with dynamic gene expression within cancers. Remarkably, in more than 82% of cancers, modules substantially improve survival stratification compared with conventional clinical factors and prominent genomic aberrations. The prognostic potential of cancer modules generalizes to external cohorts better than conventionally used single-gene features. Finally, a machine-learning framework demonstrates the combined predictive power of multiple modules, yielding prognostic models that perform substantially better than existing histopathological and clinical factors in common use.
Collapse
Affiliation(s)
- Balaji Santhanam
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
- Department of Systems Biology, Columbia University, New York, NY 10032, USA
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA
- Irving Institute for Cancer Dynamics, Columbia University, New York, NY 10032, USA
| | - Panos Oikonomou
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
- Department of Systems Biology, Columbia University, New York, NY 10032, USA
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA
- Irving Institute for Cancer Dynamics, Columbia University, New York, NY 10032, USA
| | - Saeed Tavazoie
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
- Department of Systems Biology, Columbia University, New York, NY 10032, USA
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA
- Irving Institute for Cancer Dynamics, Columbia University, New York, NY 10032, USA
| |
Collapse
|
18
|
Caputo V, Ciardiello F, Corte CMD, Martini G, Troiani T, Napolitano S. Diagnostic value of liquid biopsy in the era of precision medicine: 10 years of clinical evidence in cancer. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:102-138. [PMID: 36937316 PMCID: PMC10017193 DOI: 10.37349/etat.2023.00125] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 11/13/2022] [Indexed: 03/06/2023] Open
Abstract
Liquid biopsy is a diagnostic repeatable test, which in last years has emerged as a powerful tool for profiling cancer genomes in real-time with minimal invasiveness and tailoring oncological decision-making. It analyzes different blood-circulating biomarkers and circulating tumor DNA (ctDNA) is the preferred one. Nevertheless, tissue biopsy remains the gold standard for molecular evaluation of solid tumors whereas liquid biopsy is a complementary tool in many different clinical settings, such as treatment selection, monitoring treatment response, cancer clonal evolution, prognostic evaluation, as well as the detection of early disease and minimal residual disease (MRD). A wide number of technologies have been developed with the aim of increasing their sensitivity and specificity with acceptable costs. Moreover, several preclinical and clinical studies have been conducted to better understand liquid biopsy clinical utility. Anyway, several issues are still a limitation of its use such as false positive and negative results, results interpretation, and standardization of the panel tests. Although there has been rapid development of the research in these fields and recent advances in the clinical setting, many clinical trials and studies are still needed to make liquid biopsy an instrument of clinical routine. This review provides an overview of the current and future clinical applications and opening questions of liquid biopsy in different oncological settings, with particular attention to ctDNA liquid biopsy.
Collapse
Affiliation(s)
- Vincenza Caputo
- Medical Oncology, Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80131 Napoli, Italy
| | - Fortunato Ciardiello
- Medical Oncology, Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80131 Napoli, Italy
| | - Carminia Maria Della Corte
- Medical Oncology, Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80131 Napoli, Italy
| | - Giulia Martini
- Medical Oncology, Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80131 Napoli, Italy
| | - Teresa Troiani
- Medical Oncology, Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80131 Napoli, Italy
| | - Stefania Napolitano
- Medical Oncology, Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80131 Napoli, Italy
| |
Collapse
|
19
|
He N, Xi Y, Yu D, Yu C, Shen W. Construction of IL-1 signalling pathway correlation model in lung adenocarcinoma and association with immune microenvironment prognosis and immunotherapy: Multi-data validation. Front Immunol 2023; 14:1116789. [PMID: 36865560 PMCID: PMC9972222 DOI: 10.3389/fimmu.2023.1116789] [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: 12/05/2022] [Accepted: 02/01/2023] [Indexed: 02/12/2023] Open
Abstract
Numerous studies have confirmed the inextricable link between inflammation and malignancy, which is also involved in developing lung adenocarcinoma, where IL-1 signalling is crucial. However, the predictive role of single gene biomarkers is insufficient, and more accurate prognostic models are needed. We downloaded data related to lung adenocarcinoma patients from the GDC, GEO, TISCH2 and TCGA databases for data analysis, model construction and differential gene expression analysis. The genes of IL-1 signalling-related factors were screened from published papers for subgroup typing and predictive correlation analysis. Five prognostic genes associated with IL-1 signalling were finally identified to construct prognostic prediction models. The K-M curves indicated that the prognostic models had significant predictive efficacy. Further immune infiltration scores showed that IL-1 signalling was mainly associated with enhanced immune cells, drug sensitivity of model genes was analysed using the GDSC database, and correlation of critical memories with cell subpopulation components was observed using single-cell analysis. In conclusion, we propose a predictive model based on IL-1 signalling-related factors, a non-invasive predictive approach for genomic characterisation, in predicting patients' survival outcomes. The therapeutic response has shown satisfactory and effective performance. More interdisciplinary areas combining medicine and electronics will be explored in the future.
Collapse
Affiliation(s)
- Ningning He
- Department of Thoracic Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
| | - Yong Xi
- Department of Thoracic Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China,*Correspondence: Yong Xi,
| | - Dongyue Yu
- College of Life Sciences, Nankai University, Tianjin, China
| | - Chaoqun Yu
- Department of Thoracic Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
| | - Weiyu Shen
- Department of Thoracic Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
20
|
Stulpinas A, Sereika M, Vitkeviciene A, Imbrasaite A, Krestnikova N, Kalvelyte AV. Crosstalk between protein kinases AKT and ERK1/2 in human lung tumor-derived cell models. Front Oncol 2023; 12:1045521. [PMID: 36686779 PMCID: PMC9848735 DOI: 10.3389/fonc.2022.1045521] [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: 09/15/2022] [Accepted: 11/28/2022] [Indexed: 01/06/2023] Open
Abstract
There is no doubt that cell signaling manipulation is a key strategy for anticancer therapy. Furthermore, cell state determines drug response. Thus, establishing the relationship between cell state and therapeutic sensitivity is essential for the development of cancer therapies. In the era of personalized medicine, the use of patient-derived ex vivo cell models is a promising approach in the translation of key research findings into clinics. Here, we were focused on the non-oncogene dependencies of cell resistance to anticancer treatments. Signaling-related mechanisms of response to inhibitors of MEK/ERK and PI3K/AKT pathways (regulators of key cellular functions) were investigated using a panel of patients' lung tumor-derived cell lines with various stemness- and EMT-related markers, varying degrees of ERK1/2 and AKT phosphorylation, and response to anticancer treatment. The study of interactions between kinases was the goal of our research. Although MEK/ERK and PI3K/AKT interactions are thought to be cell line-specific, where oncogenic mutations have a decisive role, we demonstrated negative feedback loops between MEK/ERK and PI3K/AKT signaling pathways in all cell lines studied, regardless of genotype and phenotype differences. Our work showed that various and distinct inhibitors of ERK signaling - selumetinib, trametinib, and SCH772984 - increased AKT phosphorylation, and conversely, inhibitors of AKT - capivasertib, idelalisib, and AKT inhibitor VIII - increased ERK phosphorylation in both control and cisplatin-treated cells. Interaction between kinases, however, was dependent on cellular state. The feedback between ERK and AKT was attenuated by the focal adhesion kinase inhibitor PF573228, and in cells grown in suspension, showing the possible role of extracellular contacts in the regulation of crosstalk between kinases. Moreover, studies have shown that the interplay between MEK/ERK and PI3K/AKT signaling pathways may be dependent on the strength of the chemotherapeutic stimulus. The study highlights the importance of spatial location of the cells and the strength of the treatment during anticancer therapy.
Collapse
|
21
|
Rolfs F, de Goeij-de Haas RR, Knol JC, Piersma SR, Jimenez CR. Phosphoproteomics After Guanidinium Thiocyanate Extraction of Tissue Biopsies. Methods Mol Biol 2023; 2718:285-302. [PMID: 37665466 DOI: 10.1007/978-1-0716-3457-8_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: 09/05/2023]
Abstract
Proteogenomic analysis is emerging as an advantageous tool to assist personalized therapy decisions in clinical health care and integrates complementary information from the genome, transcriptome, and (phospho)proteome. A prerequisite for such analysis is a workflow for the simultaneous isolation of DNA, RNA, and protein from a single sample that does not compromise the different biological molecules and their examination. Focusing on the phosphoproteomic aspect of this workflow, we here provide detailed information on our protocol, which is based on commonly used acid guanidinium thiocyanate-phenol-chloroform (AGPC) extraction with RNA-Bee. We describe the necessary steps for biopsy collection, cryoprocessing, and protein extraction. We further share our practice on protein digestion and cleanup of small samples (200 μg protein) and describe settings for automated IMAC-based phosphopeptide enrichment with the AssayMAP Bravo platform.
Collapse
Affiliation(s)
- Frank Rolfs
- Department Medical Oncology, OncoProteomics Laboratory, Cancer Center Amsterdam, Amsterdam UMC, Location VUmc, Amsterdam, The Netherlands
| | - Richard R de Goeij-de Haas
- Department Medical Oncology, OncoProteomics Laboratory, Cancer Center Amsterdam, Amsterdam UMC, Location VUmc, Amsterdam, The Netherlands
| | - Jaco C Knol
- Department Medical Oncology, OncoProteomics Laboratory, Cancer Center Amsterdam, Amsterdam UMC, Location VUmc, Amsterdam, The Netherlands
| | - Sander R Piersma
- Department Medical Oncology, OncoProteomics Laboratory, Cancer Center Amsterdam, Amsterdam UMC, Location VUmc, Amsterdam, The Netherlands
| | - Connie R Jimenez
- Department Medical Oncology, OncoProteomics Laboratory, Cancer Center Amsterdam, Amsterdam UMC, Location VUmc, Amsterdam, The Netherlands.
| |
Collapse
|
22
|
Houweling M, Giczewska A, Abdul K, Nieuwenhuis N, Küçükosmanoglu A, Pastuszak K, Buijsman RC, Wesseling P, Wedekind L, Noske D, Supernat A, Bailey D, Watts C, Wurdinger T, Westerman BA. Screening of predicted synergistic multi-target therapies in glioblastoma identifies new treatment strategies. Neurooncol Adv 2023; 5:vdad073. [PMID: 37455945 PMCID: PMC10347974 DOI: 10.1093/noajnl/vdad073] [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] [Indexed: 07/18/2023] Open
Abstract
Background IDH-wildtype glioblastoma (GBM) is a highly malignant primary brain tumor with a median survival of 15 months after standard of care, which highlights the need for improved therapy. Personalized combination therapy has shown to be successful in many other tumor types and could be beneficial for GBM patients. Methods We performed the largest drug combination screen to date in GBM, using a high-throughput effort where we selected 90 drug combinations for their activity onto 25 patient-derived GBM cultures. 43 drug combinations were selected for interaction analysis based on their monotherapy efficacy and were tested in a short-term (3 days) as well as long-term (18 days) assay. Synergy was assessed using dose-equivalence and multiplicative survival metrics. Results We observed a consistent synergistic interaction for 15 out of 43 drug combinations on patient-derived GBM cultures. From these combinations, 11 out of 15 drug combinations showed a longitudinal synergistic effect on GBM cultures. The highest synergies were observed in the drug combinations Lapatinib with Thapsigargin and Lapatinib with Obatoclax Mesylate, both targeting epidermal growth factor receptor and affecting the apoptosis pathway. To further elaborate on the apoptosis cascade, we investigated other, more clinically relevant, apoptosis inducers and observed a strong synergistic effect while combining Venetoclax (BCL targeting) and AZD5991 (MCL1 targeting). Conclusions Overall, we have identified via a high-throughput drug screening several new treatment strategies for GBM. Moreover, an exceptionally strong synergistic interaction was discovered between kinase targeting and apoptosis induction which is suitable for further clinical evaluation as multi-targeted combination therapy.
Collapse
Affiliation(s)
- Megan Houweling
- Department of Neurosurgery, Amsterdam UMC location Vrije Universiteit Amsterdam, Boelelaan 1117, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Brain tumor center Amsterdam, Amsterdam, The Netherlands
- WINDOW consortium, Amsterdam, The Netherlands (www.window-consortium.org)
| | | | | | - Ninke Nieuwenhuis
- Department of Neurosurgery, Amsterdam UMC location Vrije Universiteit Amsterdam, Boelelaan 1117, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Brain tumor center Amsterdam, Amsterdam, The Netherlands
| | - Asli Küçükosmanoglu
- Department of Neurosurgery, Amsterdam UMC location Vrije Universiteit Amsterdam, Boelelaan 1117, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Brain tumor center Amsterdam, Amsterdam, The Netherlands
- WINDOW consortium, Amsterdam, The Netherlands (www.window-consortium.org)
| | - Krzysztof Pastuszak
- Medical University of Gdańsk, Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, 80-211 Gdańsk, Poland
- Department of Algorithms and Systems Modelling, Faculty of Electronics, Telecommunication and Informatics, Gdańsk University of Technology, 80-233 Gdańsk, Poland
- Medical University of Gdańsk, Centre of Biostatistics and Bioinformatics Analysis, 80-211 Gdańsk, Poland
| | | | - Pieter Wesseling
- Department of Pathology, Amsterdam UMC location Vrije Universiteit Amsterdam, Boelelaan 1117, Amsterdam, The Netherlands
- Princess Maxima Center for Pediatric Oncology, Laboratory for Childhood Cancer Pathology, Utrecht, The Netherlands
| | - Laurine Wedekind
- Department of Neurosurgery, Amsterdam UMC location Vrije Universiteit Amsterdam, Boelelaan 1117, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Brain tumor center Amsterdam, Amsterdam, The Netherlands
- WINDOW consortium, Amsterdam, The Netherlands (www.window-consortium.org)
| | - David Noske
- Department of Neurosurgery, Amsterdam UMC location Vrije Universiteit Amsterdam, Boelelaan 1117, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Brain tumor center Amsterdam, Amsterdam, The Netherlands
| | - Anna Supernat
- Medical University of Gdańsk, Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, 80-211 Gdańsk, Poland
- Medical University of Gdańsk, Centre of Biostatistics and Bioinformatics Analysis, 80-211 Gdańsk, Poland
| | - David Bailey
- IOTA Pharmaceuticals Ltd, St Johns Innovation Centre, Cowley Road, Cambridge, CB4 0WS, UK
- WINDOW consortium, Amsterdam, The Netherlands (www.window-consortium.org)
| | - Colin Watts
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
- WINDOW consortium, Amsterdam, The Netherlands (www.window-consortium.org)
| | - Thomas Wurdinger
- Department of Neurosurgery, Amsterdam UMC location Vrije Universiteit Amsterdam, Boelelaan 1117, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Brain tumor center Amsterdam, Amsterdam, The Netherlands
- WINDOW consortium, Amsterdam, The Netherlands (www.window-consortium.org)
| | - Bart A Westerman
- Corresponding Author: Dr. Bart A. Westerman, Department of Neurosurgery, Amsterdam UMC location Vrije Universiteit Amsterdam, Boelelaan 1117, Amsterdam, The Netherlands ()
| |
Collapse
|
23
|
Landscape of somatic alterations in large-scale solid tumors from an Asian population. Nat Commun 2022; 13:4264. [PMID: 35871175 PMCID: PMC9308789 DOI: 10.1038/s41467-022-31780-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/01/2022] [Indexed: 12/24/2022] Open
Abstract
Extending the benefits of tumor molecular profiling for all cancer patients requires a comprehensive analysis of tumor genomes across distinct patient populations worldwide. In this study, we perform deep next-generation DNA sequencing (NGS) from tumor tissues and matched blood specimens from over 10,000 patients in China by using a 450-gene comprehensive assay, developed and implemented under international clinical regulations. We perform a comprehensive comparison of somatically altered genes, the distribution of tumor mutational burden (TMB), gene fusion patterns, and the spectrum of various somatic alterations between Chinese and American patient populations. Here, we show 64% of cancers from Chinese patients in this study have clinically actionable genomic alterations, which may affect clinical decisions related to targeted therapy or immunotherapy. These findings describe the similarities and differences between tumors from Chinese and American patients, providing valuable information for personalized medicine. Understanding the mutation landscape of cancer may enable the development of more targeted therapies. Here, the authors sequence a panel of genes in a large Asian cohort and compare to American cohorts and find 64% of the Asian patients have actionable mutations.
Collapse
|
24
|
Singha M, Pu L, Stanfield BA, Uche IK, Rider PJF, Kousoulas KG, Ramanujam J, Brylinski M. Artificial intelligence to guide precision anticancer therapy with multitargeted kinase inhibitors. BMC Cancer 2022; 22:1211. [PMID: 36434556 PMCID: PMC9694576 DOI: 10.1186/s12885-022-10293-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 11/07/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Vast amounts of rapidly accumulating biological data related to cancer and a remarkable progress in the field of artificial intelligence (AI) have paved the way for precision oncology. Our recent contribution to this area of research is CancerOmicsNet, an AI-based system to predict the therapeutic effects of multitargeted kinase inhibitors across various cancers. This approach was previously demonstrated to outperform other deep learning methods, graph kernel models, molecular docking, and drug binding pocket matching. METHODS CancerOmicsNet integrates multiple heterogeneous data by utilizing a deep graph learning model with sophisticated attention propagation mechanisms to extract highly predictive features from cancer-specific networks. The AI-based system was devised to provide more accurate and robust predictions than data-driven therapeutic discovery using gene signature reversion. RESULTS Selected CancerOmicsNet predictions obtained for "unseen" data are positively validated against the biomedical literature and by live-cell time course inhibition assays performed against breast, pancreatic, and prostate cancer cell lines. Encouragingly, six molecules exhibited dose-dependent antiproliferative activities, with pan-CDK inhibitor JNJ-7706621 and Src inhibitor PP1 being the most potent against the pancreatic cancer cell line Panc 04.03. CONCLUSIONS CancerOmicsNet is a promising AI-based platform to help guide the development of new approaches in precision oncology involving a variety of tumor types and therapeutics.
Collapse
Affiliation(s)
- Manali Singha
- grid.64337.350000 0001 0662 7451Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803 USA
| | - Limeng Pu
- grid.64337.350000 0001 0662 7451Center for Computation and Technology, Louisiana State University, Baton Rouge, LA 70803 USA
| | - Brent A. Stanfield
- grid.64337.350000 0001 0662 7451Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803 USA
| | - Ifeanyi K. Uche
- grid.64337.350000 0001 0662 7451Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803 USA ,grid.64337.350000 0001 0662 7451Division of Biotechnology and Molecular Medicine, Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803 USA ,grid.279863.10000 0000 8954 1233School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112 USA
| | - Paul J. F. Rider
- grid.64337.350000 0001 0662 7451Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803 USA ,grid.64337.350000 0001 0662 7451Division of Biotechnology and Molecular Medicine, Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803 USA
| | - Konstantin G. Kousoulas
- grid.64337.350000 0001 0662 7451Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803 USA ,grid.64337.350000 0001 0662 7451Division of Biotechnology and Molecular Medicine, Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803 USA
| | - J. Ramanujam
- grid.64337.350000 0001 0662 7451Center for Computation and Technology, Louisiana State University, Baton Rouge, LA 70803 USA ,grid.64337.350000 0001 0662 7451Division of Electrical and Computer Engineering, Louisiana State University, Baton Rouge, LA 70803 USA
| | - Michal Brylinski
- grid.64337.350000 0001 0662 7451Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803 USA ,grid.64337.350000 0001 0662 7451Center for Computation and Technology, Louisiana State University, Baton Rouge, LA 70803 USA
| |
Collapse
|
25
|
Zhanpeng H, Jiekang W. A Multiview Clustering Method With Low-Rank and Sparsity Constraints for Cancer Subtyping. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:3213-3223. [PMID: 34705654 DOI: 10.1109/tcbb.2021.3122917] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Multiomics data clustering is one of the major challenges in the field of precision medicine. Integration of multiomics data for cancer subtyping can improve the understanding on cancer and reveal systems-level insights. How to integrate multiomics data for accurate cancer subtyping is an interesting and challenging research problem. To capture the global and the local structure of omics data, a novel framework for integrating multiomics data is proposed for cancer subtyping. Multiview clustering with low-rank and sparsity constraints (MVCLRS) can measure the local similarities of samples in each omics data and obtain global consensus structures by integrating the multiomics data. The main insight provided by MVCLRS is that low-rank sparse subspace clustering for the construction of an affinity matrix can best capture the local similarities in omics data. Extensive testing is conducted on 10 real world cancer datasets with multiomics from The Cancer Genome Atlas. Compared with 10 state-of-the-art multiomics clustering algorithms, the MVCLRS performs better in the 10 cancer datasets by providing its clustering results with at least one enriched clinical label in nine of ten cancer subtypes, the most of any method.
Collapse
|
26
|
Mariappan R, Jayagopal A, Sien HZ, Rajan V. Neural Collective Matrix Factorization for integrated analysis of heterogeneous biomedical data. Bioinformatics 2022; 38:4554-4561. [PMID: 35929808 DOI: 10.1093/bioinformatics/btac543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 06/30/2022] [Accepted: 08/03/2022] [Indexed: 12/24/2022] Open
Abstract
MOTIVATION In many biomedical studies, there arises the need to integrate data from multiple directly or indirectly related sources. Collective matrix factorization (CMF) and its variants are models designed to collectively learn from arbitrary collections of matrices. The latent factors learnt are rich integrative representations that can be used in downstream tasks, such as clustering or relation prediction with standard machine-learning models. Previous CMF-based methods have numerous modeling limitations. They do not adequately capture complex non-linear interactions and do not explicitly model varying sparsity and noise levels in the inputs, and some cannot model inputs with multiple datatypes. These inadequacies limit their use on many biomedical datasets. RESULTS To address these limitations, we develop Neural Collective Matrix Factorization (NCMF), the first fully neural approach to CMF. We evaluate NCMF on relation prediction tasks of gene-disease association prediction and adverse drug event prediction, using multiple datasets. In each case, data are obtained from heterogeneous publicly available databases and used to learn representations to build predictive models. NCMF is found to outperform previous CMF-based methods and several state-of-the-art graph embedding methods for representation learning in our experiments. Our experiments illustrate the versatility and efficacy of NCMF in representation learning for seamless integration of heterogeneous data. AVAILABILITY AND IMPLEMENTATION https://github.com/ajayago/NCMF_bioinformatics. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Ragunathan Mariappan
- Department of Information Systems and Analytics, School of Computing, National University of Singapore, Singapore 117417, Singapore
| | - Aishwarya Jayagopal
- Department of Information Systems and Analytics, School of Computing, National University of Singapore, Singapore 117417, Singapore
| | - Ho Zong Sien
- Department of Information Systems and Analytics, School of Computing, National University of Singapore, Singapore 117417, Singapore
| | - Vaibhav Rajan
- Department of Information Systems and Analytics, School of Computing, National University of Singapore, Singapore 117417, Singapore
| |
Collapse
|
27
|
Kang W, Tong Y, Zhang W, Jian M, Zhang A, Ren G, Fan H, Yang J. Computational Biology Predicts the Efficacy of Tumor Immune Checkpoint Blockade. BIOMED RESEARCH INTERNATIONAL 2022; 2022:6087751. [PMID: 36212709 PMCID: PMC9534640 DOI: 10.1155/2022/6087751] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/09/2022] [Accepted: 09/16/2022] [Indexed: 12/02/2022]
Abstract
Tumor immunotherapy is considered as one of the most promising methods in cancer treatment in recent years. Immune checkpoint blockade (ICB) can activate immune cells to destroy tumors by relieving the inhibitory pathway of tumor cells to immune cells. In silico prediction of the ICB response is an important step toward achieving effective and personalized cancer immunotherapy. Although immune checkpoint inhibitors have shown exciting clinical effects in the treatment of many types of tumors, there are still some clinical problems in practical application, such as low response rate and large individualized differences. How to predict the efficacy of effective individualized immune checkpoint inhibitors for tumor patients based on specific biomarkers and computational models is one of the key issues in the immunotherapy of this kind of tumor. In our work, from the five levels of genome level, transcription level, epigenetic level, microbial taxonomy level, and the immune cell infiltration profile level, the biomarkers and in silico calculation methods that affect the efficacy of tumor immune checkpoint inhibitors are comprehensively summarized.
Collapse
Affiliation(s)
- Wenyi Kang
- Department of Oncology, The First Affiliated Hospital of Yangtze University, Jingzhou, 434000 Hubei, China
| | - Yao Tong
- School of Medicine, Wuhan University of Science and Technology, Wuhan, China 430061
| | - Weijia Zhang
- Department of Oncology, The First Affiliated Hospital of Yangtze University, Jingzhou, 434000 Hubei, China
| | - Mengru Jian
- Department of Oncology, The First Affiliated Hospital of Yangtze University, Jingzhou, 434000 Hubei, China
| | - Anqi Zhang
- Department of Oncology, The First Affiliated Hospital of Yangtze University, Jingzhou, 434000 Hubei, China
| | - Guoqing Ren
- Department of Laboratory Medicine, Chuzhou Maternal and Child Health Care and Family Planning Service Center, Chuzhou 239000, China
| | - Hao Fan
- Huanggang Central Hospital of Yangtze University, Huanggang 43800, China
| | - Jiyuan Yang
- Department of Oncology, The First Affiliated Hospital of Yangtze University, Jingzhou, 434000 Hubei, China
| |
Collapse
|
28
|
Rong D, Chen X, Xiao J, Liu D, Ni X, Tong X, Wang H. Histone methylation modification patterns and relevant M-RiskScore in acute myeloid leukemia. Heliyon 2022; 8:e10610. [PMID: 36164519 PMCID: PMC9508520 DOI: 10.1016/j.heliyon.2022.e10610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 07/13/2022] [Accepted: 09/07/2022] [Indexed: 12/05/2022] Open
Abstract
Objective We tried to identify novel molecular subtypes of acute myeloid leukemia (AML) associated with histone methylation and established a relevant scoring system to predict treatment response and prognosis of AML. Methods Gene expression data and clinical characteristics of patients with AML were obtained from The Cancer Genome Atlas (TCGA) database and Gene Expression Omnibus (GEO) database. Molecular subtyping was carried out by consensus clustering analysis, based on the expression of 24 histone methylation modification regulators (HMMRs). The clinical and biological features of each clustered pattern were taken into account. The scoring system was constructed by using differential expression analysis, Cox regression method and lasso regression analysis. Subsequently, the scoring system in the roles of prognostic and chemotherapeutic prediction of AML were explored. Finally, an independent GSE dataset was used for validating the established clustering system. Results Two distinct subtypes of AML were identified based on the expression of the 24 HMMRs, which exhibited remarkable differences in several clinical and biological characteristics, including HMMRs expression, AML-M0 distribution, NPM1 mutation, tumor mutation burden, somatic mutations, pathway activation, immune cell infiltration and patient survival. The scoring system, M-RiskScore, was established. Integrated analysis demonstrated that patients with the low M-RiskScore displayed a prominent survival advantage and a good response to decitabine treatment, while patients with high M-RiskScore have resistance to decitabine, but they could benefit from IA regimen therapy. Conclusion Detection of HMMRs expression would be a potential strategy for AML subtyping. Meanwhile, targeting histone methylation would be a preferred strategy for either AML-M0 or NPM1 mutant patients. M-RiskScore was a useful prognostic biomarker and a guide for the choice of appropriate chemotherapy strategy.
Collapse
Affiliation(s)
- Dade Rong
- The First Affiliated Hospital, Sun Yat-sen University, 58 Second Zhongshan Road, Guangzhou, 510080, China.,Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 74 Second Zhongshan Road, Guangzhou, 510080, China.,Faculty of Health Sciences, University of Macau, Macau, China
| | - Xiaomin Chen
- The First Affiliated Hospital, Sun Yat-sen University, 58 Second Zhongshan Road, Guangzhou, 510080, China.,GenePlus, Beijing, China
| | - Jing Xiao
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Department of Clinical Laboratory, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, 519000, China
| | - Daiyuan Liu
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 74 Second Zhongshan Road, Guangzhou, 510080, China
| | - Xiangna Ni
- The First Affiliated Hospital, Sun Yat-sen University, 58 Second Zhongshan Road, Guangzhou, 510080, China
| | - Xiuzhen Tong
- The First Affiliated Hospital, Sun Yat-sen University, 58 Second Zhongshan Road, Guangzhou, 510080, China
| | - Haihe Wang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 74 Second Zhongshan Road, Guangzhou, 510080, China
| |
Collapse
|
29
|
Iranzo J, Gruenhagen G, Calle-Espinosa J, Koonin EV. Pervasive conditional selection of driver mutations and modular epistasis networks in cancer. Cell Rep 2022; 40:111272. [PMID: 36001960 DOI: 10.1016/j.celrep.2022.111272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 04/18/2022] [Accepted: 08/05/2022] [Indexed: 11/19/2022] Open
Abstract
Cancer driver mutations often display mutual exclusion or co-occurrence, underscoring the key role of epistasis in carcinogenesis. However, estimating the magnitude of epistasis and quantifying its effect on tumor evolution remains a challenge. We develop a method (Coselens) to quantify conditional selection on the excess of nonsynonymous substitutions in cancer genes. Coselens infers the number of drivers per gene in different partitions of a cancer genomics dataset using covariance-based mutation models and determines whether coding mutations in a gene affect selection for drivers in any other gene. Using Coselens, we identify 296 conditionally selected gene pairs across 16 cancer types in the TCGA dataset. Conditional selection affects 25%-50% of driver substitutions in tumors with >2 drivers. Conditionally co-selected genes form modular networks, whose structures challenge the traditional interpretation of within-pathway mutual exclusivity and across-pathway synergy, suggesting a more complex scenario where gene-specific across-pathway epistasis shapes differentiated cancer subtypes.
Collapse
Affiliation(s)
- Jaime Iranzo
- Centro de Biotecnología y Genómica de Plantas, Universidad Politécnica de Madrid (UPM) - Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Madrid, Spain; Institute for Biocomputation and Physics of Complex Systems (BIFI), University of Zaragoza, Zaragoza, Spain.
| | - George Gruenhagen
- Institute of Bioengineering and Biosciences, School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Jorge Calle-Espinosa
- Centro de Biotecnología y Genómica de Plantas, Universidad Politécnica de Madrid (UPM) - Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Madrid, Spain
| | - Eugene V Koonin
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
30
|
Strzebonska K, Blukacz M, Wasylewski MT, Polak M, Gyawali B, Waligora M. Risk and benefit for umbrella trials in oncology: a systematic review and meta-analysis. BMC Med 2022; 20:219. [PMID: 35799149 PMCID: PMC9264503 DOI: 10.1186/s12916-022-02420-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 05/30/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Umbrella clinical trials in precision oncology are designed to tailor therapies to the specific genetic changes within a tumor. Little is known about the risk/benefit ratio for umbrella clinical trials. The aim of our systematic review with meta-analysis was to evaluate the efficacy and safety profiles in cancer umbrella trials testing targeted drugs or a combination of targeted therapy with chemotherapy. METHODS Our study was prospectively registered in PROSPERO (CRD42020171494). We searched Embase and PubMed for cancer umbrella trials testing targeted agents or a combination of targeted therapies with chemotherapy. We included solid tumor studies published between 1 January 2006 and 7 October 2019. We measured the risk using drug-related grade 3 or higher adverse events (AEs), and the benefit by objective response rate (ORR), progression-free survival (PFS), and overall survival (OS). When possible, data were meta-analyzed. RESULTS Of the 6207 records identified, we included 31 sub-trials or arms of nine umbrella trials (N = 1637). The pooled overall ORR was 17.7% (95% confidence interval [CI] 9.5-25.9). The ORR for targeted therapies in the experimental arms was significantly lower than the ORR for a combination of targeted therapy drugs with chemotherapy: 13.3% vs 39.0%; p = 0.005. The median PFS was 2.4 months (95% CI 1.9-2.9), and the median OS was 7.1 months (95% CI 6.1-8.4). The overall drug-related death rate (drug-related grade 5 AEs rate) was 0.8% (95% CI 0.3-1.4), and the average drug-related grade 3/4 AE rate per person was 0.45 (95% CI 0.40-0.50). CONCLUSIONS Our findings suggest that, on average, one in five cancer patients in umbrella trials published between 1 January 2006 and 7 October 2019 responded to a given therapy, while one in 125 died due to drug toxicity. Our findings do not support the expectation of increased patient benefit in cancer umbrella trials. Further studies should investigate whether umbrella trial design and the precision oncology approach improve patient outcomes.
Collapse
Affiliation(s)
- Karolina Strzebonska
- Research Ethics in Medicine Study Group (REMEDY), Faculty of Health Sciences, Jagiellonian University Medical College, Kraków, Poland
| | - Mateusz Blukacz
- Institute of Psychology, University of Silesia, Katowice, Poland
- Institute of Public Health, Faculty of Health Sciences, Jagiellonian University Medical College, Kraków, Poland
| | - Mateusz T. Wasylewski
- Research Ethics in Medicine Study Group (REMEDY), Faculty of Health Sciences, Jagiellonian University Medical College, Kraków, Poland
| | - Maciej Polak
- Research Ethics in Medicine Study Group (REMEDY), Faculty of Health Sciences, Jagiellonian University Medical College, Kraków, Poland
- Department of Epidemiology and Population Studies, Institute of Public Health, Faculty of Health Sciences, Jagiellonian University Medical College, Kraków, Poland
| | - Bishal Gyawali
- Department of Oncology and the Department of Public Health Sciences, Queen’s University, Kingston, Ontario Canada
| | - Marcin Waligora
- Research Ethics in Medicine Study Group (REMEDY), Faculty of Health Sciences, Jagiellonian University Medical College, Kraków, Poland
| |
Collapse
|
31
|
Hamamoto R, Takasawa K, Machino H, Kobayashi K, Takahashi S, Bolatkan A, Shinkai N, Sakai A, Aoyama R, Yamada M, Asada K, Komatsu M, Okamoto K, Kameoka H, Kaneko S. Application of non-negative matrix factorization in oncology: one approach for establishing precision medicine. Brief Bioinform 2022; 23:6628783. [PMID: 35788277 PMCID: PMC9294421 DOI: 10.1093/bib/bbac246] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/06/2022] [Accepted: 05/25/2022] [Indexed: 12/19/2022] Open
Abstract
The increase in the expectations of artificial intelligence (AI) technology has led to machine learning technology being actively used in the medical field. Non-negative matrix factorization (NMF) is a machine learning technique used for image analysis, speech recognition, and language processing; recently, it is being applied to medical research. Precision medicine, wherein important information is extracted from large-scale medical data to provide optimal medical care for every individual, is considered important in medical policies globally, and the application of machine learning techniques to this end is being handled in several ways. NMF is also introduced differently because of the characteristics of its algorithms. In this review, the importance of NMF in the field of medicine, with a focus on the field of oncology, is described by explaining the mathematical science of NMF and the characteristics of the algorithm, providing examples of how NMF can be used to establish precision medicine, and presenting the challenges of NMF. Finally, the direction regarding the effective use of NMF in the field of oncology is also discussed.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Rina Aoyama
- Showa University Graduate School of Medicine School of Medicine
| | | | - Ken Asada
- RIKEN Center for Advanced Intelligence Project
| | | | | | | | | |
Collapse
|
32
|
Mohammad T, Singh P, Jairajpuri DS, Al-Keridis LA, Alshammari N, Adnan M, Dohare R, Hassan MI. Differential Gene Expression and Weighted Correlation Network Dynamics in High-Throughput Datasets of Prostate Cancer. Front Oncol 2022; 12:881246. [PMID: 35719950 PMCID: PMC9198298 DOI: 10.3389/fonc.2022.881246] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 05/03/2022] [Indexed: 12/13/2022] Open
Abstract
Precision oncology is an absolute need today due to the emergence of treatment resistance and heterogeneity among cancerous profiles. Target-propelled cancer therapy is one of the treasures of precision oncology which has come together with substantial medical accomplishment. Prostate cancer is one of the most common cancers in males, with tremendous biological heterogeneity in molecular and clinical behavior. The spectrum of molecular abnormalities and varying clinical patterns in prostate cancer suggest substantial heterogeneity among different profiles. To identify novel therapeutic targets and precise biomarkers implicated with prostate cancer, we performed a state-of-the-art bioinformatics study, beginning with analyzing high-throughput genomic datasets from The Cancer Genome Atlas (TCGA). Weighted gene co-expression network analysis (WGCNA) suggests a set of five dysregulated hub genes (MAF, STAT6, SOX2, FOXO1, and WNT3A) that played crucial roles in biological pathways associated with prostate cancer progression. We found overexpressed STAT6 and SOX2 and proposed them as candidate biomarkers and potential targets in prostate cancer. Furthermore, the alteration frequencies in STAT6 and SOX2 and their impact on the patients' survival were explored through the cBioPortal platform. The Kaplan-Meier survival analysis suggested that the alterations in the candidate genes were linked to the decreased overall survival of the patients. Altogether, the results signify that STAT6 and SOX2 and their genomic alterations can be explored in therapeutic interventions of prostate cancer for precision oncology, utilizing early diagnosis and target-propelled therapy.
Collapse
Affiliation(s)
- Taj Mohammad
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Prithvi Singh
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Deeba Shamim Jairajpuri
- Department of Medical Biochemistry, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | - Lamya Ahmed Al-Keridis
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Nawaf Alshammari
- Department of Biology, College of Science, University of Hail, Hail, Saudi Arabia
| | - Mohd Adnan
- Department of Biology, College of Science, University of Hail, Hail, Saudi Arabia
| | - Ravins Dohare
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
33
|
Zheng D, Yu L, Wei Z, Xia K, Guo W. N6-Methyladenosine-Related lncRNAs Are Potential Prognostic Biomarkers and Correlated With Tumor Immune Microenvironment in Osteosarcoma. Front Genet 2022; 12:805607. [PMID: 35186011 PMCID: PMC8847230 DOI: 10.3389/fgene.2021.805607] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 12/29/2021] [Indexed: 12/30/2022] Open
Abstract
N6-methyladenosine (m6A) and long non-coding RNAs (lncRNAs) play vital roles in the prognostic value and immune microenvironment of malignant tumors. Here, we constructed a m6A-related lncRNA signature in osteosarcoma samples from TCGA dataset and analyzed the association of the signature with tumor immune microenvironment. m6A-related lncRNAs were identified by performing Pearson's correlation analysis and were used to construct a novel m6A-related lncRNA signature in osteosarcoma. Validation in testing and entire cohorts confirmed the satisfactory accuracy of the risk signature. Principal-component analysis verifies the grouping ability of the risk signature. Functional enrichment analyses connected immune with the risk signature based on the six m6A-related lncRNAs. When patients were separated into high- and low-risk group based on their risk scores, we found that patients in the high-risk group had lower stromal scores, immune scores, and ESTIMATE scores, while the tumor purity was higher in the high-risk group than that in the low-risk group. As for immune cell infiltration, the proportion of monocytes was significantly higher in the low-risk group than that in the high-risk group. Of the six lncRNAs, AC004812.2 was a protective factor in osteosarcoma and low expression of AC004812.2 predicted worse overall survival. Overexpression of AC004812.2 inhibited 143B cell proliferation and increased the expression levels of IGF2BP1 and YTHDF1. In all, our m6A-related lncRNA signature was a potential prognostic biomarker and correlated with tumor immune microenvironment and immune cell infiltration, and AC004812.2 might be an important regulator of m6A modification and a promising therapeutic target in osteosarcoma.
Collapse
Affiliation(s)
- Di Zheng
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ling Yu
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhun Wei
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Kezhou Xia
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Weichun Guo
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
34
|
Templeton AR, Jeffery PL, Thomas PB, Perera MPJ, Ng G, Calabrese AR, Nicholls C, Mackenzie NJ, Wood J, Bray LJ, Vela I, Thompson EW, Williams ED. Patient-Derived Explants as a Precision Medicine Patient-Proximal Testing Platform Informing Cancer Management. Front Oncol 2022; 11:767697. [PMID: 34988013 PMCID: PMC8721047 DOI: 10.3389/fonc.2021.767697] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 11/30/2021] [Indexed: 12/12/2022] Open
Abstract
Precision medicine approaches that inform clinical management of individuals with cancer are progressively advancing. Patient-derived explants (PDEs) provide a patient-proximal ex vivo platform that can be used to assess sensitivity to standard of care (SOC) therapies and novel agents. PDEs have several advantages as a patient-proximal model compared to current preclinical models, as they maintain the phenotype and microenvironment of the individual tumor. However, the longevity of PDEs is not compatible with the timeframe required to incorporate candidate therapeutic options identified by whole exome sequencing (WES) of the patient’s tumor. This review investigates how PDE longevity varies across tumor streams and how this is influenced by tissue preparation. Improving longevity of PDEs will enable individualized therapeutics testing, and thus contribute to improving outcomes for people with cancer.
Collapse
Affiliation(s)
- Abby R Templeton
- School of Biomedical Sciences at the Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, Australia.,Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD, Australia.,Queensland Bladder Cancer Initiative (QBCI), Brisbane, QLD, Australia.,Australian Prostate Cancer Research Centre - Queensland (APCRC-Q), Brisbane, QLD, Australia
| | - Penny L Jeffery
- School of Biomedical Sciences at the Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, Australia.,Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD, Australia.,Queensland Bladder Cancer Initiative (QBCI), Brisbane, QLD, Australia.,Australian Prostate Cancer Research Centre - Queensland (APCRC-Q), Brisbane, QLD, Australia
| | - Patrick B Thomas
- School of Biomedical Sciences at the Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, Australia.,Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD, Australia.,Queensland Bladder Cancer Initiative (QBCI), Brisbane, QLD, Australia.,Australian Prostate Cancer Research Centre - Queensland (APCRC-Q), Brisbane, QLD, Australia
| | - Mahasha P J Perera
- School of Biomedical Sciences at the Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, Australia.,Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD, Australia.,Queensland Bladder Cancer Initiative (QBCI), Brisbane, QLD, Australia.,Australian Prostate Cancer Research Centre - Queensland (APCRC-Q), Brisbane, QLD, Australia.,Department of Urology, Princess Alexandra Hospital (PAH), Brisbane, QLD, Australia
| | - Gary Ng
- Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD, Australia.,Department of Medical Oncology, Princess Alexandra Hospital (PAH), Brisbane, QLD, Australia
| | - Alivia R Calabrese
- School of Biomedical Sciences at the Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, Australia.,Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD, Australia.,Queensland Bladder Cancer Initiative (QBCI), Brisbane, QLD, Australia.,Australian Prostate Cancer Research Centre - Queensland (APCRC-Q), Brisbane, QLD, Australia
| | - Clarissa Nicholls
- School of Biomedical Sciences at the Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, Australia.,Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD, Australia.,Queensland Bladder Cancer Initiative (QBCI), Brisbane, QLD, Australia
| | - Nathan J Mackenzie
- School of Biomedical Sciences at the Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, Australia.,Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD, Australia.,Queensland Bladder Cancer Initiative (QBCI), Brisbane, QLD, Australia.,Australian Prostate Cancer Research Centre - Queensland (APCRC-Q), Brisbane, QLD, Australia
| | - Jack Wood
- School of Biomedical Sciences at the Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, Australia.,Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD, Australia.,Queensland Bladder Cancer Initiative (QBCI), Brisbane, QLD, Australia
| | - Laura J Bray
- Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD, Australia.,School of Mechanical, Medical and Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD, Australia.,Australian Research Council (ARC) Training Centre for Cell and Tissue Engineering, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Ian Vela
- School of Biomedical Sciences at the Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, Australia.,Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD, Australia.,Queensland Bladder Cancer Initiative (QBCI), Brisbane, QLD, Australia.,Australian Prostate Cancer Research Centre - Queensland (APCRC-Q), Brisbane, QLD, Australia.,Department of Urology, Princess Alexandra Hospital (PAH), Brisbane, QLD, Australia
| | - Erik W Thompson
- School of Biomedical Sciences at the Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, Australia.,Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD, Australia
| | - Elizabeth D Williams
- School of Biomedical Sciences at the Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, Australia.,Centre for Personalised Analysis of Cancers (CPAC), Brisbane, QLD, Australia.,Queensland Bladder Cancer Initiative (QBCI), Brisbane, QLD, Australia.,Australian Prostate Cancer Research Centre - Queensland (APCRC-Q), Brisbane, QLD, Australia
| |
Collapse
|
35
|
Liany H, Lin Y, Jeyasekharan A, Rajan V. An Algorithm to Mine Therapeutic Motifs for Cancer from Networks of Genetic Interactions. IEEE J Biomed Health Inform 2022; 26:2830-2838. [PMID: 34990373 DOI: 10.1109/jbhi.2022.3141076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Study of pairwise genetic interactions, such as mutually exclusive mutations, has led to understanding of underlying mechanisms in cancer. Investigation of various combinatorial motifs within networks of such interactions can lead to deeper insights into its mutational landscape and inform therapy development. One such motif called the Between-Pathway Model (BPM) represents redundant or compensatory pathways that can be therapeutically exploited. Finding such BPM motifs is challenging since most formulations require solving variants of the NP-complete maximum weight bipartite subgraph problem. In this paper we design an algorithm based on Integer Linear Programming (ILP) to solve this problem. In our experiments, our approach outperforms the best previous method to mine BPM motifs. Further, our ILP-based approach allows us to easily model additional application-specific constraints. We illustrate this advantage through a new application of BPM motifs that can potentially aid in finding combination therapies to combat cancer.
Collapse
|
36
|
Luo R, Liu H, Cheng Z. Protein scaffolds: Antibody alternative for cancer diagnosis and therapy. RSC Chem Biol 2022; 3:830-847. [PMID: 35866165 PMCID: PMC9257619 DOI: 10.1039/d2cb00094f] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/23/2022] [Indexed: 12/01/2022] Open
Abstract
Although antibodies are well developed and widely used in cancer therapy and diagnostic fields, some defects remain, such as poor tissue penetration, long in vivo metabolic retention, potential cytotoxicity, patent limitation, and high production cost. These issues have led scientists to explore and develop novel antibody alternatives. Protein scaffolds are small monomeric proteins with stable tertiary structures and mutable residues, which emerged in the 1990s. By combining robust gene engineering and phage display techniques, libraries with sufficient diversity could be established for target binding scaffold selection. Given the properties of small size, high affinity, and excellent specificity and stability, protein scaffolds have been applied in basic research, and preclinical and clinical fields over the past two decades. To date, more than 20 types of protein scaffolds have been developed, with the most frequently used being affibody, adnectin, ANTICALIN®, DARPins, and knottin. In this review, we focus on the protein scaffold applications in cancer therapy and diagnosis in the last 5 years, and discuss the pros and cons, and strategies of optimization and design. Although antibodies are well developed and widely used in cancer therapy and diagnostic fields, some defects remain, such as poor tissue penetration, long in vivo metabolic retention, potential cytotoxicity, patent limitation, and high production cost.![]()
Collapse
Affiliation(s)
- Renli Luo
- Department of Molecular Medicine, College of Life and Health Sciences, Northeastern University Shenyang China
| | - Hongguang Liu
- Department of Molecular Medicine, College of Life and Health Sciences, Northeastern University Shenyang China
| | - Zhen Cheng
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
- Drug Discovery Shandong Laboratory, Bohai Rim Advanced Research Institute for Drug Discovery Yantai Shandong 264117 China
| |
Collapse
|
37
|
Son J, Fridén J, Lieber RL. Biomechanical Modeling of Brachialis-to-Wrist Extensor Muscle Transfer Function for Daily Activities in Tetraplegia. JB JS Open Access 2022; 7:JBJSOA-D-22-00018. [DOI: 10.2106/jbjs.oa.22.00018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
38
|
Wei Q, Ramsey SA. Predicting chemotherapy response using a variational autoencoder approach. BMC Bioinformatics 2021; 22:453. [PMID: 34551729 PMCID: PMC8456615 DOI: 10.1186/s12859-021-04339-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 08/17/2021] [Indexed: 01/14/2023] Open
Abstract
Background Multiple studies have shown the utility of transcriptome-wide RNA-seq profiles as features for machine learning-based prediction of response to chemotherapy in cancer. While tumor transcriptome profiles are publicly available for thousands of tumors for many cancer types, a relatively modest number of tumor profiles are clinically annotated for response to chemotherapy. The paucity of labeled examples and the high dimension of the feature data limit performance for predicting therapeutic response using fully-supervised classification methods. Recently, multiple studies have established the utility of a deep neural network approach, the variational autoencoder (VAE), for generating meaningful latent features from original data. Here, we report the first study of a semi-supervised approach using VAE-encoded tumor transcriptome features and regularized gradient boosted decision trees (XGBoost) to predict chemotherapy drug response for five cancer types: colon, pancreatic, bladder, breast, and sarcoma. Results We found: (1) VAE-encoding of the tumor transcriptome preserves the cancer type identity of the tumor, suggesting preservation of biologically relevant information; and (2) as a feature-set for supervised classification to predict response-to-chemotherapy, the unsupervised VAE encoding of the tumor’s gene expression profile leads to better area under the receiver operating characteristic curve and area under the precision-recall curve classification performance than the original gene expression profile or the PCA principal components or the ICA components of the gene expression profile, in four out of five cancer types that we tested. Conclusions Given high-dimensional “omics” data, the VAE is a powerful tool for obtaining a nonlinear low-dimensional embedding; it yields features that retain biological patterns that distinguish between different types of cancer and that enable more accurate tumor transcriptome-based prediction of response to chemotherapy than would be possible using the original data or their principal components. Supplementary Information The online version contains supplementary material available at 10.1186/s12859-021-04339-6.
Collapse
Affiliation(s)
- Qi Wei
- School of Electrical Engineering and Computer Science, Oregon State University, Corvallis, OR, USA.
| | - Stephen A Ramsey
- Department of Biomedical Sciences, Oregon State University, Corvallis, OR, USA
| |
Collapse
|
39
|
Lapuente-Santana Ó, van Genderen M, Hilbers PA, Finotello F, Eduati F. Interpretable systems biomarkers predict response to immune-checkpoint inhibitors. PATTERNS (NEW YORK, N.Y.) 2021; 2:100293. [PMID: 34430923 PMCID: PMC8369166 DOI: 10.1016/j.patter.2021.100293] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/22/2021] [Accepted: 05/31/2021] [Indexed: 02/07/2023]
Abstract
Cancer cells can leverage several cell-intrinsic and -extrinsic mechanisms to escape immune system recognition. The inherent complexity of the tumor microenvironment, with its multicellular and dynamic nature, poses great challenges for the extraction of biomarkers of immune response and immunotherapy efficacy. Here, we use RNA-sequencing (RNA-seq) data combined with different sources of prior knowledge to derive system-based signatures of the tumor microenvironment, quantifying immune-cell composition and intra- and intercellular communications. We applied multi-task learning to these signatures to predict different hallmarks of immune responses and derive cancer-type-specific models based on interpretable systems biomarkers. By applying our models to independent RNA-seq data from cancer patients treated with PD-1/PD-L1 inhibitors, we demonstrated that our method to Estimate Systems Immune Response (EaSIeR) accurately predicts therapeutic outcome. We anticipate that EaSIeR will be a valuable tool to provide a holistic description of immune responses in complex and dynamic systems such as tumors using available RNA-seq data.
Collapse
Affiliation(s)
- Óscar Lapuente-Santana
- Department of Biomedical Engineering, Eindhoven University of Technology, 5612 AZ Eindhoven, the Netherlands
| | - Maisa van Genderen
- Department of Biomedical Engineering, Eindhoven University of Technology, 5612 AZ Eindhoven, the Netherlands
| | - Peter A.J. Hilbers
- Department of Biomedical Engineering, Eindhoven University of Technology, 5612 AZ Eindhoven, the Netherlands
| | - Francesca Finotello
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Federica Eduati
- Department of Biomedical Engineering, Eindhoven University of Technology, 5612 AZ Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, 5612 AZ Eindhoven, the Netherlands
| |
Collapse
|
40
|
Reel PS, Reel S, Pearson E, Trucco E, Jefferson E. Using machine learning approaches for multi-omics data analysis: A review. Biotechnol Adv 2021; 49:107739. [PMID: 33794304 DOI: 10.1016/j.biotechadv.2021.107739] [Citation(s) in RCA: 316] [Impact Index Per Article: 79.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/01/2021] [Accepted: 03/25/2021] [Indexed: 02/06/2023]
Abstract
With the development of modern high-throughput omic measurement platforms, it has become essential for biomedical studies to undertake an integrative (combined) approach to fully utilise these data to gain insights into biological systems. Data from various omics sources such as genetics, proteomics, and metabolomics can be integrated to unravel the intricate working of systems biology using machine learning-based predictive algorithms. Machine learning methods offer novel techniques to integrate and analyse the various omics data enabling the discovery of new biomarkers. These biomarkers have the potential to help in accurate disease prediction, patient stratification and delivery of precision medicine. This review paper explores different integrative machine learning methods which have been used to provide an in-depth understanding of biological systems during normal physiological functioning and in the presence of a disease. It provides insight and recommendations for interdisciplinary professionals who envisage employing machine learning skills in multi-omics studies.
Collapse
Affiliation(s)
- Parminder S Reel
- Division of Population Health and Genomics, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Smarti Reel
- Division of Population Health and Genomics, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Ewan Pearson
- Division of Population Health and Genomics, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Emanuele Trucco
- VAMPIRE project, Computing, School of Science and Engineering, University of Dundee, Dundee, United Kingdom
| | - Emily Jefferson
- Division of Population Health and Genomics, School of Medicine, University of Dundee, Dundee, United Kingdom.
| |
Collapse
|
41
|
Wu M, Hong G, Chen Y, Ye L, Zhang K, Cai K, Yang H, Long X, Gao W, Li H. Personalized drug testing in a patient with non-small-cell lung cancer using cultured cancer cells from pleural effusion. J Int Med Res 2021; 48:300060520955058. [PMID: 32954884 PMCID: PMC7509736 DOI: 10.1177/0300060520955058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Objective Patients with non-small-cell lung cancer (NSCLC) and primary or acquired resistance do not respond to targeted drugs. We explored whether cancer cells can be cultured from liquid biopsies from patients with primary resistance to tyrosine kinase inhibitors (TKIs). We aimed to predict patients’ responses to drugs according to in vitro drug testing results. Methods Cancer cell cultures were established from the pleural effusion of a patient with TKI-resistant NSCLC using a conditional reprogramming technique. Phenotypic drug sensitivity tests were performed using the Cell Counting Kit-8 assay. We tested individual drugs and compared the synergistic and inhibitory effects of drug combinations. Results The results of our in vitro sensitivity test using the combination of cisplatin and pemetrexed were correlated with the patient’s response. Conclusion This represents the first successful report of predictive testing for combination therapy in patients with epidermal growth factor receptor-mutant NSCLC and primary TKI resistance. This strategy should be applicable to both chemotherapies and targeted therapies, and it will significantly improve the clinical treatment and management of patients with NSCLC and primary or acquired resistance to targeted therapies, as well as patients lacking targetable mutations.
Collapse
Affiliation(s)
- Ming Wu
- Wuhan University Shenzhen Institute, Shenzhen, Guangdong, China
| | - Guodai Hong
- Shenzhen Luohu People's Hospital, Shenzhen, Guangdong, China
| | - Yu Chen
- Wuhan University Shenzhen Institute, Shenzhen, Guangdong, China
| | - Lina Ye
- Wuhan University Shenzhen Institute, Shenzhen, Guangdong, China
| | - Kang Zhang
- Wuhan University Shenzhen Institute, Shenzhen, Guangdong, China
| | - Kaihong Cai
- Wuhan University Shenzhen Institute, Shenzhen, Guangdong, China
| | - Huadong Yang
- Wuhan University Shenzhen Institute, Shenzhen, Guangdong, China
| | - Xiang Long
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Wenbin Gao
- Shenzhen Luohu People's Hospital, Shenzhen, Guangdong, China
| | - Hui Li
- Wuhan University Shenzhen Institute, Shenzhen, Guangdong, China
| |
Collapse
|
42
|
Bioinformatics analysis of potential core genes for glioblastoma. Biosci Rep 2021; 40:225797. [PMID: 32667033 PMCID: PMC7385582 DOI: 10.1042/bsr20201625] [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: 05/18/2020] [Revised: 07/14/2020] [Accepted: 07/14/2020] [Indexed: 01/15/2023] Open
Abstract
Background: Glioblastoma (GBM) has a high degree of malignancy, aggressiveness and recurrence rate. However, there are limited options available for the treatment of GBM, and they often result in poor prognosis and unsatisfactory outcomes. Materials and methods: In order to identify potential core genes in GBM that may provide new therapeutic insights, we analyzed three gene chips (GSE2223, GSE4290 and GSE50161) screened from the GEO database. Differentially expressed genes (DEG) from the tissues of GBM and normal brain were screened using GEO2R. To determine the functional annotation and pathway of DEG, Gene Ontology (GO) and KEGG pathway enrichment analysis were conducted using DAVID database. Protein interactions of DEG were visualized using PPI network on Cytoscape software. Next, 10 Hub nodes were screened from the differentially expressed network using MCC algorithm on CytoHubba software and subsequently identified as Hub genes. Finally, the relationship between Hub genes and the prognosis of GBM patients was described using GEPIA2 survival analysis web tool. Results: A total of 37 up-regulated and 187 down-regulated genes were identified through microarray analysis. Amongst the 10 Hub genes selected, SV2B appeared to be the only gene associated with poor prognosis in glioblastoma based on the survival analysis. Conclusion: Our study suggests that high expression of SV2B is associated with poor prognosis in GBM patients. Whether SV2B can be used as a new therapeutic target for GBM requires further validation.
Collapse
|
43
|
Mussap M, Noto A, Piras C, Atzori L, Fanos V. Slotting metabolomics into routine precision medicine. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2021. [DOI: 10.1080/23808993.2021.1911639] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Michele Mussap
- Department of Surgical Science, University of Cagliari, Monserrato, Italy
| | - Antonio Noto
- Department of Medical Sciences and Public Health, University of Cagliari, Monserrato, Italy
| | - Cristina Piras
- Department of Surgical Science, University of Cagliari, Monserrato, Italy
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Luigi Atzori
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Vassilios Fanos
- Department of Surgical Science, University of Cagliari, Monserrato, Italy
| |
Collapse
|
44
|
Spolaor S, Scheve M, Firat M, Cazzaniga P, Besozzi D, Nobile MS. Screening for Combination Cancer Therapies With Dynamic Fuzzy Modeling and Multi-Objective Optimization. Front Genet 2021; 12:617935. [PMID: 33868363 PMCID: PMC8044361 DOI: 10.3389/fgene.2021.617935] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 03/05/2021] [Indexed: 11/13/2022] Open
Abstract
Combination therapies proved to be a valuable strategy in the fight against cancer, thanks to their increased efficacy in inducing tumor cell death and in reducing tumor growth, metastatic potential, and the risk of developing drug resistance. The identification of effective combinations of drug targets generally relies on costly and time consuming processes based on in vitro experiments. Here, we present a novel computational approach that, by integrating dynamic fuzzy modeling with multi-objective optimization, allows to efficiently identify novel combination cancer therapies, with a relevant saving in working time and costs. We tested this approach on a model of oncogenic K-ras cancer cells characterized by a marked Warburg effect. The computational approach was validated by its capability in finding out therapies already known in the literature for this type of cancer cell. More importantly, our results show that this method can suggest potential therapies consisting in a small number of molecular targets. In the model of oncogenic K-ras cancer cells, for instance, we identified combination of up to three targets, which affect different cellular pathways that are crucial for cancer proliferation and survival.
Collapse
Affiliation(s)
- Simone Spolaor
- Department of Informatics, Systems and Communication, University of Milano-Bicocca, Milan, Italy
| | - Martijn Scheve
- Department of Industrial Engineering & Innovation Sciences, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Murat Firat
- Department of Industrial Engineering & Innovation Sciences, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Paolo Cazzaniga
- Department of Human and Social Sciences, University of Bergamo, Bergamo, Italy
- SYSBIO/ISBE.IT Research Centre of Systems Biology, Milan, Italy
- Bicocca Bioinformatics Biostatistics and Bioimaging Centre (B4), Milan, Italy
| | - Daniela Besozzi
- Department of Informatics, Systems and Communication, University of Milano-Bicocca, Milan, Italy
- SYSBIO/ISBE.IT Research Centre of Systems Biology, Milan, Italy
- Bicocca Bioinformatics Biostatistics and Bioimaging Centre (B4), Milan, Italy
| | - Marco S Nobile
- Department of Industrial Engineering & Innovation Sciences, Eindhoven University of Technology, Eindhoven, Netherlands
- SYSBIO/ISBE.IT Research Centre of Systems Biology, Milan, Italy
- Bicocca Bioinformatics Biostatistics and Bioimaging Centre (B4), Milan, Italy
| |
Collapse
|
45
|
Ülgen E, Can Ö, Bilguvar K, Akyerli Boylu C, Kılıçturgay Yüksel Ş, Erşen Danyeli A, Sezerman OU, Yakıcıer MC, Pamir MN, Özduman K. Sequential filtering for clinically relevant variants as a method for clinical interpretation of whole exome sequencing findings in glioma. BMC Med Genomics 2021; 14:54. [PMID: 33622343 PMCID: PMC7903763 DOI: 10.1186/s12920-021-00904-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 02/02/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In the clinical setting, workflows for analyzing individual genomics data should be both comprehensive and convenient for clinical interpretation. In an effort for comprehensiveness and practicality, we attempted to create a clinical individual whole exome sequencing (WES) analysis workflow, allowing identification of genomic alterations and presentation of neurooncologically-relevant findings. METHODS The analysis workflow detects germline and somatic variants and presents: (1) germline variants, (2) somatic short variants, (3) tumor mutational burden (TMB), (4) microsatellite instability (MSI), (5) somatic copy number alterations (SCNA), (6) SCNA burden, (7) loss of heterozygosity, (8) genes with double-hit, (9) mutational signatures, and (10) pathway enrichment analyses. Using the workflow, 58 WES analyses from matched blood and tumor samples of 52 patients were analyzed: 47 primary and 11 recurrent diffuse gliomas. RESULTS The median mean read depths were 199.88 for tumor and 110.955 for normal samples. For germline variants, a median of 22 (14-33) variants per patient was reported. There was a median of 6 (0-590) reported somatic short variants per tumor. A median of 19 (0-94) broad SCNAs and a median of 6 (0-12) gene-level SCNAs were reported per tumor. The gene with the most frequent somatic short variants was TP53 (41.38%). The most frequent chromosome-/arm-level SCNA events were chr7 amplification, chr22q loss, and chr10 loss. TMB in primary gliomas were significantly lower than in recurrent tumors (p = 0.002). MSI incidence was low (6.9%). CONCLUSIONS We demonstrate that WES can be practically and efficiently utilized for clinical analysis of individual brain tumors. The results display that NOTATES produces clinically relevant results in a concise but exhaustive manner.
Collapse
Affiliation(s)
- Ege Ülgen
- Department of Biostatistics and Medical Informatics, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Özge Can
- Department of Medical Engineering, Faculty of Engineering, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Kaya Bilguvar
- Department of Genetics, School of Medicine, Yale University, New Haven, CT, USA
- Yale Center for Genome Analysis, West Haven, CT, USA
| | - Cemaliye Akyerli Boylu
- Department of Medical Biology, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Şirin Kılıçturgay Yüksel
- Department of Medical Biology, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Ayça Erşen Danyeli
- Department of Pathology, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - O Uğur Sezerman
- Department of Biostatistics and Medical Informatics, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - M Cengiz Yakıcıer
- Department of Molecular Biology, School of Arts and Sciences, Acibadem Mehmet Ali Aydinlar University Istanbul, Istanbul, Turkey
| | - M Necmettin Pamir
- Department of Neurosurgery, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Altunizade Mahallesi, Yurtcan Sok. No:1, Üsküdar, Istanbul, 34662, Turkey
| | - Koray Özduman
- Department of Neurosurgery, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Altunizade Mahallesi, Yurtcan Sok. No:1, Üsküdar, Istanbul, 34662, Turkey.
| |
Collapse
|
46
|
Chen X, Shen C, Wei Z, Zhang R, Wang Y, Jiang L, Chen K, Qiu S, Zhang Y, Zhang T, Chen B, Xu Y, Feng Q, Huang J, Zhong Z, Li H, Che G, Xiao K. Patient-derived non-small cell lung cancer xenograft mirrors complex tumor heterogeneity. Cancer Biol Med 2021; 18:184-198. [PMID: 33628593 PMCID: PMC7877179 DOI: 10.20892/j.issn.2095-3941.2020.0012] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 06/28/2020] [Indexed: 02/05/2023] Open
Abstract
Objective Patient-derived xenograft (PDX) models have shown great promise in preclinical and translational applications, but their consistency with primary tumors in phenotypic, genetic, and pharmacodynamic heterogeneity has not been well-studied. This study aimed to establish a PDX repository for non-small cell lung cancer (NSCLC) and to further elucidate whether it could preserve the heterogeneity within and between tumors in patients. Methods A total of 75 surgically resected NSCLC specimens were implanted into immunodeficient NOD/SCID mice. Based on the successful establishment of the NSCLC PDX model, we compared the expressions of vimentin, Ki67, EGFR, and PD-L1 proteins between cancer tissues and PDX models using hematoxylin and eosin staining and immunohistochemical staining. In addition, we detected whole gene expression profiling between primary tumors and PDX generations. We also performed whole exome sequencing (WES) analysis in 17 first generation xenografts to further assess whether PDXs retained the patient heterogeneities. Finally, paclitaxel, cisplatin, doxorubicin, atezolizumab, afatininb, and AZD4547 were used to evaluate the responses of PDX models to the standard-of-care agents. Results A large collection of serially transplantable PDX models for NSCLC were successfully developed. The histology and pathological immunohistochemistry of PDX xenografts were consistent with the patients' tumor samples. WES and RNA-seq further confirmed that PDX accurately replicated the molecular heterogeneities of primary tumors. Similar to clinical patients, PDX models responded differentially to the standard-of-care treatment, including chemo-, targeted- and immuno-therapeutics. Conclusions Our established PDX models of NSCLC faithfully reproduced the molecular, histopathological, and therapeutic characteristics, as well as the corresponding tumor heterogeneities, which provides a clinically relevant platform for drug screening, biomarker discovery, and translational research.
Collapse
Affiliation(s)
- Xuanming Chen
- National Chengdu Center for Safety Evaluation of Drugs and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610000, China
- Sichuan Kangcheng Biotechnology Co., Ltd. Chengdu 610000, China
| | - Cheng Shen
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Zhe Wei
- Sichuan Kangcheng Biotechnology Co., Ltd. Chengdu 610000, China
| | - Rui Zhang
- Sichuan Kangcheng Biotechnology Co., Ltd. Chengdu 610000, China
| | - Yongsheng Wang
- GCP Center, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Lili Jiang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Ke Chen
- Sichuan Kangcheng Biotechnology Co., Ltd. Chengdu 610000, China
| | - Shuang Qiu
- Sichuan Kangcheng Biotechnology Co., Ltd. Chengdu 610000, China
| | - Yuanli Zhang
- Sichuan Kangcheng Biotechnology Co., Ltd. Chengdu 610000, China
| | - Ting Zhang
- Sichuan Kangcheng Biotechnology Co., Ltd. Chengdu 610000, China
- Laboratory of Nonhuman Primate Disease Modeling Research, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Bin Chen
- Center for Infectious Diseases, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Yanjun Xu
- Sichuan Kangcheng Biotechnology Co., Ltd. Chengdu 610000, China
| | - Qiyi Feng
- National Chengdu Center for Safety Evaluation of Drugs and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610000, China
- Sichuan Kangcheng Biotechnology Co., Ltd. Chengdu 610000, China
| | - Jinxing Huang
- National Chengdu Center for Safety Evaluation of Drugs and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610000, China
- Sichuan Kangcheng Biotechnology Co., Ltd. Chengdu 610000, China
| | - Zhihui Zhong
- Sichuan Kangcheng Biotechnology Co., Ltd. Chengdu 610000, China
- Laboratory of Nonhuman Primate Disease Modeling Research, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Hongxia Li
- National Chengdu Center for Safety Evaluation of Drugs and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Guowei Che
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Kai Xiao
- National Chengdu Center for Safety Evaluation of Drugs and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610000, China
- Sichuan Kangcheng Biotechnology Co., Ltd. Chengdu 610000, China
| |
Collapse
|
47
|
Chen AP, Kummar S, Moore N, Rubinstein LV, Zhao Y, Williams PM, Palmisano A, Sims D, O'Sullivan Coyne G, Rosenberger CL, Simpson M, Raghav KPS, Meric-Bernstam F, Leong S, Waqar S, Foster JC, Konaté MM, Das B, Karlovich C, Lih CJ, Polley E, Simon R, Li MC, Piekarz R, Doroshow JH. Molecular Profiling-Based Assignment of Cancer Therapy (NCI-MPACT): A Randomized Multicenter Phase II Trial. JCO Precis Oncol 2021; 5:PO.20.00372. [PMID: 33928209 PMCID: PMC8078898 DOI: 10.1200/po.20.00372] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/10/2020] [Accepted: 11/24/2020] [Indexed: 12/19/2022] Open
Abstract
This trial assessed the utility of applying tumor DNA sequencing to treatment selection for patients with advanced, refractory cancer and somatic mutations in one of four signaling pathways by comparing the efficacy of four study regimens that were either matched to the patient's aberrant pathway (experimental arm) or not matched to that pathway (control arm). MATERIALS AND METHODS Adult patients with an actionable mutation of interest were randomly assigned 2:1 to receive either (1) a study regimen identified to target the aberrant pathway found in their tumor (veliparib with temozolomide or adavosertib with carboplatin [DNA repair pathway], everolimus [PI3K pathway], or trametinib [RAS/RAF/MEK pathway]), or (2) one of the same four regimens, but chosen from among those not targeting that pathway. RESULTS Among 49 patients treated in the experimental arm, the objective response rate was 2% (95% CI, 0% to 10.9%). One of 20 patients (5%) in the experimental trametinib cohort had a partial response. There were no responses in the other cohorts. Although patients and physicians were blinded to the sequencing and random assignment results, a higher pretreatment dropout rate was observed in the control arm (22%) compared with the experimental arm (6%; P = .038), suggesting that some patients may have had prior tumor mutation profiling performed that led to a lack of participation in the control arm. CONCLUSION Further investigation, better annotation of predictive biomarkers, and the development of more effective agents are necessary to inform treatment decisions in an era of precision cancer medicine. Increasing prevalence of tumor mutation profiling and preference for targeted therapy make it difficult to use a randomized phase II design to evaluate targeted therapy efficacy in an advanced disease setting.
Collapse
Affiliation(s)
- Alice P. Chen
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Shivaani Kummar
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR
| | - Nancy Moore
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | | | - Yingdong Zhao
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - P. Mickey Williams
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Alida Palmisano
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
- General Dynamics Information Technology (GDIT), Falls Church, VA
| | - David Sims
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD
| | | | | | - Mel Simpson
- Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Kanwal P. S. Raghav
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Funda Meric-Bernstam
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Saiama Waqar
- Department of Medical Oncology, Washington University School of Medicine, St Louis, MO
| | - Jared C. Foster
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Mariam M. Konaté
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Biswajit Das
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Chris Karlovich
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Chih-Jian Lih
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Eric Polley
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN
| | - Richard Simon
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Ming-Chung Li
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Richard Piekarz
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - James H. Doroshow
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
- Center for Cancer Research, National Cancer Institute, Bethesda, MD
| |
Collapse
|
48
|
Ghazarian AA, Simonds NI, Lai GY, Mechanic LE. Opportunities for Gene and Environment Research in Cancer: An Updated Review of NCI's Extramural Grant Portfolio. Cancer Epidemiol Biomarkers Prev 2020; 30:576-583. [PMID: 33323360 DOI: 10.1158/1055-9965.epi-20-1264] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/28/2020] [Accepted: 12/11/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The study of gene-environment (GxE) interactions is a research priority for the NCI. Previously, our group analyzed NCI's extramural grant portfolio from fiscal years (FY) 2007 to 2009 to determine the state of the science in GxE research. This study builds upon our previous effort and examines changes in the landscape of GxE cancer research funded by NCI. METHODS The NCI grant portfolio was examined from FY 2010 to 2018 using the iSearch application. A time-trend analysis was conducted to explore changes over the study interval. RESULTS A total of 107 grants met the search criteria and were abstracted. The most common cancer types studied were breast (19.6%) and colorectal (18.7%). Most grants focused on GxE using specific candidate genes (69.2%) compared with agnostic approaches using genome-wide (26.2%) or whole-exome/whole-genome next-generation sequencing (NGS) approaches (19.6%); some grants used more than one approach to assess genetic variation. More funded grants incorporated NGS technologies in FY 2016-2018 compared with prior FYs. Environmental exposures most commonly examined were energy balance (46.7%) and drugs/treatment (40.2%). Over the time interval, we observed a decrease in energy balance applications with a concurrent increase in drug/treatment applications. CONCLUSIONS Research in GxE interactions has continued to concentrate on common cancers, while there have been some shifts in focus of genetic and environmental exposures. Opportunities exist to study less common cancers, apply new technologies, and increase racial/ethnic diversity. IMPACT This analysis of NCI's extramural grant portfolio updates previous efforts and provides a review of NCI grant support for GxE research.
Collapse
Affiliation(s)
- Armen A Ghazarian
- Environmental Epidemiology Branch, Epidemiology and Genomics Research Program (EGRP), Division of Cancer Control and Population Sciences (DCCPS), NCI, Bethesda, Maryland
| | | | - Gabriel Y Lai
- Environmental Epidemiology Branch, Epidemiology and Genomics Research Program (EGRP), Division of Cancer Control and Population Sciences (DCCPS), NCI, Bethesda, Maryland
| | - Leah E Mechanic
- Genomic Epidemiology Branch, EGRP, DCCPS, NCI, Bethesda, Maryland.
| |
Collapse
|
49
|
Hamamoto R, Suvarna K, Yamada M, Kobayashi K, Shinkai N, Miyake M, Takahashi M, Jinnai S, Shimoyama R, Sakai A, Takasawa K, Bolatkan A, Shozu K, Dozen A, Machino H, Takahashi S, Asada K, Komatsu M, Sese J, Kaneko S. Application of Artificial Intelligence Technology in Oncology: Towards the Establishment of Precision Medicine. Cancers (Basel) 2020; 12:E3532. [PMID: 33256107 PMCID: PMC7760590 DOI: 10.3390/cancers12123532] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 11/21/2020] [Accepted: 11/24/2020] [Indexed: 02/07/2023] Open
Abstract
In recent years, advances in artificial intelligence (AI) technology have led to the rapid clinical implementation of devices with AI technology in the medical field. More than 60 AI-equipped medical devices have already been approved by the Food and Drug Administration (FDA) in the United States, and the active introduction of AI technology is considered to be an inevitable trend in the future of medicine. In the field of oncology, clinical applications of medical devices using AI technology are already underway, mainly in radiology, and AI technology is expected to be positioned as an important core technology. In particular, "precision medicine," a medical treatment that selects the most appropriate treatment for each patient based on a vast amount of medical data such as genome information, has become a worldwide trend; AI technology is expected to be utilized in the process of extracting truly useful information from a large amount of medical data and applying it to diagnosis and treatment. In this review, we would like to introduce the history of AI technology and the current state of medical AI, especially in the oncology field, as well as discuss the possibilities and challenges of AI technology in the medical field.
Collapse
Affiliation(s)
- Ryuji Hamamoto
- Division of Molecular Modification and Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (M.Y.); (K.K.); (N.S.); (M.T.); (R.S.); (A.S.); (K.T.); (A.B.); (K.S.); (A.D.); (H.M.); (S.T.); (K.A.); (M.K.); (J.S.); (S.K.)
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo 103-0027, Japan
- Department of NCC Cancer Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Kruthi Suvarna
- Indian Institute of Technology Bombay, Powai, Mumbai 400 076, India;
| | - Masayoshi Yamada
- Division of Molecular Modification and Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (M.Y.); (K.K.); (N.S.); (M.T.); (R.S.); (A.S.); (K.T.); (A.B.); (K.S.); (A.D.); (H.M.); (S.T.); (K.A.); (M.K.); (J.S.); (S.K.)
- Department of Endoscopy, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku Tokyo 104-0045, Japan
| | - Kazuma Kobayashi
- Division of Molecular Modification and Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (M.Y.); (K.K.); (N.S.); (M.T.); (R.S.); (A.S.); (K.T.); (A.B.); (K.S.); (A.D.); (H.M.); (S.T.); (K.A.); (M.K.); (J.S.); (S.K.)
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo 103-0027, Japan
- Department of NCC Cancer Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Norio Shinkai
- Division of Molecular Modification and Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (M.Y.); (K.K.); (N.S.); (M.T.); (R.S.); (A.S.); (K.T.); (A.B.); (K.S.); (A.D.); (H.M.); (S.T.); (K.A.); (M.K.); (J.S.); (S.K.)
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo 103-0027, Japan
- Department of NCC Cancer Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Mototaka Miyake
- Department of Diagnostic Radiology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan;
| | - Masamichi Takahashi
- Division of Molecular Modification and Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (M.Y.); (K.K.); (N.S.); (M.T.); (R.S.); (A.S.); (K.T.); (A.B.); (K.S.); (A.D.); (H.M.); (S.T.); (K.A.); (M.K.); (J.S.); (S.K.)
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Shunichi Jinnai
- Department of Dermatologic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan;
| | - Ryo Shimoyama
- Division of Molecular Modification and Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (M.Y.); (K.K.); (N.S.); (M.T.); (R.S.); (A.S.); (K.T.); (A.B.); (K.S.); (A.D.); (H.M.); (S.T.); (K.A.); (M.K.); (J.S.); (S.K.)
| | - Akira Sakai
- Division of Molecular Modification and Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (M.Y.); (K.K.); (N.S.); (M.T.); (R.S.); (A.S.); (K.T.); (A.B.); (K.S.); (A.D.); (H.M.); (S.T.); (K.A.); (M.K.); (J.S.); (S.K.)
- Department of NCC Cancer Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Ken Takasawa
- Division of Molecular Modification and Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (M.Y.); (K.K.); (N.S.); (M.T.); (R.S.); (A.S.); (K.T.); (A.B.); (K.S.); (A.D.); (H.M.); (S.T.); (K.A.); (M.K.); (J.S.); (S.K.)
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo 103-0027, Japan
| | - Amina Bolatkan
- Division of Molecular Modification and Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (M.Y.); (K.K.); (N.S.); (M.T.); (R.S.); (A.S.); (K.T.); (A.B.); (K.S.); (A.D.); (H.M.); (S.T.); (K.A.); (M.K.); (J.S.); (S.K.)
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo 103-0027, Japan
| | - Kanto Shozu
- Division of Molecular Modification and Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (M.Y.); (K.K.); (N.S.); (M.T.); (R.S.); (A.S.); (K.T.); (A.B.); (K.S.); (A.D.); (H.M.); (S.T.); (K.A.); (M.K.); (J.S.); (S.K.)
| | - Ai Dozen
- Division of Molecular Modification and Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (M.Y.); (K.K.); (N.S.); (M.T.); (R.S.); (A.S.); (K.T.); (A.B.); (K.S.); (A.D.); (H.M.); (S.T.); (K.A.); (M.K.); (J.S.); (S.K.)
| | - Hidenori Machino
- Division of Molecular Modification and Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (M.Y.); (K.K.); (N.S.); (M.T.); (R.S.); (A.S.); (K.T.); (A.B.); (K.S.); (A.D.); (H.M.); (S.T.); (K.A.); (M.K.); (J.S.); (S.K.)
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo 103-0027, Japan
| | - Satoshi Takahashi
- Division of Molecular Modification and Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (M.Y.); (K.K.); (N.S.); (M.T.); (R.S.); (A.S.); (K.T.); (A.B.); (K.S.); (A.D.); (H.M.); (S.T.); (K.A.); (M.K.); (J.S.); (S.K.)
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo 103-0027, Japan
| | - Ken Asada
- Division of Molecular Modification and Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (M.Y.); (K.K.); (N.S.); (M.T.); (R.S.); (A.S.); (K.T.); (A.B.); (K.S.); (A.D.); (H.M.); (S.T.); (K.A.); (M.K.); (J.S.); (S.K.)
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo 103-0027, Japan
| | - Masaaki Komatsu
- Division of Molecular Modification and Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (M.Y.); (K.K.); (N.S.); (M.T.); (R.S.); (A.S.); (K.T.); (A.B.); (K.S.); (A.D.); (H.M.); (S.T.); (K.A.); (M.K.); (J.S.); (S.K.)
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo 103-0027, Japan
| | - Jun Sese
- Division of Molecular Modification and Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (M.Y.); (K.K.); (N.S.); (M.T.); (R.S.); (A.S.); (K.T.); (A.B.); (K.S.); (A.D.); (H.M.); (S.T.); (K.A.); (M.K.); (J.S.); (S.K.)
- Humanome Lab, 2-4-10 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Syuzo Kaneko
- Division of Molecular Modification and Cancer Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (M.Y.); (K.K.); (N.S.); (M.T.); (R.S.); (A.S.); (K.T.); (A.B.); (K.S.); (A.D.); (H.M.); (S.T.); (K.A.); (M.K.); (J.S.); (S.K.)
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo 103-0027, Japan
| |
Collapse
|
50
|
Virumbrales-Muñoz M, Chen J, Ayuso J, Lee M, Abel EJ, Beebe DJ. Organotypic primary blood vessel models of clear cell renal cell carcinoma for single-patient clinical trials. LAB ON A CHIP 2020; 20:4420-4432. [PMID: 33103699 PMCID: PMC8743028 DOI: 10.1039/d0lc00252f] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Clear cell renal cell carcinoma (ccRCC) is a common genitourinary cancer associated with the development of abnormal tumor angiogenesis. Although multiple anti-angiogenic therapies have been developed, responses to individual treatment are highly variable between patients. Thus, the use of one-patient clinical trials has been suggested as an alternative to standard trials. We used a microfluidic device to generate organotypic primary patient-specific blood vessel models using normal (NEnC) and tumor-associated primary CD31+ selected cells (TEnC). Our model was able to recapitulate differences in angiogenic sprouting and vessel permeability that characterize normal and tumor-associated vessels. We analyzed the expression profile of vessel models to define vascular normalization in a patient-specific manner. Using this data, we identified actionable targets to normalize TEnC vessel function to a more NEnC-like phenotype. Finally, we tested two of these drugs in our patient-specific models to determine the efficiency in restoring vessel function showing the potential of the model for single-patient clinical trials.
Collapse
Affiliation(s)
- María Virumbrales-Muñoz
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, 1111 Highland Avenue, Madison, Wisconsin 53705, USA.
| | | | | | | | | | | |
Collapse
|