1
|
Desponds E, Croci D, Wosika V, Hadadi N, Fonseca Costa SS, Ciarloni L, Ongaro M, Zdimerova H, Leblond MM, Hosseinian Ehrensberger S, Romero P, Verdeil G. Immuno-Transcriptomic Profiling of Blood and Tumor Tissue Identifies Gene Signatures Associated with Immunotherapy Response in Metastatic Bladder Cancer. Cancers (Basel) 2024; 16:433. [PMID: 38275874 PMCID: PMC10814931 DOI: 10.3390/cancers16020433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/11/2024] [Accepted: 01/15/2024] [Indexed: 01/27/2024] Open
Abstract
Blood-based biomarkers represent ideal candidates for the development of non-invasive immuno-oncology-based assays. However, to date, no blood biomarker has been validated to predict clinical responses to immunotherapy. In this study, we used next-generation sequencing (RNAseq) on bulk RNA extracted from whole blood and tumor samples in a pre-clinical MIBC mouse model. We aimed to identify biomarkers associated with immunotherapy response and assess the potential application of simple non-invasive blood biomarkers as a therapeutic decision-making assay compared to tissue-based biomarkers. We established that circulating immune cells and the tumor microenvironment (TME) display highly organ-specific transcriptional responses to ICIs. Interestingly, in both, a common lymphocytic activation signature can be identified associated with the efficient response to immunotherapy, including a blood-specific CD8+ T cell activation/proliferation signature which predicts the immunotherapy response.
Collapse
Affiliation(s)
- Emma Desponds
- Department of Oncology UNIL CHUV, University of Lausanne, 1015 Lausanne, Switzerland; (E.D.); (M.O.); (H.Z.); (M.M.L.)
- Ludwig Institute for Cancer Research, University of Lausanne, 1015 Lausanne, Switzerland
| | - Davide Croci
- Novigenix SA, 1066 Epalinges, Switzerland; (D.C.); (N.H.); (S.S.F.C.); (L.C.); (S.H.E.); (P.R.)
| | - Victoria Wosika
- Novigenix SA, 1066 Epalinges, Switzerland; (D.C.); (N.H.); (S.S.F.C.); (L.C.); (S.H.E.); (P.R.)
| | - Noushin Hadadi
- Novigenix SA, 1066 Epalinges, Switzerland; (D.C.); (N.H.); (S.S.F.C.); (L.C.); (S.H.E.); (P.R.)
| | - Sara S. Fonseca Costa
- Novigenix SA, 1066 Epalinges, Switzerland; (D.C.); (N.H.); (S.S.F.C.); (L.C.); (S.H.E.); (P.R.)
| | - Laura Ciarloni
- Novigenix SA, 1066 Epalinges, Switzerland; (D.C.); (N.H.); (S.S.F.C.); (L.C.); (S.H.E.); (P.R.)
| | - Marco Ongaro
- Department of Oncology UNIL CHUV, University of Lausanne, 1015 Lausanne, Switzerland; (E.D.); (M.O.); (H.Z.); (M.M.L.)
- Ludwig Institute for Cancer Research, University of Lausanne, 1015 Lausanne, Switzerland
| | - Hana Zdimerova
- Department of Oncology UNIL CHUV, University of Lausanne, 1015 Lausanne, Switzerland; (E.D.); (M.O.); (H.Z.); (M.M.L.)
- Ludwig Institute for Cancer Research, University of Lausanne, 1015 Lausanne, Switzerland
| | - Marine M. Leblond
- Department of Oncology UNIL CHUV, University of Lausanne, 1015 Lausanne, Switzerland; (E.D.); (M.O.); (H.Z.); (M.M.L.)
- Ludwig Institute for Cancer Research, University of Lausanne, 1015 Lausanne, Switzerland
| | | | - Pedro Romero
- Novigenix SA, 1066 Epalinges, Switzerland; (D.C.); (N.H.); (S.S.F.C.); (L.C.); (S.H.E.); (P.R.)
| | - Grégory Verdeil
- Department of Oncology UNIL CHUV, University of Lausanne, 1015 Lausanne, Switzerland; (E.D.); (M.O.); (H.Z.); (M.M.L.)
- Ludwig Institute for Cancer Research, University of Lausanne, 1015 Lausanne, Switzerland
| |
Collapse
|
2
|
Hou JY, Li N, Wang J, Gao LJ, Chang JS, Cao JM. Histone crotonylation of peripheral blood mononuclear cells is a potential biomarker for diagnosis of colorectal cancer. Epigenetics Chromatin 2023; 16:35. [PMID: 37749610 PMCID: PMC10521402 DOI: 10.1186/s13072-023-00509-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 09/17/2023] [Indexed: 09/27/2023] Open
Abstract
BACKGROUND Blood-based tests have public appeal in screening cancers due to their minimally invasive nature, ability to integrate with other routine blood tests, and high compliance. This study aimed to investigate whether certain epigenetic modulation of peripheral blood mononuclear cells (PBMCs) could be a biomarker of colorectal cancer (CRC). RESULTS Western blotting of histones in the PBMCs from 40 colorectal cancer patients and 40 healthy controls was performed to identify the crotonylation sites of proteins. The correlation of crotonylation with tumor staging and diagnostic efficacy were analyzed. Crotonylation of H2BK12 (H2BK12cr) was identified significantly upregulated in the PBMCs of CRC patients compared to healthy controls, and were closely related to distant metastasis (P = 0.0478) and late TNM stage (P = 0.0201). Receiver operator characteristic curve (ROC) analysis demonstrated that the area under curve (AUC) of H2BK12cr was 0.8488, the sensitivity was 70%, and the specificity was 92.5%. The H2BK12cr parameter significantly increased the diagnostic effectiveness of CRC compared with the commercial carcinoembryonic antigen assays. CONCLUSIONS The H2BK12cr level in PBMCs of CRC patients has a potential to be a biomarker for distinguishing CRC patients from healthy controls with the advantages of easy operation and high diagnostic efficacy.
Collapse
Affiliation(s)
- Jia-Yi Hou
- Department of Clinical Laboratory, Shanxi Provincial Academy of Traditional Chinese Medicine, Taiyuan, China
| | - Ning Li
- Department of Gastrointestinal and Pancreatic Surgery and Hernia and Abdominal Surgery, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Jie Wang
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Key Laboratory of Cellular Physiology of Shanxi Province, and the Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Li-Juan Gao
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Key Laboratory of Cellular Physiology of Shanxi Province, and the Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Jia-Song Chang
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Key Laboratory of Cellular Physiology of Shanxi Province, and the Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Ji-Min Cao
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Key Laboratory of Cellular Physiology of Shanxi Province, and the Department of Physiology, Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
3
|
Goyal R, Wassie MM, Winter JM, Lathlean TJ, Young GP, Symonds EL. Progress in the field of noninvasive diagnostics for colorectal cancer: a systematic review for the accuracy of blood-based biomarkers for detection of advanced pre-cancerous lesions. Expert Rev Mol Diagn 2023; 23:1233-1250. [PMID: 38044883 DOI: 10.1080/14737159.2023.2290646] [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: 05/23/2023] [Accepted: 11/22/2023] [Indexed: 12/05/2023]
Abstract
BACKGROUND Early detection of pre-cancerous adenomas through screening can reduce colorectal cancer (CRC) incidence. Fecal immunochemical tests are commonly used, but have limited sensitivity for pre-cancerous lesions. Blood-based screening may improve test sensitivity. This systematic review and meta-analysis was conducted to evaluate the accuracy of blood-based biomarkers for detection of advanced pre-cancerous lesions. RESEARCH DESIGN AND METHODS We present the accuracy of blood-based biomarkers for the detection of advanced pre-cancerous lesions. EMBASE, Web of Science and PubMed databases were searched, with study populations limited to adults diagnosed with advanced pre-cancerous lesions at colonoscopy, who had a blood-based biomarker test analyzed with reports of sensitivity and specificity. RESULTS 69 studies were identified, which assessed 133 unique biomarkers sets. The best performing test was a panel of 6 miRNAs, with a sensitivity of 95% and specificity of 90% for advanced pre-cancerous lesions. Only 6 biomarkers demonstrated sensitivity ≥ 50% and specificity ≥ 90% for the detection of advanced pre-cancerous lesions. CONCLUSION Many different blood-based biomarkers have been assessed for detection of advanced pre-cancerous lesions, but few have progressed beyond the discovery stage. While some biomarkers have reported high sensitivity and specificity, larger prospective studies in unbiased intended-use screening populations are required for validation.
Collapse
Affiliation(s)
- Rishabh Goyal
- Department of Medicine, College of Medicine and Public Health, Flinders University, Bedford Park, Australia
| | - Molla M Wassie
- Cancer Research, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, Australia
| | - Jean M Winter
- Cancer Research, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, Australia
| | - Timothy Jh Lathlean
- Cancer Research, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, Australia
- ROSA Research Centre, South Australian Health and Medical Research Institue, Adelaide, Australia
| | - Graeme P Young
- Cancer Research, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, Australia
| | - Erin L Symonds
- Cancer Research, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, Australia
- Gastroenterology Department, Flinders Medical Centre, Bedford Park, Australia
| |
Collapse
|
4
|
Huang CS, Terng HJ, Hwang YT. Gene-Function-Based Clusters Explore Intricate Networks of Gene Expression of Circulating Tumor Cells in Patients with Colorectal Cancer. Biomedicines 2023; 11:biomedicines11010145. [PMID: 36672653 PMCID: PMC9855519 DOI: 10.3390/biomedicines11010145] [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: 11/19/2022] [Revised: 12/22/2022] [Accepted: 01/02/2023] [Indexed: 01/11/2023] Open
Abstract
Colorectal cancer (CRC) is a complex disease characterized by dynamically deregulated gene expression and crosstalk between signaling pathways. In this study, a new approach based on gene-function-based clusters was introduced to explore the CRC-associated networks of gene expression. Each cluster contained genes involved in coordinated regulatory activity, such as RAS signaling, the cell cycle process, transcription, or translation. A retrospective case-control study was conducted with the inclusion of 119 patients with histologically confirmed colorectal cancer and 308 controls. The quantitative expression data of 15 genes were obtained from the peripheral blood samples of all participants to investigate cluster-gene and gene-gene interactions. DUSP6, MDM2, and EIF2S3 were consistently selected as CRC-associated factors with high significance in all logistic models. CPEB4 became an insignificant factor only when combined with the clusters for cell cycle processes and for transcription. The CPEB4/DUSP6 complex was a prerequisite for the significance of MMD, whereas EXT2, RNF4, ZNF264, WEE1, and MCM4 were affected by more than two clusters. Intricate networks among MMD, RAS signaling factors (DUSP6, GRB2, and NF1), and translation factors (EIF2S3, CPEB4, and EXT2) were also revealed. Our results suggest that limited G1/S transition, uncontrolled DNA replication, and the cap-independent initiation of translation may be dominant and concurrent scenarios in circulating tumor cells derived from colorectal cancer. This gene-function-based cluster approach is simple and useful for revealing intricate CRC-associated gene expression networks. These findings may provide clues to the metastatic mechanisms of circulating tumor cells in patients with colorectal cancer.
Collapse
Affiliation(s)
- Chi-Shuan Huang
- Division of Colorectal Surgery, Cheng Hsin General Hospital, Taipei 11220, Taiwan
| | | | - Yi-Ting Hwang
- Department of Statistics, National Taipei University, New Taipei City 22102, Taiwan
- Correspondence:
| |
Collapse
|
5
|
Sattar RSA, Verma R, Nimisha, Kumar A, Dar GM, Apurva, Sharma AK, Kumari I, Ahmad E, Ali A, Mahajan B, Saluja SS. Diagnostic and prognostic biomarkers in colorectal cancer and the potential role of exosomes in drug delivery. Cell Signal 2022; 99:110413. [PMID: 35907519 DOI: 10.1016/j.cellsig.2022.110413] [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: 04/14/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 11/03/2022]
Abstract
Colorectal cancer (CRC) is third most common cancer with second most common cause of death worldwide. One fourth to one fifth of the CRC cases are detected at advance stage. Early detection of colorectal cancer might help in decreasing mortality and morbidity worldwide. CRC being a heterogeneous disease, new non-invasive approaches are needed to complement and improve the screening and management of CRC. Reliable and early detectable biomarkers would improve diagnosis, prognosis, therapeutic responses, and will enable the prediction of drug response and recurrence risk. Over the past decades molecular research has demonstrated the potentials of CTCs, ctDNAs, circulating mRNA, ncRNAs, and exosomes as tumor biomarkers. Non-invasive screening approaches using fecal samples for identification of altered gut microbes in CRC is also gaining attention. Exosomes can be potential candidates that can be employed in the drug delivery system. Further, the integration of in vitro, in vivo and in silico models that involve CRC biomarkers will help to understand the interactions occurring at the cellular level. This review summarizes recent update on CRC biomarkers and their application along with the nanoparticles followed by the application of organoid culture in CRC.
Collapse
Affiliation(s)
- Real Sumayya Abdul Sattar
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Renu Verma
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Nimisha
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Arun Kumar
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Ghulam Mehdi Dar
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Apurva
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Abhay Kumar Sharma
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Indu Kumari
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Ejaj Ahmad
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Asgar Ali
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Bhawna Mahajan
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India; Department of Biochemistry, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Sundeep Singh Saluja
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India; Department of GI Surgery, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India.
| |
Collapse
|
6
|
Abstract
INTRODUCTION Colorectal cancer (CRC) is the second leading cause of cancer-related deaths globally. Nonetheless, with early detection of CRC or its precancerous lesions, mortality, and CRC incidence can be reduced. Although colonoscopy is currently the gold standard for CRC screening and diagnosis, its invasive nature, and troublesome bowel preparation deter patient participation. Therefore, there is a need to expand the use of noninvasive or minimally invasive methods to increase patient compliance. AREAS COVERED This review summarizes advances in different methods for CRC screening, including stool bacterial and metagenomic markers, fecal proteins, genetic and epigenetic markers in blood and stools, and imaging modalities. The cost-effectiveness of these methods is also discussed. FIT is more cost-effective compared to virtual colonoscopy, mSEPT9 test, and Multitarget Stool DNA test, while the cost-effectiveness of other noninvasive methods requires further evaluation. EXPERT OPINION Recent evidence has well demonstrated the usefulness of gut microbiome and certain fecal bacterial markers in the noninvasive diagnosis of CRC and its precancerous lesions. Many of the fecal biomarkers, from host cells or the gut environment, show better diagnostic sensitivity than FIT. New screening tests based on these fecal biomarkers can be expected to replace FIT with higher cost-effectiveness in the near future.
Collapse
Affiliation(s)
- Sarah Cheuk Hei Chan
- Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Jessie Qiaoyi Liang
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, Cuhk Shenzhen Research Institute, the Chinese University of Hong Kong, Shatin, Hong Kong, China
| |
Collapse
|
7
|
Čelešnik H, Potočnik U. Peripheral Blood Transcriptome in Breast Cancer Patients as a Source of Less Invasive Immune Biomarkers for Personalized Medicine, and Implications for Triple Negative Breast Cancer. Cancers (Basel) 2022; 14:591. [PMID: 35158858 PMCID: PMC8833511 DOI: 10.3390/cancers14030591] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 01/14/2022] [Accepted: 01/19/2022] [Indexed: 12/21/2022] Open
Abstract
Transcriptome studies of peripheral blood cells can advance our understanding of the systemic immune response to the presence of cancer and the mechanisms underlying cancer onset and progression. This enables the identification of novel minimally invasive immune biomarkers for early cancer detection and personalized cancer management and may bring forward new immunotherapy options. Recent blood gene expression analyses in breast cancer (BC) identified distinct patient subtypes that differed in the immune reaction to cancer and were distinct from the clinical BC subtypes, which are categorized based on expression of specific receptors on tumor cells. Introducing new BC subtypes based on peripheral blood gene expression profiles may be appropriate, since it may assist in BC prognosis, the identification of patients likely to benefit from immunotherapy, and treatment efficacy monitoring. Triple-negative breast cancer (TNBC) is an aggressive, heterogeneous, and difficult-to-treat disease, and identification of novel biomarkers for this BC is crucial for clinical decision-making. A few studies have reported TNBC-enriched blood transcriptional signatures, mostly related to strong inflammation and augmentation of altered immune signaling, that can differentiate TNBC from other classical BC subtypes and facilitate diagnosis. Future research is geared toward transitioning from expression signatures in unfractionated blood cells to those in immune cell subpopulations.
Collapse
Affiliation(s)
- Helena Čelešnik
- Faculty of Chemistry and Chemical Engineering, University of Maribor, Smetanova Ulica 17, 2000 Maribor, Slovenia;
- Center for Human Genetics & Pharmacogenomics, Faculty of Medicine, University of Maribor, Taborska Ulica 8, 2000 Maribor, Slovenia
| | - Uroš Potočnik
- Faculty of Chemistry and Chemical Engineering, University of Maribor, Smetanova Ulica 17, 2000 Maribor, Slovenia;
- Center for Human Genetics & Pharmacogenomics, Faculty of Medicine, University of Maribor, Taborska Ulica 8, 2000 Maribor, Slovenia
- Department for Science and Research, University Medical Centre Maribor, Ljubljanska Ulica 5, 2000 Maribor, Slovenia
| |
Collapse
|
8
|
Wen G, Zhou T, Gu W. The potential of using blood circular RNA as liquid biopsy biomarker for human diseases. Protein Cell 2021; 12:911-946. [PMID: 33131025 PMCID: PMC8674396 DOI: 10.1007/s13238-020-00799-3] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 10/09/2020] [Indexed: 12/14/2022] Open
Abstract
Circular RNA (circRNA) is a novel class of single-stranded RNAs with a closed loop structure. The majority of circRNAs are formed by a back-splicing process in pre-mRNA splicing. Their expression is dynamically regulated and shows spatiotemporal patterns among cell types, tissues and developmental stages. CircRNAs have important biological functions in many physiological processes, and their aberrant expression is implicated in many human diseases. Due to their high stability, circRNAs are becoming promising biomarkers in many human diseases, such as cardiovascular diseases, autoimmune diseases and human cancers. In this review, we focus on the translational potential of using human blood circRNAs as liquid biopsy biomarkers for human diseases. We highlight their abundant expression, essential biological functions and significant correlations to human diseases in various components of peripheral blood, including whole blood, blood cells and extracellular vesicles. In addition, we summarize the current knowledge of blood circRNA biomarkers for disease diagnosis or prognosis.
Collapse
Affiliation(s)
- Guoxia Wen
- State Key Laboratory of Bioelectronics, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Tong Zhou
- Department of Physiology and Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV, 89557, USA.
| | - Wanjun Gu
- State Key Laboratory of Bioelectronics, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, 210096, China.
| |
Collapse
|
9
|
Ferlizza E, Solmi R, Sgarzi M, Ricciardiello L, Lauriola M. The Roadmap of Colorectal Cancer Screening. Cancers (Basel) 2021; 13:1101. [PMID: 33806465 PMCID: PMC7961708 DOI: 10.3390/cancers13051101] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 02/24/2021] [Accepted: 02/27/2021] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is the third most common form of cancer in terms of incidence and the second in terms of mortality worldwide. CRC develops over several years, thus highlighting the importance of early diagnosis. National screening programs based on fecal occult blood tests and subsequent colonoscopy have reduced the incidence and mortality, however improvements are needed since the participation rate remains low and the tests present a high number of false positive results. This review provides an overview of the CRC screening globally and the state of the art in approaches aimed at improving accuracy and participation in CRC screening, also considering the need for gender and age differentiation. New fecal tests and biomarkers such as DNA methylation, mutation or integrity, proteins and microRNAs are explored, including recent investigations into fecal microbiota. Liquid biopsy approaches, involving novel biomarkers and panels, such as circulating mRNA, micro- and long-non-coding RNA, DNA, proteins and extracellular vesicles are discussed. The approaches reported are based on quantitative PCR methods that could be easily applied to routine screening, or arrays and sequencing assays that should be better exploited to describe and identify candidate biomarkers in blood samples.
Collapse
Affiliation(s)
- Enea Ferlizza
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40138 Bologna, Italy; (R.S.); (M.S); (M.L.)
| | - Rossella Solmi
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40138 Bologna, Italy; (R.S.); (M.S); (M.L.)
| | - Michela Sgarzi
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40138 Bologna, Italy; (R.S.); (M.S); (M.L.)
| | - Luigi Ricciardiello
- Gastroenterology Unit, Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy;
| | - Mattia Lauriola
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40138 Bologna, Italy; (R.S.); (M.S); (M.L.)
| |
Collapse
|
10
|
Gan X, Wang T, Chen ZY, Zhang KH. Blood-derived molecular signatures as biomarker panels for the early detection of colorectal cancer. Mol Biol Rep 2020; 47:8159-8168. [DOI: 10.1007/s11033-020-05838-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 09/10/2020] [Indexed: 12/24/2022]
|
11
|
Transcriptomic Analyses Revealed Systemic Alterations in Gene Expression in Circulation and Tumor Microenvironment of Colorectal Cancer Patients. Cancers (Basel) 2019; 11:cancers11121994. [PMID: 31835892 PMCID: PMC6966620 DOI: 10.3390/cancers11121994] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 12/03/2019] [Accepted: 12/05/2019] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is among the leading causes of cancer-related deaths worldwide, underscoring a need for better understanding of the disease and development of novel diagnostic biomarkers and therapeutic interventions. Herein, we performed transcriptome analyses on peripheral blood mononuclear cells (PBMCs), CRC tumor tissue and adjacent normal tissue from 10 CRC patients and PBMCs from 15 healthy controls. Up regulated transcripts from CRC PBMCs were associated with functions related to immune cell trafficking and cellular movement, while downregulated transcripts were enriched in cellular processes related to cell death. Most affected signaling networks were those involved in tumor necrosis factor (TNF) and interleukin signaling. The expression of selected immune-related genes from the RNA-Seq data were further validated using qRT-PCR. Transcriptome analysis of CRC tumors and ingenuity pathway analysis revealed enrichment in several functional categories related to cellular movement, cell growth and proliferation, DNA replication, recombination and repair, while functional categories related to cell death were suppressed. Upstream regulator analysis revealed activation of ERBB2 and FOXM1 networks. Interestingly, there were 18 common upregulated and 36 common downregulated genes when comparing PBMCs and tumor tissue, suggesting transcriptomic changes in the tumor microenvironment could be reflected, in part, in the periphery with potential utilization as disease biomarkers.
Collapse
|
12
|
Chen S, Liu Y, Wang Y, Xue Z. LncRNA CCAT1 Promotes Colorectal Cancer Tumorigenesis Via A miR-181b-5p/TUSC3 Axis. Onco Targets Ther 2019; 12:9215-9225. [PMID: 31807005 PMCID: PMC6842281 DOI: 10.2147/ott.s216718] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 09/12/2019] [Indexed: 01/17/2023] Open
Abstract
AIM The aim was to determine the function and molecular mechanism of long non-coding RNA colon cancer associated transcript-1(lncRNA CCAT1) in the development of colorectal cancer (CRC). METHODS CCAT1 mRNA expression levels were determined in CRC tissues and cells using reverse transcription-quantitative polymerase chain reaction. Cell Counting Kit-8 and colony formation assays were used to examine the effects of CCAT1 on the proliferation of CRC cells. Luciferase reporter gene analysis was used to confirm the target gene of microRNA-181b-5p (miR-181b-5p) in CRC cells. Tumor xenografts were subsequently used to investigate the role of CCAT1 in CRC growth in vivo. RESULTS The relative mRNA expression levels of CCAT1 were significantly higher in CRC tissues and cell lines compared with the normal tissues or cells. CCAT1 knockdown significantly inhibited CRC cell proliferation in vitro and in vivo. Bioinformatics and luciferase reporter assays showed that miR-181b-5p was a direct target of CCAT1, and the expression of miR-181b-5p was negatively correlated with the expression of CCAT1 in CRC tissues. Furthermore, CCAT1 positively regulated the level of tumor suppressor candidate 3 (TUSC3) by competing with miR-181b-5p in CRC cells. CONCLUSION These data suggested that lncRNA CCAT1 promoted colorectal cancer tumorigenesis via a miR-181b-5p/TUSC3 axis.
Collapse
Affiliation(s)
- Si Chen
- Department of Colorectal and Anal Surgery
| | | | | | - Zhaoping Xue
- Anaesthesia Wake Room, First Hospital of Jilin University, Changchun, Jilin130021, People’s Republic of China
| |
Collapse
|
13
|
Jeun M, Lee HJ, Park S, Do E, Choi J, Sung Y, Hong S, Kim S, Kim D, Kang JY, Son H, Joo J, Song EM, Hwang SW, Park SH, Yang D, Ye BD, Byeon J, Choe J, Yang S, Moinova H, Markowitz SD, Lee KH, Myung S. A Novel Blood-Based Colorectal Cancer Diagnostic Technology Using Electrical Detection of Colon Cancer Secreted Protein-2. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1802115. [PMID: 31179210 PMCID: PMC6548955 DOI: 10.1002/advs.201802115] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 03/02/2019] [Indexed: 05/15/2023]
Abstract
Colorectal cancer (CRC) is the second-leading cause of cancer-related mortality worldwide, which may be effectively reduced by early screening. Colon cancer secreted protein-2 (CCSP-2) is a promising blood marker for CRC. An electric-field effect colorectal sensor (E-FECS), an ion-sensitive field-effect transistor under dual gate operation with nanostructure is developed, to quantify CCSP-2 directly from patient blood samples. The sensing performance of the E-FECS is verified in 7 controls and 7 CRC samples, and it is clinically validated on 30 controls, 30 advanced adenomas, and 81 CRC cases. The concentration of CCSP-2 is significantly higher in plasma samples from CRC and advanced adenoma compared with controls (both P < 0.001). Sensitivity and specificity for CRC versus controls are 44.4% and 86.7%, respectively (AUC of 0.67), and 43.3% and 86.7%, respectively, for advanced adenomas (AUC of 0.67). CCSP-2 detects a greater number of CRC cases than carcinoembryonic antigen does (45.6% vs 24.1%), and the combination of the two markers detects an even greater number of cases (53.2%). The E-FECS system successfully detects CCSP-2 in a wide range of samples including early stage cancers and advanced adenoma. CCSP-2 has potential for use as a blood-based biomarker for CRC.
Collapse
Affiliation(s)
- Minhong Jeun
- Center for BiomaterialsBiomedical Research InstituteKorea Institute of Science and Technology (KIST)5 Hwarangno 14‐gilSeongbuk‐guSeoul02792Republic of Korea
| | - Hyo Jeong Lee
- Health Screening & Promotion CenterAsan Medical Center88 Olympic‐ro 43‐gilSongpa‐guSeoul05505Republic of Korea
| | - Sungwook Park
- Center for BiomaterialsBiomedical Research InstituteKorea Institute of Science and Technology (KIST)5 Hwarangno 14‐gilSeongbuk‐guSeoul02792Republic of Korea
- Division of Bio‐Medical Science & TechnologyKIST School – Korea University of Science and Technology (UST)5 Hwarangno 14‐gilSeongbuk‐guSeoul02792Republic of Korea
| | - Eun‐ju Do
- Asan Institute for Life SciencesAsan Medical Center88 Olympic‐ro 43‐gilSongpa‐guSeoul05505Republic of Korea
| | - Jaewon Choi
- Center for BiomaterialsBiomedical Research InstituteKorea Institute of Science and Technology (KIST)5 Hwarangno 14‐gilSeongbuk‐guSeoul02792Republic of Korea
| | - You‐Na Sung
- Department of PathologyAsan Medical CenterUniversity of Ulsan College of Medicine88 Olympic‐ro 43‐gilSongpa‐guSeoul05505Republic of Korea
| | - Seung‐Mo Hong
- Department of PathologyAsan Medical CenterUniversity of Ulsan College of Medicine88 Olympic‐ro 43‐gilSongpa‐guSeoul05505Republic of Korea
| | - Sang‐Yeob Kim
- Asan Institute for Life SciencesAsan Medical Center88 Olympic‐ro 43‐gilSongpa‐guSeoul05505Republic of Korea
- Department of Convergence MedicineUniversity of Ulsan College of Medicine88 Olympic‐ro 43‐gilSongpa‐guSeoul05505Republic of Korea
| | - Dong‐Hee Kim
- Asan Institute for Life SciencesAsan Medical Center88 Olympic‐ro 43‐gilSongpa‐guSeoul05505Republic of Korea
| | - Ja Young Kang
- Asan Institute for Life SciencesAsan Medical Center88 Olympic‐ro 43‐gilSongpa‐guSeoul05505Republic of Korea
| | - Hye‐Nam Son
- Asan Institute for Life SciencesAsan Medical Center88 Olympic‐ro 43‐gilSongpa‐guSeoul05505Republic of Korea
| | - Jinmyoung Joo
- Asan Institute for Life SciencesAsan Medical Center88 Olympic‐ro 43‐gilSongpa‐guSeoul05505Republic of Korea
- Department of Convergence MedicineUniversity of Ulsan College of Medicine88 Olympic‐ro 43‐gilSongpa‐guSeoul05505Republic of Korea
| | - Eun Mi Song
- Department of GastroenterologyAsan Medical CenterUniversity of Ulsan College of Medicine88 Olympic‐ro 43‐gilSongpa‐guSeoul05505Republic of Korea
| | - Sung Wook Hwang
- Department of GastroenterologyAsan Medical CenterUniversity of Ulsan College of Medicine88 Olympic‐ro 43‐gilSongpa‐guSeoul05505Republic of Korea
| | - Sang Hyoung Park
- Department of GastroenterologyAsan Medical CenterUniversity of Ulsan College of Medicine88 Olympic‐ro 43‐gilSongpa‐guSeoul05505Republic of Korea
| | - Dong‐Hoon Yang
- Department of GastroenterologyAsan Medical CenterUniversity of Ulsan College of Medicine88 Olympic‐ro 43‐gilSongpa‐guSeoul05505Republic of Korea
| | - Byong Duk Ye
- Department of GastroenterologyAsan Medical CenterUniversity of Ulsan College of Medicine88 Olympic‐ro 43‐gilSongpa‐guSeoul05505Republic of Korea
| | - Jeong‐Sik Byeon
- Department of GastroenterologyAsan Medical CenterUniversity of Ulsan College of Medicine88 Olympic‐ro 43‐gilSongpa‐guSeoul05505Republic of Korea
| | - Jaewon Choe
- Health Screening & Promotion CenterAsan Medical Center88 Olympic‐ro 43‐gilSongpa‐guSeoul05505Republic of Korea
- Department of GastroenterologyAsan Medical CenterUniversity of Ulsan College of Medicine88 Olympic‐ro 43‐gilSongpa‐guSeoul05505Republic of Korea
| | - Suk‐Kyun Yang
- Department of GastroenterologyAsan Medical CenterUniversity of Ulsan College of Medicine88 Olympic‐ro 43‐gilSongpa‐guSeoul05505Republic of Korea
| | - Helen Moinova
- Department of Medicine and Case Comprehensive Cancer CenterCase Western Reserve University10900 Euclid AveClevelandOHUSA
| | - Sanford D. Markowitz
- Department of Medicine and Case Comprehensive Cancer CenterCase Western Reserve University10900 Euclid AveClevelandOHUSA
- University Hospitals Seidman Cancer Center10900 Euclid AveClevelandOHUSA
| | - Kwan Hyi Lee
- Center for BiomaterialsBiomedical Research InstituteKorea Institute of Science and Technology (KIST)5 Hwarangno 14‐gilSeongbuk‐guSeoul02792Republic of Korea
- Division of Bio‐Medical Science & TechnologyKIST School – Korea University of Science and Technology (UST)5 Hwarangno 14‐gilSeongbuk‐guSeoul02792Republic of Korea
| | - Seung‐Jae Myung
- Asan Institute for Life SciencesAsan Medical Center88 Olympic‐ro 43‐gilSongpa‐guSeoul05505Republic of Korea
- Department of Convergence MedicineUniversity of Ulsan College of Medicine88 Olympic‐ro 43‐gilSongpa‐guSeoul05505Republic of Korea
- Department of GastroenterologyAsan Medical CenterUniversity of Ulsan College of Medicine88 Olympic‐ro 43‐gilSongpa‐guSeoul05505Republic of Korea
| |
Collapse
|
14
|
Jee HG, Kim BA, Kim M, Yu HW, Choi JY, Kim SJ, Lee KE. Expression of SLC5A5 in Circulating Tumor Cells May Distinguish Follicular Thyroid Carcinomas from Adenomas: Implications for Blood-Based Preoperative Diagnosis. J Clin Med 2019; 8:jcm8020257. [PMID: 30781659 PMCID: PMC6406463 DOI: 10.3390/jcm8020257] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 02/13/2019] [Accepted: 02/13/2019] [Indexed: 12/25/2022] Open
Abstract
Preoperative diagnosis of thyroid nodules reduces unnecessary surgery. Circulating tumor cells (CTCs) may contain information of primary tumor(s). We asked whether the peripheral blood expression of genes specific for circulating tumor cells (CTCs) differentiates benign thyroid nodules from malignant nodules. Peripheral blood mononuclear cells from thyroid nodule patients (n = 20) were isolated preoperatively and the expression of seven CTC-associated genes was measured in patients with thyroid nodule(s) (n = 20). Among the tested genes, the expression of SLC5A5 and LGALS3 were validated in a larger number of patients (n = 64) and our results show that SLC5A5 expression differentiated follicular adenomas from follicular carcinomas (area under the curve (AUC) = 0.831). The expression of SLC5A5 in CTCs may preoperatively distinguish thyroid follicular adenomas from follicular carcinomas.
Collapse
Affiliation(s)
- Hyeon-Gun Jee
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea.
- Healthcare Innovation Park, Seoul National University Bundang Hospital, Seoungnam 13605, Korea.
| | - Byoung-Ae Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea.
| | - Minjun Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea.
| | - Hyeong Won Yu
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam 13620, Korea.
| | - June Young Choi
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam 13620, Korea.
| | - Su-Jin Kim
- Department of Surgery, Seoul National University Hospital and College of Medicine, Seoul 03080, Korea.
| | - Kyu Eun Lee
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea.
- Department of Surgery, Seoul National University Hospital and College of Medicine, Seoul 03080, Korea.
| |
Collapse
|
15
|
Immune Condition of Colorectal Cancer Patients Featured by Serum Chemokines and Gene Expressions of CD4+ Cells in Blood. Can J Gastroenterol Hepatol 2018; 2018:7436205. [PMID: 29992127 PMCID: PMC6016223 DOI: 10.1155/2018/7436205] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 04/20/2018] [Accepted: 05/09/2018] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC), the most common malignancy worldwide, causes inflammation. We explored the inflammatory pathophysiology of CRC by assessing the peripheral blood parameters. METHODS The differences in gene expression profiles of whole blood cells and cell subpopulations between CRC patients and healthy controls were analyzed using DNA microarray. Serum cytokine/chemokine concentrations in CRC patients and healthy controls were measured via multiplex detection immunoassays. In addition, we explored correlations between the expression levels of certain genes of peripheral CD4+ cells and serum chemokine concentrations. RESULTS The gene expression profiles of peripheral CD4+ cells of CRC patients differed from those of healthy controls, but this was not true of CD8+ cells, CD14+ cells, CD15+ cells, or CD19+ cells. Serum IL-8 and eotaxin-1 levels were significantly elevated in CRC patients, and the levels substantially correlated with the expression levels of certain genes of CD4+ cells. Interestingly, the relationships between gene expression levels in peripheral CD4+ cells and serum IL-8 and eotaxin-1 levels resembled those of monocytes/macrophages, not T cells. CONCLUSIONS Serum IL-8 and eotaxin-1 concentrations increased and were associated with changes in the gene expression of peripheral CD4+ cells in CRC patients.
Collapse
|
16
|
Neuropilin-1 Associated Molecules in the Blood Distinguish Poor Prognosis Breast Cancer: A Cross-Sectional Study. Sci Rep 2017; 7:3301. [PMID: 28607365 PMCID: PMC5468252 DOI: 10.1038/s41598-017-03280-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 04/28/2017] [Indexed: 12/12/2022] Open
Abstract
Circulating plasma and peripheral blood mononuclear (PBMCs) cells provide an informative snapshot of the systemic physiological state. Moreover, they provide a non-invasively accessible compartment to identify biomarkers for personalized medicine in advanced breast cancer. The role of Neuropilin-1 (NRP-1) and its interacting molecules in breast tumor tissue was correlated with cancer progression; however, the clinical impact of their systemic levels was not extensively evaluated. In this cross-sectional study, we found that circulating and tumor tissue expression of NRP-1 and circulating placental growth factor (PlGF) increase in advanced nodal and metastatic breast cancer compared with locally advanced disease. Tumor tissue expression of NRP-1 and PlGF is also upregulated in triple negative breast cancer (TNBC) compared to other subtypes. Conversely, in PBMCs, NRP-1 and its interacting molecules SEMA4A and SNAI1 are significantly downregulated in breast cancer patients compared to healthy controls, indicating a protective role. Moreover, we report differential PBMC expression profiles that correlate inversely with disease stage (SEMA4A, SNAI1, PLXNA1 and VEGFR3) and can differentiate between the TNBC and non-TNBC tumor subtypes (VEGFR3 and PLXNA1). This work supports the importance of NRP-1-associated molecules in circulation to characterize poor prognosis breast cancer and emphasizes on their role as favorable drug targets.
Collapse
|
17
|
Stewart JP, Richman S, Maughan T, Lawler M, Dunne PD, Salto-Tellez M. Standardising RNA profiling based biomarker application in cancer-The need for robust control of technical variables. Biochim Biophys Acta Rev Cancer 2017; 1868:258-272. [PMID: 28549623 DOI: 10.1016/j.bbcan.2017.05.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 05/21/2017] [Accepted: 05/22/2017] [Indexed: 01/10/2023]
Abstract
Histopathology-based staging of colorectal cancer (CRC) has utility in assessing the prognosis of patient subtypes, but as yet cannot accurately predict individual patient's treatment response. Transcriptomics approaches, using array based or next generation sequencing (NGS) platforms, of formalin fixed paraffin embedded tissue can be harnessed to develop multi-gene biomarkers for predicting both prognosis and treatment response, leading to stratification of treatment. While transcriptomics can shape future biomarker development, currently <1% of published biomarkers become clinically validated tests, often due to poor study design or lack of independent validation. In this review of a large number of CRC transcriptional studies, we identify recurrent sources of technical variability that encompass collection, preservation and storage of malignant tissue, nucleic acid extraction, methods to quantitate RNA transcripts and data analysis pipelines. We propose a series of defined steps for removal of these confounding issues, to ultimately aid in the development of more robust clinical biomarkers.
Collapse
Affiliation(s)
- James P Stewart
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, UK; Northern Ireland Molecular Pathology Laboratory, Queen's University Belfast, UK
| | - Susan Richman
- Department of Pathology and Tumour Biology, St James University Hospital, Leeds, UK
| | - Tim Maughan
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, UK
| | - Mark Lawler
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, UK
| | - Philip D Dunne
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, UK
| | - Manuel Salto-Tellez
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, UK; Northern Ireland Molecular Pathology Laboratory, Queen's University Belfast, UK.
| |
Collapse
|
18
|
Samatov TR, Galatenko VV, Block A, Shkurnikov MY, Tonevitsky AG, Schumacher U. Novel biomarkers in cancer: The whole is greater than the sum of its parts. Semin Cancer Biol 2016; 45:50-57. [PMID: 27639751 DOI: 10.1016/j.semcancer.2016.09.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 09/08/2016] [Indexed: 02/07/2023]
Abstract
The major issues hampering progress in the treatment of cancer patients are distant metastases and drug resistance to chemotherapy. Metastasis formation is a very complex process, and looking at gene signatures alone is not enough to get deep insight into it. This paper reviews traditional and novel approaches to identify gene signature biomarkers and intratumoural fluid pressure both as a novel way of creating predictive markers and as an obstacle to cancer therapy. Finally recently developed in vitro systems to predict the response of individual patient derived cancer explants to chemotherapy are discussed.
Collapse
Affiliation(s)
- Timur R Samatov
- SRC Bioclinicum, Ugreshskaya str 2/85, 115088, Moscow, Russia; Moscow State University of Mechanical Engineering, Bolshaya Semenovskaya str 38, 107023, Moscow, Russia
| | - Vladimir V Galatenko
- SRC Bioclinicum, Ugreshskaya str 2/85, 115088, Moscow, Russia; Lomonosov Moscow State University, Leninskie Gory, 119991, Moscow, Russia; National Research University Higher School of Economics, Kochnovsky Pass 3, 125319 Moscow, Russia
| | - Andreas Block
- Department of Oncology and Hematology, University Cancer Center, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Maxim Yu Shkurnikov
- P. Hertsen Moscow Oncology Research Institute, National Center of Medical Radiological Research, 3 Second Botkinsky Lane, Moscow, 125284, Russia
| | - Alexander G Tonevitsky
- Lomonosov Moscow State University, Leninskie Gory, 119991, Moscow, Russia; P. Hertsen Moscow Oncology Research Institute, National Center of Medical Radiological Research, 3 Second Botkinsky Lane, Moscow, 125284, Russia
| | - Udo Schumacher
- Department of Anatomy and Experimental Morphology, University Cancer Center, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany, Germany.
| |
Collapse
|
19
|
Ciarloni L, Ehrensberger SH, Imaizumi N, Monnier-Benoit S, Nichita C, Myung SJ, Kim JS, Song SY, Kim TI, van der Weg B, Meier R, Borovicka J, Beglinger C, Vallet C, Maerten P, Rüegg C, Dorta G. Development and Clinical Validation of a Blood Test Based on 29-Gene Expression for Early Detection of Colorectal Cancer. Clin Cancer Res 2016; 22:4604-11. [PMID: 27126992 DOI: 10.1158/1078-0432.ccr-15-2057] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 04/09/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE A blood test for early detection of colorectal cancer is a valuable tool for testing asymptomatic individuals and reducing colorectal cancer-related mortality. The objective of this study was to develop and validate a novel blood test able to differentiate patients with colorectal cancer and adenomatous polyps (AP) from individuals with a negative colonoscopy. EXPERIMENTAL DESIGN A case-control, multicenter clinical study was designed to collect blood samples from patients referred for colonoscopy or surgery. Predictive algorithms were developed on 75 controls, 61 large AP (LAP) ≥1 cm, and 45 colorectal cancer cases and independently validated on 74 controls, 42 LAP, and 52 colorectal cancer cases (23 stages I-II) as well as on 245 cases including other colorectal findings and diseases other than colorectal cancer. The test is based on a 29-gene panel expressed in peripheral blood mononuclear cells alone or in combination with established plasma tumor markers. RESULTS The 29-gene algorithm detected colorectal cancer and LAP with a sensitivity of 79.5% and 55.4%, respectively, with 90.0% specificity. Combination with the protein tumor markers carcinoembryonic antigen (CEA) and CYFRA21-2 resulted in a specificity increase (92.2%) with a sensitivity for colorectal cancer and LAP detection of 78.1% and 52.3%, respectively. CONCLUSIONS We report the validation of a novel blood test, Colox®, for the detection of colorectal cancer and LAP based on a 29-gene panel and the CEA and CYFRA21-1 plasma biomarkers. The performance and convenience of this routine blood test provide physicians a useful tool to test average-risk individuals unwilling to undergo upfront colonoscopy. Clin Cancer Res; 22(18); 4604-11. ©2016 AACR.
Collapse
Affiliation(s)
| | | | | | | | - Cristina Nichita
- Department of Gastroenterology and Hepatology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Seung-Jae Myung
- Asan Medical Centre, Department of Gastroenterology, University of Ulsan College of Medicine, Seoul, South Korea
| | - Joo Sung Kim
- Department of Internal Medicine, College of Medicine, Seoul National University, Seoul, South Korea
| | - Si Young Song
- Severance Hospital, Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Tae Il Kim
- Severance Hospital, Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | | | - Rémy Meier
- Kantonsspital Liestal, Gastroenterology, Hepatology and Nutrition Department, University Hospital, Liestal, Switzerland
| | - Jan Borovicka
- Kantonsspital St. Gallen, Department of Gastroenterology and Hepatology, St. Gallen, Switzerland
| | | | - Cédric Vallet
- Ensemble Hospitalier de la Côte, Surgery Services, Morges, Switzerland
| | | | - Curzio Rüegg
- Novigenix SA, Epalinges, Switzerland. Department of Medicine, Faculty of Science, University of Fribourg, Fribourg, Switzerland.
| | - Gian Dorta
- Department of Gastroenterology and Hepatology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland.
| |
Collapse
|
20
|
Gross ME. Blood-based gene expression profiling in castrate-resistant prostate cancer. BMC Med 2015; 13:219. [PMID: 26365516 PMCID: PMC4568585 DOI: 10.1186/s12916-015-0463-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 08/27/2015] [Indexed: 11/26/2022] Open
Abstract
Castrate-resistant prostate cancer (CRPC), the most life-threatening form of prostate cancer, has recently been the focus of many successful new treatments. Contemporary trials highlight the heterogeneous prognosis of CRPC as overall survival times vary greatly across different patient sub-groups. As presented in BMC Medicine, Wang et al. identify a blood-based prognostic signature in CRPC. Their approach is notable for discovery and validation of a four-gene model based on a whole-blood expression signature sampled from three distinct clinical cohorts. Further, the marker selection process incorporates an understanding of biological pathways expressed in myeloid or lymphoid cells which may provide some insight into host-tumor interactions as reflected in the peripheral blood. While the study includes a multivariate analysis accounting for many important clinical variables, larger datasets with more complete clinical information and sufficient follow-up are needed to confirm the independent significance of the four-gene expression model in a way which may better inform the care of CRPC patients.Please see related article: http://www.biomedcentral.com/1741-7015/13/201 .
Collapse
Affiliation(s)
- Mitchell E Gross
- Center for Applied Molecular Medicine, Keck School of Medicine, University of Southern California, 9033 Wilshire Boulevard, Suite 300, Beverly Hills, CA, 90211, USA.
| |
Collapse
|