1
|
Ghanegolmohammadi F, Eslami M, Ohya Y. Systematic data analysis pipeline for quantitative morphological cell phenotyping. Comput Struct Biotechnol J 2024; 23:2949-2962. [PMID: 39104709 PMCID: PMC11298594 DOI: 10.1016/j.csbj.2024.07.012] [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: 05/06/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 08/07/2024] Open
Abstract
Quantitative morphological phenotyping (QMP) is an image-based method used to capture morphological features at both the cellular and population level. Its interdisciplinary nature, spanning from data collection to result analysis and interpretation, can lead to uncertainties, particularly among those new to this actively growing field. High analytical specificity for a typical QMP is achieved through sophisticated approaches that can leverage subtle cellular morphological changes. Here, we outline a systematic workflow to refine the QMP methodology. For a practical review, we describe the main steps of a typical QMP; in each step, we discuss the available methods, their applications, advantages, and disadvantages, along with the R functions and packages for easy implementation. This review does not cover theoretical backgrounds, but provides several references for interested researchers. It aims to broaden the horizons for future phenome studies and demonstrate how to exploit years of endeavors to achieve more with less.
Collapse
Affiliation(s)
- Farzan Ghanegolmohammadi
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| | - Mohammad Eslami
- Harvard Ophthalmology AI Lab, Schepen’s Eye Research Institute of Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, USA
| | - Yoshikazu Ohya
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| |
Collapse
|
2
|
Hu J, Jiang Q, Mao W, Zhong S, Sun H, Mao K. STARD7 could be an immunological and prognostic biomarker: from pan-cancer analysis to hepatocellular carcinoma validation. Discov Oncol 2024; 15:543. [PMID: 39390226 PMCID: PMC11467145 DOI: 10.1007/s12672-024-01434-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 10/07/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND As the emergence of technologies such as sequencing and gene mapping, significant advancements have been made in understanding the landscape of tumors. However, the effective treatment of tumors continues to pose a tremendous challenge in clinical practice, which highlights the importance of predicting tumor markers and studying drug resistance mechanisms. The prognosis and differential expression of STARD7 in human pan-cancer were investigated by bioinformatic methods and experimental verification. METHODS The expression, diagnostic, and prognostic significance of the STARD7 gene were comprehensive analyzed using bioinformatics techniques. Furthermore, we validated our projected outcomes in liver cancer through experimental methodologies, including the use of qRT-PCR, CCK8 and transwell assays. RESULTS The STARD7 gene exhibits differential expression in 25 tumors, with high expression observed in 22 tumors. These distinct expression patterns within different tumor types are closely associated with poor prognosis and diagnosis. Furthermore, the STARD7 gene plays a role in regulating the tumor immune microenvironment. Methylation levels of STARD7 vary among 20 types of tumors and are correlated with survival outcomes. Furthermore, the experiment results demonstrated that STARD7 is highly expressed in hepatocellular carcinoma cells. Suppression of STARD7 significantly impedes the proliferation, migration, and invasion of HepG-2 and SMMC-7721 cells. CONCLUSIONS STARD7 has the potential to function as a crucial prognostic biomarker and exhibit correlation with tumor immunity in various types of human cancers. The implications of our findings extend to informing cancer immune-therapy and promoting the advancement of precision immune-oncology.
Collapse
Affiliation(s)
- Jie Hu
- Department of Pharmacy, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, No.100 Minjiang Road, Kecheng District, Quzhou, 324000, Zhejiang, China
| | - Qiu Jiang
- Department of Pharmacy, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, No.100 Minjiang Road, Kecheng District, Quzhou, 324000, Zhejiang, China
| | - Weili Mao
- Department of Pharmacy, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, No.100 Minjiang Road, Kecheng District, Quzhou, 324000, Zhejiang, China
| | - Songyang Zhong
- Department of Pharmacy, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, No.100 Minjiang Road, Kecheng District, Quzhou, 324000, Zhejiang, China
| | - Huayu Sun
- Department of Pharmacy, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, No.100 Minjiang Road, Kecheng District, Quzhou, 324000, Zhejiang, China.
| | - Kaili Mao
- Department of Pharmacy, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, No.100 Minjiang Road, Kecheng District, Quzhou, 324000, Zhejiang, China.
| |
Collapse
|
3
|
Di Cosimo S, Silvestri M, De Marco C, Calzoni A, De Santis MC, Carnevale MG, Reduzzi C, Cristofanilli M, Cappelletti V. Low-pass whole genome sequencing of circulating tumor cells to evaluate chromosomal instability in triple-negative breast cancer. Sci Rep 2024; 14:20479. [PMID: 39227622 PMCID: PMC11372142 DOI: 10.1038/s41598-024-71378-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 08/27/2024] [Indexed: 09/05/2024] Open
Abstract
Chromosomal Instability (CIN) is a common and evolving feature in breast cancer. Large-scale Transitions (LSTs), defined as chromosomal breakages leading to gains or losses of at least 10 Mb, have recently emerged as a metric of CIN due to their standardized definition across platforms. Herein, we report the feasibility of using low-pass Whole Genome Sequencing to assess LSTs, copy number alterations (CNAs) and their relationship in individual circulating tumor cells (CTCs) of triple-negative breast cancer (TNBC) patients. Initial assessment of LSTs in breast cancer cell lines consistently showed wide-ranging values (median 22, range 4-33, mean 21), indicating heterogeneous CIN. Subsequent analysis of CTCs revealed LST values (median 3, range 0-18, mean 5), particularly low during treatment, suggesting temporal changes in CIN levels. CNAs averaged 30 (range 5-49), with loss being predominant. As expected, CTCs with higher LSTs values exhibited increased CNAs. A CNA-based classifier of individual patient-derived CTCs, developed using machine learning, identified genes associated with both DNA proliferation and repair, such as RB1, MYC, and EXO1, as significant predictors of CIN. The model demonstrated a high predictive accuracy with an Area Under the Curve (AUC) of 0.89. Overall, these findings suggest that sequencing CTCs holds the potential to facilitate CIN evaluation and provide insights into its dynamic nature over time, with potential implications for monitoring TNBC progression through iterative assessments.
Collapse
Affiliation(s)
- Serena Di Cosimo
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Via Venezian 1, 20100, Milan, Italy
| | - Marco Silvestri
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Via Venezian 1, 20100, Milan, Italy.
- Isinnova S.R.L, Brescia, Italy.
| | - Cinzia De Marco
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Via Venezian 1, 20100, Milan, Italy
| | - Alessia Calzoni
- Isinnova S.R.L, Brescia, Italy
- Department of Information Engineering, University of Brescia, Brescia, Italy
| | - Maria Carmen De Santis
- Department of Radiation Oncology, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milan, Italy
- Breast Unit, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milan, Italy
| | - Maria Grazia Carnevale
- Department of Radiation Oncology, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milan, Italy
- Breast Unit, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Milan, Italy
| | - Carolina Reduzzi
- Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, USA
| | | | - Vera Cappelletti
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale Dei Tumori Di Milano, Via Venezian 1, 20100, Milan, Italy
| |
Collapse
|
4
|
Grewal US, Gaddam SJ, Beg MS, Brown TJ. Targeted therapies in advanced biliary malignancies: a clinical review. Expert Rev Anticancer Ther 2024; 24:869-880. [PMID: 39083012 DOI: 10.1080/14737140.2024.2387612] [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: 03/27/2024] [Accepted: 07/30/2024] [Indexed: 08/06/2024]
Abstract
INTRODUCTION Despite several therapeutic advancements, the proportion of patients with advanced biliary tract cancers (BTC) surviving 5 years from diagnosis remains dismal. The increasing recognition of targetable genetic alterations in BTCs has ushered in a new era in the treatment of these patients. Newer therapeutic agents targeting mutations such as isocitrate dehydrogenase (IDH), fibroblastic growth factor receptor (FGFR), human epidermal growth factor receptor (HER), and so on have established a new standard of care for treatment upon progression on frontline therapy in patients with disease harboring these mutations. AREAS COVERED The current review aims to concisely summarize progress with various targeted therapy options for BTC. We also briefly discuss future directions in clinical and translational research for the adoption of a personalized approach for the treatment of unresectable or advanced BTC. EXPERT OPINION Several new agents continue to emerge as feasible treatment options for patients with advanced BTC harboring targetable mutations. There is a growing need to identify mechanisms to conquer primary and acquired resistance to these agents. The identification of potential biomarkers that predict response to targeted therapy may be helpful in adopting a more tailored approach. All patients receiving treatment for advanced BTC should undergo tissue genomic profiling at diagnosis.
Collapse
Affiliation(s)
- Udhayvir S Grewal
- Division of Hematology and Oncology, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Shiva J Gaddam
- Division of Hematology and Oncology, Department of Medicine, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | | | - Timothy J Brown
- Division of Hematology and Medical Oncology, Department of Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
5
|
Gemelli M, Albini A, Catalano G, Incarbone M, Cannone M, Balladore E, Ricotta R, Pelosi G. Navigating resistance to ALK inhibitors in the lorlatinib era: a comprehensive perspective on NSCLC. Expert Rev Anticancer Ther 2024; 24:347-361. [PMID: 38630549 DOI: 10.1080/14737140.2024.2344648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 04/15/2024] [Indexed: 04/19/2024]
Abstract
INTRODUCTION The emergence of anaplastic lymphoma kinase (ALK) rearrangements in non-small cell lung cancer (NSCLC) has revolutionized targeted therapy. This dynamic landscape, featuring novel ALK inhibitors and combination therapies, necessitates a profound understanding of resistance mechanisms for effective treatment strategies. Recognizing two primary categories - on-target and off-target resistance - underscores the need for comprehensive assessment. AREAS COVERED This review delves into the intricacies of resistance to ALK inhibitors, exploring complexities in identification and management. Molecular testing, pivotal for early detection and accurate diagnosis, forms the foundation for patient stratification and resistance management. The literature search methodology involved comprehensive exploration of Pubmed and Embase. The multifaceted perspective encompasses new therapeutic horizons, ongoing clinical trials, and their clinical implications post the recent approval of lorlatinib. EXPERT OPINION Our expert opinion encapsulates the critical importance of understanding resistance mechanisms in the context of ALK inhibitors for shaping successful treatment approaches. With a focus on molecular testing and comprehensive assessment, this review contributes valuable insights to the evolving landscape of NSCLC therapy.
Collapse
Affiliation(s)
- Maria Gemelli
- Medical Oncology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milan, Italy
| | - Adriana Albini
- Departement of Scientific Directorate, European Institute of Oncology (IEO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Gianpiero Catalano
- Radiation Oncology Center, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milan, Italy
| | - Matteo Incarbone
- Department of Surgery, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milan, Italy
| | - Maria Cannone
- Inter-Hospital Division of Pathology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milan, Italy
| | - Emanuela Balladore
- Inter-Hospital Division of Pathology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milan, Italy
| | - Riccardo Ricotta
- Medical Oncology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milan, Italy
| | - Giuseppe Pelosi
- Inter-Hospital Division of Pathology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
6
|
Kersch CN, Kim M, Stoller J, Barajas RF, Park JE. Imaging Genomics of Glioma Revisited: Analytic Methods to Understand Spatial and Temporal Heterogeneity. AJNR Am J Neuroradiol 2024; 45:537-548. [PMID: 38548303 PMCID: PMC11288537 DOI: 10.3174/ajnr.a8148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 11/09/2023] [Indexed: 04/12/2024]
Abstract
An improved understanding of the cellular and molecular biologic processes responsible for brain tumor development, growth, and resistance to therapy is fundamental to improving clinical outcomes. Imaging genomics is the study of the relationships between microscopic, genetic, and molecular biologic features and macroscopic imaging features. Imaging genomics is beginning to shift clinical paradigms for diagnosing and treating brain tumors. This article provides an overview of imaging genomics in gliomas, in which imaging data including hallmarks such as IDH-mutation, MGMT methylation, and EGFR-mutation status can provide critical insights into the pretreatment and posttreatment stages. This article will accomplish the following: 1) review the methods used in imaging genomics, including visual analysis, quantitative analysis, and radiomics analysis; 2) recommend suitable analytic methods for imaging genomics according to biologic characteristics; 3) discuss the clinical applicability of imaging genomics; and 4) introduce subregional tumor habitat analysis with the goal of guiding future radiogenetics research endeavors toward translation into critically needed clinical applications.
Collapse
Affiliation(s)
- Cymon N Kersch
- From the Department of Radiation Medicine (C.N.K.), Oregon Health and Science University, Portland, Oregon
| | - Minjae Kim
- Department of Radiology and Research Institute of Radiology (M.K., J.E.P.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jared Stoller
- Department of Diagnostic Radiology (J.S., R.F.B.), Oregon Health and Science University, Portland, Oregon
| | - Ramon F Barajas
- Department of Diagnostic Radiology (J.S., R.F.B.), Oregon Health and Science University, Portland, Oregon
- Knight Cancer Institute (R.F.B.), Oregon Health and Science University, Portland, Oregon
- Advanced Imaging Research Center (R.F.B.), Oregon Health and Science University, Portland, Oregon
| | - Ji Eun Park
- Department of Radiology and Research Institute of Radiology (M.K., J.E.P.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
7
|
LaPelusa M, Cann C, Ciombor KK, Eng C. Mutational Signature Changes in Patients With Metastatic Squamous Cell Carcinoma of the Anal Canal. Oncologist 2024; 29:e475-e486. [PMID: 38103030 PMCID: PMC10994269 DOI: 10.1093/oncolo/oyad326] [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: 09/05/2023] [Accepted: 11/14/2023] [Indexed: 12/17/2023] Open
Abstract
PURPOSE We examined the concordance of genetic mutations between pretreatment tumor tissue and posttreatment circulating tumor DNA (ctDNA) in patients with metastatic squamous cell carcinoma of the anal canal (SCCA) and assessed the impact of therapy on this concordance. METHODS We analyzed next-generation sequencing reports from pretreatment tumor tissue and posttreatment ctDNA in 11 patients with metastatic SCCA treated at Vanderbilt University Medical Center between 2017 and 2021. RESULTS Among the mutations identified in posttreatment ctDNA, 34.5% were also found in pretreatment tumor tissue, while 47.6% of pretreatment tumor tissue mutations were found in posttreatment ctDNA. Four patients had preservation of potentially actionable mutations in both pretreatment tissue and posttreatment ctDNA, while 7 patients had newly identified mutations in posttreatment ctDNA that were not present in pretreatment tumor tissue. CONCLUSION Patients with SCCA demonstrate a high degree of temporal mutational heterogeneity. This supports the hypothesis that ctDNA can serve as a real-time tracking mechanism for solid tumors' molecular evolution in response to therapy. Our findings highlight the potential of ctDNA in identifying emerging actionable mutations, supplementing information from tissue-based genomic assessments. Further research, ideally with larger and multi-institutional cohorts, is needed to validate our findings in this relatively rare tumor type.
Collapse
Affiliation(s)
- Michael LaPelusa
- Department of Internal Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Christopher Cann
- Division of Hematology and Oncology, Department of Internal Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kristen K Ciombor
- Division of Hematology and Oncology, Department of Internal Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cathy Eng
- Division of Hematology and Oncology, Department of Internal Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
8
|
Lynch A, Bradford S, Burkard ME. The reckoning of chromosomal instability: past, present, future. Chromosome Res 2024; 32:2. [PMID: 38367036 DOI: 10.1007/s10577-024-09746-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 01/11/2024] [Accepted: 01/27/2024] [Indexed: 02/19/2024]
Abstract
Quantitative measures of CIN are crucial to our understanding of its role in cancer. Technological advances have changed the way CIN is quantified, offering increased accuracy and insight. Here, we review measures of CIN through its rise as a field, discuss considerations for its measurement, and look forward to future quantification of CIN.
Collapse
Affiliation(s)
- Andrew Lynch
- UW Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
- McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, WI, USA
- Division of Hematology/Oncology, Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Shermineh Bradford
- UW Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
- McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, WI, USA
- Division of Hematology/Oncology, Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Mark E Burkard
- UW Carbone Cancer Center, University of Wisconsin, Madison, WI, USA.
- McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, WI, USA.
- Division of Hematology/Oncology, Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA.
| |
Collapse
|
9
|
Wang X, Bai H, Zhang J, Wang Z, Duan J, Cai H, Cao Z, Lin Q, Ding X, Sun Y, Zhang W, Xu X, Chen H, Zhang D, Feng X, Wan J, Zhang J, He J, Wang J. Genetic Intratumor Heterogeneity Remodels the Immune Microenvironment and Induces Immune Evasion in Brain Metastasis of Lung Cancer. J Thorac Oncol 2024; 19:252-272. [PMID: 37717855 DOI: 10.1016/j.jtho.2023.09.276] [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: 06/06/2023] [Revised: 08/18/2023] [Accepted: 09/07/2023] [Indexed: 09/19/2023]
Abstract
INTRODUCTION Brain metastasis, with the highest incidence in patients with lung cancer, significantly worsens prognosis and poses challenges to clinical management. To date, how brain metastasis evades immune elimination remains unknown. METHODS Whole-exome sequencing and RNA sequencing were performed on 30 matched brain metastasis, primary lung adenocarcinoma, and normal tissues. Data from The Cancer Genome Atlas primary lung adenocarcinoma cohort, including multiplex immunofluorescence, were used to support the findings of bioinformatics analysis. RESULTS Our study highlights the key role of intratumor heterogeneity of genomic alterations in the metastasis process, mainly caused by homologous recombination deficiency or other somatic copy number alteration-associated mutation mechanisms, leading to increased genomic instability and genomic complexity. We further proposed a selection model of brain metastatic evolution in which intratumor heterogeneity drives immune remodeling, leading to immune escape through different mechanisms under local immune pressure. CONCLUSIONS Our findings provide novel insights into the metastatic process and immune escape mechanisms of brain metastasis and pave the way for precise immunotherapeutic strategies for patients with lung cancer with brain metastasis.
Collapse
Affiliation(s)
- Xin Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hua Bai
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiyang Zhang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhijie Wang
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianchun Duan
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hongqing Cai
- Department of Neurosurgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zheng Cao
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qingtang Lin
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiaosheng Ding
- Department of Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yiting Sun
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Zhang
- Department of Translational Medicine, 3D Medicines Inc., Shanghai, China
| | - Xiaoya Xu
- Department of Translational Medicine, 3D Medicines Inc., Shanghai, China
| | - Hao Chen
- Department of Translational Medicine, 3D Medicines Inc., Shanghai, China
| | - Dadong Zhang
- Department of Translational Medicine, 3D Medicines Inc., Shanghai, China
| | - Xiaoli Feng
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jinghai Wan
- Department of Neurosurgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianjun Zhang
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie Wang
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
10
|
Giunta EF, Malapelle U, Russo A, De Giorgi U. Blood-based liquid biopsy in advanced prostate cancer. Crit Rev Oncol Hematol 2024; 194:104241. [PMID: 38122919 DOI: 10.1016/j.critrevonc.2023.104241] [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: 08/16/2023] [Revised: 11/25/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023] Open
Abstract
Prostate cancer is characterized by several genetic alterations which could impact prognosis and therapeutic decisions in the advanced disease. Tissue biopsy is still considered the gold standard approach for molecular characterization in prostate cancer, but it has several limitations, including the possibility of insufficient/inadequate tumor tissue to be analyzed. Blood-based liquid biopsy is a non-invasive method to investigate tumor cell derivatives in the bloodstream, being a valid alternative to tissue biopsy for molecular characterization but also for predictive and/or prognostic purposes. In this review, we analyze the most relevant evidence in this field, focusing on clinically relevant targets such as HRD genetic alterations and also focusing on the differences between tissue and liquid biopsy in light of the data from the latest clinical trials.
Collapse
Affiliation(s)
- Emilio Francesco Giunta
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) 'Dino Amadori', Meldola, FC, Italy.
| | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Antonio Russo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Ugo De Giorgi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) 'Dino Amadori', Meldola, FC, Italy
| |
Collapse
|
11
|
Pellegrino S, Fonti R, Vallone C, Morra R, Matano E, De Placido S, Del Vecchio S. Coefficient of Variation in Metastatic Lymph Nodes Determined by 18F-FDG PET/CT in Patients with Advanced NSCLC: Combination with Coefficient of Variation in Primary Tumors. Cancers (Basel) 2024; 16:279. [PMID: 38254770 PMCID: PMC10813913 DOI: 10.3390/cancers16020279] [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: 12/05/2023] [Revised: 01/03/2024] [Accepted: 01/06/2024] [Indexed: 01/24/2024] Open
Abstract
Purpose The aim of the present study was to test whether the coefficient of variation (CoV) of 18F-FDG PET/CT images of metastatic lymph nodes and primary tumors may predict clinical outcome in patients with advanced non-small cell lung cancer (NSCLC). Materials and Methods Fifty-eight NSCLC patients who had undergone 18F-FDG PET/CT at diagnosis were evaluated. SUVmax, SUVmean, CoV, MTV and TLG were determined in targeted lymph nodes and corresponding primary tumors along with Total MTV (MTVTOT) and Whole-Body TLG (TLGWB) of all malignant lesions. Univariate analysis was performed using Cox proportional hazards regression whereas the Kaplan-Meier method and log-rank tests were used for survival analysis. Results Fifty-eight metastatic lymph nodes were analyzed and average values of SUVmax, SUVmean, CoV, MTV and TLG were 11.89 ± 8.54, 4.85 ± 1.90, 0.37 ± 0.16, 46.16 ± 99.59 mL and 256.84 ± 548.27 g, respectively, whereas in primary tumors they were 11.92 ± 6.21, 5.47 ± 2.34, 0.36 ± 0.14, 48.03 ± 64.45 mL and 285.21 ± 397.95 g, respectively. At univariate analysis, overall survival (OS) was predicted by SUVmax (p = 0.0363), SUVmean (p = 0.0200) and CoV (p = 0.0139) of targeted lymph nodes as well as by CoV of primary tumors (p = 0.0173), MTVTOT (p = 0.0007), TLGWB (p = 0.0129) and stage (p = 0.0122). Using Kaplan-Meier analysis, OS was significantly better in patients with CoV of targeted lymph nodes ≤ 0.29 than those with CoV > 0.29 (p = 0.0147), meanwhile patients with CoV of primary tumors > 0.38 had a better prognosis compared to those with CoV ≤ 0.38 (p = 0.0137). Finally, we combined the CoV values of targeted lymph nodes and primary tumors in all possible arrangements and a statistically significant difference was found among the four survival curves (p = 0.0133). In particular, patients with CoV of targeted lymph nodes ≤ 0.29 and CoV of primary tumors > 0.38 had the best prognosis. Conclusions The CoV of targeted lymph nodes combined with the CoV of primary tumors can predict prognosis of NSCLC patients.
Collapse
Affiliation(s)
- Sara Pellegrino
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (S.P.); (R.F.); (C.V.)
| | - Rosa Fonti
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (S.P.); (R.F.); (C.V.)
| | - Carlo Vallone
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (S.P.); (R.F.); (C.V.)
| | - Rocco Morra
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80131 Naples, Italy; (R.M.); (E.M.); (S.D.P.)
| | - Elide Matano
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80131 Naples, Italy; (R.M.); (E.M.); (S.D.P.)
| | - Sabino De Placido
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80131 Naples, Italy; (R.M.); (E.M.); (S.D.P.)
| | - Silvana Del Vecchio
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (S.P.); (R.F.); (C.V.)
| |
Collapse
|
12
|
Wu YP, Wu L, Ou J, Cao JM, Fu MY, Chen TW, Ouchi E, Hu J. Preoperative CT radiomics of esophageal squamous cell carcinoma and lymph node to predict nodal disease with a high diagnostic capability. Eur J Radiol 2024; 170:111197. [PMID: 37992611 DOI: 10.1016/j.ejrad.2023.111197] [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: 09/25/2023] [Revised: 10/12/2023] [Accepted: 11/13/2023] [Indexed: 11/24/2023]
Abstract
PURPOSE To develop CT radiomics models of resectable esophageal squamous cell carcinoma (ESCC) and lymph node (LN) to preoperatively identify LN+. MATERIALS AND METHODS 299 consecutive patients with ESCC were enrolled in the study, 140 of whom were LN+ and 159 were LN-. Of the 299 patients, 249 (from the same hospital) were randomly divided into a training cohort (n = 174) and a test cohort (n = 75). The remaining 50 patients, from a second hospital, were assigned to an external validation cohort. In the training cohort, preoperative contrast-enhanced CT radiomics features of ESCC and LN were extracted, then integrated with clinical features to develop three models: ESCC, LN and combined. The performance of these models was assessed using area under receiver operating characteristic curve (AUC), and F-1 score, which were validated in both the test cohort and external validation cohort. RESULTS An ESCC model was developed for the training cohort utilizing the 8 tumor radiomics features, and an LN model was constructed using 9 nodal radiomics features. A combined model was constructed using both ESCC and LN extracted features, in addition to cT stage and LN+ distribution. This combined model had the highest predictive ability among the three models in the training cohort (AUC = 0.948, F1-score = 0.878). The predictive ability was validated in both the test and external validation cohorts (AUC = 0.885 and 0.867, F1-score = 0.816 and 0.773, respectively). CONCLUSION To preoperatively determine LN+, the combined model is superior to models of ESCC and LN alone.
Collapse
Affiliation(s)
- Yu-Ping Wu
- Department of Radiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China; Medical Imaging Key Laboratory of Sichuan Province, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Lan Wu
- Department of Radiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Ou
- Medical Imaging Key Laboratory of Sichuan Province, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Jin-Ming Cao
- Medical Imaging Key Laboratory of Sichuan Province, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China; Department of Radiology, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong, China
| | - Mao-Yong Fu
- Department of Thoracic Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Tian-Wu Chen
- Department of Radiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China; Medical Imaging Key Laboratory of Sichuan Province, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China.
| | - Erika Ouchi
- Department of Radiology, Wayne State University, Detroit, MI, USA
| | - Jiani Hu
- Department of Radiology, Wayne State University, Detroit, MI, USA
| |
Collapse
|
13
|
Caro GD, Lam ET, Bourdon D, Blankfard M, Dharajiya N, Slade M, Williams E, Zhang D, Wenstrup R, Schwartzberg L. A novel liquid biopsy assay for detection of ERBB2 (HER2) amplification in circulating tumor cells (CTCs). J Circ Biomark 2024; 13:27-35. [PMID: 39377016 PMCID: PMC11456801 DOI: 10.33393/jcb.2024.3046] [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: 02/14/2024] [Accepted: 08/26/2024] [Indexed: 10/09/2024] Open
Abstract
Purpose Circulating tumor cell (CTC)-based ERBB2 (HER2) assay is a laboratory test developed by Epic Sciences using single-cell genomics to detect ERBB2 (HER2) amplification in CTCs found in the peripheral blood of metastatic breast cancer (MBC) patients. Patients and methods Peripheral blood was collected in Streck tubes and centrifugation was used to remove plasma and red blood cells. The remaining nucleated cells were deposited on glass slides, immunofluorescent-stained with proprietary antibodies, scanned by a high-definition digital scanner, and analyzed by a proprietary algorithm. In addition, single-cell genomics was performed on selected CTC. Analytical validation was performed using white blood cells from healthy donors and breast cancer cell lines with known levels of ERBB2 amplification. Clinical concordance was assessed on MBC patients whose blood was tested by the CTC ERBB2 (HER2) assay and those results are compared to results of matched metastatic tissue biopsy (immunohistochemistry [IHC] 3+ or IHC2+/in situ hybridization [ISH+]). Results Epic's ERBB2 (HER2) assay detected 2-fold ERBB2 amplification with 85% sensitivity and 94% specificity. In the clinical concordance study, among the 50% of the cases that had ERBB2 status results from CTCs found to be chromosomally-unstable, the CTC ERBB2 (HER2) assay showed sensitivity of 69% and specificity of 78% when compared to HER2 status by metastatic tissue biopsy. Conclusions The CTC ERBB2 (HER2) assay can consistently detect ERBB2 status in MBC cell lines and in the population of patients with MBC with detectable chromosomally unstable CTCs for whom tissue biopsy is not available or is infeasible.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Dong Zhang
- Epic Sciences, San Diego, California - USA
| | | | | |
Collapse
|
14
|
Dong H, Yang L, Shaofeng D, Lili G. Feasibility Study of Computed Tomographic Radiomics Model for the Prediction of Early and Intermediate Stage Hepatocellular Carcinoma Using BCLC Staging. Technol Cancer Res Treat 2024; 23:15330338241245943. [PMID: 38660703 PMCID: PMC11044781 DOI: 10.1177/15330338241245943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 02/29/2024] [Accepted: 03/19/2024] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a serious health concern because of its high morbidity and mortality. The prognosis of HCC largely depends on the disease stage at diagnosis. Computed tomography (CT) image textural analysis is an image analysis technique that has emerged in recent years. OBJECTIVE To probe the feasibility of a CT radiomic model for predicting early (stages 0, A) and intermediate (stage B) HCC using Barcelona Clinic Liver Cancer (BCLC) staging. METHODS A total of 190 patients with stages 0, A, or B HCC according to CT-enhanced arterial and portal vein phase images were retrospectively assessed. The lesions were delineated manually to construct a region of interest (ROI) consisting of the entire tumor mass. Consequently, the textural profiles of the ROIs were extracted by specific software. Least absolute shrinkage and selection operator dimensionality reduction was used to screen the textural profiles and obtain the area under the receiver operating characteristic curve values. RESULTS Within the test cohort, the area under the curve (AUC) values associated with arterial-phase images and BCLC stages 0, A, and B disease were 0.99, 0.98, and 0.99, respectively. The overall accuracy rate was 92.7%. The AUC values associated with portal vein phase images and BCLC stages 0, A, and B disease were 0.98, 0.95, and 0.99, respectively, with an overall accuracy of 90.9%. CONCLUSION The CT radiomic model can be used to predict the BCLC stage of early-stage and intermediate-stage HCC.
Collapse
Affiliation(s)
- Han Dong
- The Affiliated Huai’an First People's Hospital of Nanjing Medical University, Huai’an, Jiangsu Province, China
| | - Lu Yang
- The Affiliated Huai’an First People's Hospital of Nanjing Medical University, Huai’an, Jiangsu Province, China
| | | | - Guo Lili
- The Affiliated Huai’an First People's Hospital of Nanjing Medical University, Huai’an, Jiangsu Province, China
| |
Collapse
|
15
|
Hasanzadeh A, Ebadati A, Dastanpour L, Aref AR, Sahandi Zangabad P, Kalbasi A, Dai X, Mehta G, Ghasemi A, Fatahi Y, Joshi S, Hamblin MR, Karimi M. Applications of Innovation Technologies for Personalized Cancer Medicine: Stem Cells and Gene-Editing Tools. ACS Pharmacol Transl Sci 2023; 6:1758-1779. [PMID: 38093832 PMCID: PMC10714436 DOI: 10.1021/acsptsci.3c00102] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 10/19/2023] [Accepted: 10/23/2023] [Indexed: 02/16/2024]
Abstract
Personalized medicine is a new approach toward safer and even cheaper treatments with minimal side effects and toxicity. Planning a therapy based on individual properties causes an effective result in a patient's treatment, especially in a complex disease such as cancer. The benefits of personalized medicine include not only early diagnosis with high accuracy but also a more appropriate and effective therapeutic approach based on the unique clinical, genetic, and epigenetic features and biomarker profiles of a specific patient's disease. In order to achieve personalized cancer therapy, understanding cancer biology plays an important role. One of the crucial applications of personalized medicine that has gained consideration more recently due to its capability in developing disease therapy is related to the field of stem cells. We review various applications of pluripotent, somatic, and cancer stem cells in personalized medicine, including targeted cancer therapy, cancer modeling, diagnostics, and drug screening. CRISPR-Cas gene-editing technology is then discussed as a state-of-the-art biotechnological advance with substantial impacts on medical and therapeutic applications. As part of this section, the role of CRISPR-Cas genome editing in recent cancer studies is reviewed as a further example of personalized medicine application.
Collapse
Affiliation(s)
- Akbar Hasanzadeh
- Cellular
and Molecular Research Center, Iran University
of Medical Sciences, Tehran 14535, Iran
- Department
of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 14535, Iran
- Advances
Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran 14535, Iran
| | - Arefeh Ebadati
- Cellular
and Molecular Research Center, Iran University
of Medical Sciences, Tehran 14535, Iran
- Department
of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 14535, Iran
- Advances
Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran 14535, Iran
| | - Lida Dastanpour
- Cellular
and Molecular Research Center, Iran University
of Medical Sciences, Tehran 14535, Iran
- Department
of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 14535, Iran
- Advances
Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran 14535, Iran
| | - Amir R. Aref
- Department
of Medical Oncology and Belfer Center for Applied Cancer Science, Dana Farber Cancer Institute, Boston, Massachusetts 02115, United States
| | - Parham Sahandi Zangabad
- Monash
Institute of Pharmaceutical Sciences, Department of Pharmacy and Pharmaceutical
Sciences, Monash University, Parkville, Melbourne, Victoria 3052, Australia
| | - Alireza Kalbasi
- Department
of Medical Oncology, Dana-Farber Cancer
Institute, Boston, Massachusetts 02115, United States
| | - Xiaofeng Dai
- School of
Biotechnology, Jiangnan University, Wuxi 214122, China
- National
Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi 214122, China
- Jiangsu Provincial
Research Center for Bioactive Product Processing Technology, Jiangnan University, Wuxi 214122, China
| | - Geeta Mehta
- Department
of Biomedical Engineering, University of
Michigan, Ann Arbor, Michigan 48109, United States
- Department
of Materials Science and Engineering, University
of Michigan, Ann Arbor, Michigan 48109, United States
- Macromolecular
Science and Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
- Rogel Cancer
Center, University of Michigan, Ann Arbor, Michigan 48109, United States
- Precision
Health, University of Michigan, Ann Arbor, Michigan 48105, United States
| | - Amir Ghasemi
- Department
of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 14535, Iran
- Department
of Materials Science and Engineering, Sharif
University of Technology, Tehran 14588, Iran
| | - Yousef Fatahi
- Nanotechnology
Research Centre, Faculty of Pharmacy, Tehran
University of Medical Sciences, Tehran 14166, Iran
- Department
of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 14166, Iran
- Universal
Scientific Education and Research Network (USERN), Tehran 14166, Iran
| | - Suhasini Joshi
- Chemical
Biology Program, Memorial Sloan Kettering
Cancer Center, New York, New York 10065, United States
| | - Michael R. Hamblin
- Laser Research
Centre, Faculty of Health Science, University
of Johannesburg, Doornfontein 2028, South Africa
- Radiation
Biology Research Center, Iran University
of Medical Sciences, Tehran 14535, Iran
| | - Mahdi Karimi
- Cellular
and Molecular Research Center, Iran University
of Medical Sciences, Tehran 14535, Iran
- Department
of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 14535, Iran
- Oncopathology
Research Center, Iran University of Medical
Sciences, Tehran 14535, Iran
- Research
Center for Science and Technology in Medicine, Tehran University of Medical Sciences, Tehran 14166, Iran
- Applied
Biotechnology Research Centre, Tehran Medical Science, Islamic Azad University, Tehran 14166, Iran
| |
Collapse
|
16
|
Sashittal P, Zhang H, Iacobuzio-Donahue CA, Raphael BJ. ConDoR: tumor phylogeny inference with a copy-number constrained mutation loss model. Genome Biol 2023; 24:272. [PMID: 38037115 PMCID: PMC10688497 DOI: 10.1186/s13059-023-03106-5] [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: 12/27/2022] [Accepted: 11/07/2023] [Indexed: 12/02/2023] Open
Abstract
A tumor contains a diverse collection of somatic mutations that reflect its past evolutionary history and that range in scale from single nucleotide variants (SNVs) to large-scale copy-number aberrations (CNAs). However, no current single-cell DNA sequencing (scDNA-seq) technology produces accurate measurements of both SNVs and CNAs, complicating the inference of tumor phylogenies. We introduce a new evolutionary model, the constrained k-Dollo model, that uses SNVs as phylogenetic markers but constrains losses of SNVs according to clusters of cells. We derive an algorithm, ConDoR, that infers phylogenies from targeted scDNA-seq data using this model. We demonstrate the advantages of ConDoR on simulated and real scDNA-seq data.
Collapse
Affiliation(s)
| | - Haochen Zhang
- Gerstner Sloan Kettering Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, NY, USA
| | - Christine A Iacobuzio-Donahue
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, NY, USA
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, NY, USA
| | | |
Collapse
|
17
|
Jönsson H, Ahlström H, Kullberg J. Spatial mapping of tumor heterogeneity in whole-body PET-CT: a feasibility study. Biomed Eng Online 2023; 22:110. [PMID: 38007471 PMCID: PMC10675915 DOI: 10.1186/s12938-023-01173-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 11/17/2023] [Indexed: 11/27/2023] Open
Abstract
BACKGROUND Tumor heterogeneity is recognized as a predictor of treatment response and patient outcome. Quantification of tumor heterogeneity across all scales may therefore provide critical insight that ultimately improves cancer management. METHODS An image registration-based framework for the study of tumor heterogeneity in whole-body images was evaluated on a dataset of 490 FDG-PET-CT images of lung cancer, lymphoma, and melanoma patients. Voxel-, lesion- and subject-level features were extracted from the subjects' segmented lesion masks and mapped to female and male template spaces for voxel-wise analysis. Resulting lesion feature maps of the three subsets of cancer patients were studied visually and quantitatively. Lesion volumes and lesion distances in subject spaces were compared with resulting properties in template space. The strength of the association between subject and template space for these properties was evaluated with Pearson's correlation coefficient. RESULTS Spatial heterogeneity in terms of lesion frequency distribution in the body, metabolic activity, and lesion volume was seen between the three subsets of cancer patients. Lesion feature maps showed anatomical locations with low versus high mean feature value among lesions sampled in space and also highlighted sites with high variation between lesions in each cancer subset. Spatial properties of the lesion masks in subject space correlated strongly with the same properties measured in template space (lesion volume, R = 0.986, p < 0.001; total metabolic volume, R = 0.988, p < 0.001; maximum within-patient lesion distance, R = 0.997, p < 0.001). Lesion volume and total metabolic volume increased on average from subject to template space (lesion volume, 3.1 ± 52 ml; total metabolic volume, 53.9 ± 229 ml). Pair-wise lesion distance decreased on average by 0.1 ± 1.6 cm and maximum within-patient lesion distance increased on average by 0.5 ± 2.1 cm from subject to template space. CONCLUSIONS Spatial tumor heterogeneity between subsets of interest in cancer cohorts can successfully be explored in whole-body PET-CT images within the proposed framework. Whole-body studies are, however, especially prone to suffer from regional variation in lesion frequency, and thus statistical power, due to the non-uniform distribution of lesions across a large field of view.
Collapse
Affiliation(s)
- Hanna Jönsson
- Section of Radiology, Department of Surgical Sciences, Uppsala University, 751 85, Uppsala, Sweden.
| | - Håkan Ahlström
- Section of Radiology, Department of Surgical Sciences, Uppsala University, 751 85, Uppsala, Sweden
- Antaros Medical AB, BioVenture Hub, 431 53, Mölndal, Sweden
| | - Joel Kullberg
- Section of Radiology, Department of Surgical Sciences, Uppsala University, 751 85, Uppsala, Sweden
- Antaros Medical AB, BioVenture Hub, 431 53, Mölndal, Sweden
| |
Collapse
|
18
|
Wollenzien H, Tecleab YA, Szczepaniak-Sloane R, Restaino A, Kareta MS. Single-Cell Evolutionary Analysis Reveals Drivers of Plasticity and Mediators of Chemoresistance in Small Cell Lung Cancer. Mol Cancer Res 2023; 21:892-907. [PMID: 37256926 PMCID: PMC10527088 DOI: 10.1158/1541-7786.mcr-22-0881] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 03/11/2023] [Accepted: 05/24/2023] [Indexed: 06/02/2023]
Abstract
Small cell lung cancer (SCLC) is often a heterogeneous tumor, where dynamic regulation of key transcription factors can drive multiple populations of phenotypically different cells which contribute differentially to tumor dynamics. This tumor is characterized by a very low 2-year survival rate, high rates of metastasis, and rapid acquisition of chemoresistance. The heterogeneous nature of this tumor makes it difficult to study and to treat, as it is not clear how or when this heterogeneity arises. Here we describe temporal, single-cell analysis of SCLC to investigate tumor initiation and chemoresistance in both SCLC xenografts and an autochthonous SCLC model. We identify an early population of tumor cells with high expression of AP-1 network genes that are critical for tumor growth. Furthermore, we have identified and validated the cancer testis antigens (CTA) PAGE5 and GAGE2A as mediators of chemoresistance in human SCLC. CTAs have been successfully targeted in other tumor types and may be a promising avenue for targeted therapy in SCLC. IMPLICATIONS Understanding the evolutionary dynamics of SCLC can shed light on key mechanisms such as cellular plasticity, heterogeneity, and chemoresistance.
Collapse
Affiliation(s)
- Hannah Wollenzien
- Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, South Dakota, USA
- Genetics & Genomics Group, Sanford Research, Sioux Falls, South Dakota, USA
- Division of Basic Biomedical Sciences, University of South Dakota, Vermillion, South Dakota, USA
| | | | - Robert Szczepaniak-Sloane
- Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, South Dakota, USA
- Genetics & Genomics Group, Sanford Research, Sioux Falls, South Dakota, USA
| | - Anthony Restaino
- Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, South Dakota, USA
- Department of Pediatrics, Sanford School of Medicine, Sioux Falls, South Dakota, USA
| | - Michael S. Kareta
- Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, South Dakota, USA
- Genetics & Genomics Group, Sanford Research, Sioux Falls, South Dakota, USA
- Division of Basic Biomedical Sciences, University of South Dakota, Vermillion, South Dakota, USA
- Functional Genomics & Bioinformatics Core, Sanford Research, Sioux Falls, SD, USA
- Department of Pediatrics, Sanford School of Medicine, Sioux Falls, South Dakota, USA
- Department of Biochemistry, South Dakota State University, Brookings, South Dakota, USA
| |
Collapse
|
19
|
Yan M, Liu Q. The nature of cancer. Front Med 2023; 17:796-803. [PMID: 36913173 DOI: 10.1007/s11684-022-0975-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 12/05/2022] [Indexed: 03/14/2023]
Affiliation(s)
- Min Yan
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Quentin Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China.
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, 116023, China.
| |
Collapse
|
20
|
Ingles Garces AH, Porta N, Graham TA, Banerji U. Clinical trial designs for evaluating and exploiting cancer evolution. Cancer Treat Rev 2023; 118:102583. [PMID: 37331179 DOI: 10.1016/j.ctrv.2023.102583] [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: 04/03/2023] [Revised: 05/21/2023] [Accepted: 05/23/2023] [Indexed: 06/20/2023]
Abstract
The evolution of drug-resistant cell subpopulations causes cancer treatment failure. Current preclinical evidence shows that it is possible to model herding of clonal evolution and collateral sensitivity where an initial treatment could favourably influence the response to a subsequent one. Novel therapy strategies exploiting this understanding are being considered, and clinical trial designs for steering cancer evolution are needed. Furthermore, preclinical evidence suggests that different subsets of drug-sensitive and resistant clones could compete between themselves for nutrients/blood supply, and clones that populate a tumour do so at the expense of other clones. Treatment paradigms based on this clinical application of exploiting cell-cell competition include intermittent dosing regimens or cycling different treatments before progression. This will require clinical trial designs different from the conventional practice of evaluating responses to individual therapy regimens. Next-generation sequencing to assess clonal dynamics longitudinally will improve current radiological assessment of clinical response/resistance and be incorporated into trials exploiting evolution. Furthermore, if understood, clonal evolution can be used to therapeutic advantage, improving patient outcomes based on a new generation of clinical trials.
Collapse
Affiliation(s)
- Alvaro H Ingles Garces
- Drug Development Unit, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, UK
| | - Nuria Porta
- Clinical Trials and Statistical Unit, The Institute of Cancer Research, UK
| | - Trevor A Graham
- Centre for Evolution and Cancer, The Institute of Cancer Research, UK
| | - Udai Banerji
- Drug Development Unit, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, UK.
| |
Collapse
|
21
|
Ge Y, Ye T, Fu S, Jiang X, Song H, Liu B, Wang G, Wang J. Research progress of extracellular vesicles as biomarkers in immunotherapy for non-small cell lung cancer. Front Immunol 2023; 14:1114041. [PMID: 37153619 PMCID: PMC10162406 DOI: 10.3389/fimmu.2023.1114041] [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: 12/02/2022] [Accepted: 04/07/2023] [Indexed: 05/10/2023] Open
Abstract
Lung cancer is one of the most severe forms of malignancy and a leading cause of cancer-related death worldwide, of which non-small cell lung cancer (NSCLC) is the most primary type observed in the clinic. NSCLC is mainly treated with surgery, radiotherapy, and chemotherapy. Additionally, targeted therapy and immunotherapy have also shown promising results. Several immunotherapies, including immune checkpoint inhibitors, have been developed for clinical use and have benefited patients with NSCLC. However, immunotherapy faces several challenges like poor response and unknown effective population. It is essential to identify novel predictive markers to further advance precision immunotherapy for NSCLC. Extracellular vesicles (EVs) present an important research direction. In this review, we focus on the role of EVs as a biomarker in NSCLC immunotherapy considering various perspectives, including the definition and properties of EVs, their role as biomarkers in current NSCLC immunotherapy, and different EV components as biomarkers in NSCLC immunotherapy research. We describe the cross-talk between the role of EVs as biomarkers and novel technical approaches or research concepts in NSCLC immunotherapy, such as neoadjuvants, multi-omics analysis, and the tumour microenvironment. This review will provide a reference for future research to improve the benefits of immunotherapy for patients with NSCLC.
Collapse
Affiliation(s)
- Yang Ge
- Graduate School, Anhui University of Chinese Medicine, Hefei, China
| | - Ting Ye
- Graduate School, Anhui University of Chinese Medicine, Hefei, China
| | - Siyun Fu
- Department of Cellular and Molecular Biology, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Xiaoying Jiang
- Department of Science and Technology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Hang Song
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Bin Liu
- Department of Cellular and Molecular Biology, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
- *Correspondence: Bin Liu, ; Guoquan Wang, ; Jinghui Wang,
| | - Guoquan Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
- *Correspondence: Bin Liu, ; Guoquan Wang, ; Jinghui Wang,
| | - Jinghui Wang
- Department of Cellular and Molecular Biology, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
- *Correspondence: Bin Liu, ; Guoquan Wang, ; Jinghui Wang,
| |
Collapse
|
22
|
Zhang H, Zhang L, Chen S, Yao M, Ma Z, Yuan Y. Yeast-based evolutionary modeling of androgen receptor mutations and natural selection. PLoS Genet 2022; 18:e1010518. [PMID: 36459502 PMCID: PMC9718406 DOI: 10.1371/journal.pgen.1010518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 11/10/2022] [Indexed: 12/05/2022] Open
Abstract
Cancer progression is associated with the evolutionary accumulation of genetic mutations that are biologically significant. Mutations of the androgen receptor (AR) are associated with the development of prostate cancer (PCa) by responding to non-androgenic hormones, and the lack of annotations in their responsiveness to hormone ligands remains a daunting challenge. Here, we have used a yeast reporter system to quickly evaluate the responsiveness of all fifty clinical AR mutations to a variety of steroidal ligands including dihydrotestosterone (DHT), 17β-estradiol (E2), progesterone (PROG), and cyproterone acetate (CPA). Based on an AR-driven reporter that synthesizes histidine, a basic amino acid required for yeast survival and propagation, the yeast reporter system enabling clonal selection was further empowered by combining with a random DNA mutagenesis library to simulate the natural evolution of AR gene under the selective pressures of steroidal ligands. In a time-frame of 1-2 weeks, 19 AR mutants were identified, in which 11 AR mutants were validated for activation by tested steroidal compounds. The high efficiency of our artificial evolution strategy was further evidenced by a sequential selection that enabled the discovery of multipoint AR mutations and evolution directions under the pressure of steroidal ligands. In summary, our designer yeast is a portable reporter module that can be readily adapted to streamline high-throughput AR-compound screening, used as a PCa clinical reference, and combined with additional bioassay systems to further extend its potential.
Collapse
Affiliation(s)
- Haoran Zhang
- Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, People’s Republic of China
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, People’s Republic of China
| | - Lu Zhang
- Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, People’s Republic of China
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, People’s Republic of China
| | - Shaoyong Chen
- Hematology-Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Mingdong Yao
- Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, People’s Republic of China
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, People’s Republic of China
| | - Zhenyi Ma
- School of Basic Medical Sciences, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Yingjin Yuan
- Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, People’s Republic of China
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, People’s Republic of China
- * E-mail:
| |
Collapse
|
23
|
Vishnubalaji R, Shaath H, Al-Alwan M, Abdelalim EM, Alajez NM. Reciprocal interplays between MicroRNAs and pluripotency transcription factors in dictating stemness features in human cancers. Semin Cancer Biol 2022; 87:1-16. [PMID: 36354097 DOI: 10.1016/j.semcancer.2022.10.007] [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: 06/14/2022] [Revised: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022]
Abstract
The interplay between microRNAs (miRNAs) and pluripotency transcription factors (TFs) orchestrates the acquisition of cancer stem cell (CSC) features during the course of malignant transformation, rendering them essential cancer cell dependencies and therapeutic vulnerabilities. In this review, we discuss emerging themes in tumor heterogeneity, including the clonal evolution and the CSC models and their implications in resistance to cancer therapies, and then provide thorough coverage on the roles played by key TFs in maintaining normal and malignant stem cell pluripotency and plasticity. In addition, we discuss the reciprocal interactions between miRNAs and MYC, OCT4, NANOG, SOX2, and KLF4 pluripotency TFs and their contributions to tumorigenesis. We provide our view on the potential to interfere with key miRNA-TF networks through the use of RNA-based therapeutics as single agents or in combination with other therapeutic strategies, to abrogate the CSC state and render tumor cells more responsive to standard and targeted therapies.
Collapse
Affiliation(s)
- Radhakrishnan Vishnubalaji
- Translational Cancer and Immunity Center (TCIC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar
| | - Hibah Shaath
- Translational Cancer and Immunity Center (TCIC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar
| | - Monther Al-Alwan
- Stem Cell and Tissue Re-Engineering Program, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia; College of Medicine, Al-Faisal University, Riyadh 11533, Saudi Arabia
| | - Essam M Abdelalim
- Diabetes Research Center (DRC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, PO Box 34110, Doha, Qatar; College of Health & Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar
| | - Nehad M Alajez
- Translational Cancer and Immunity Center (TCIC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar; College of Health & Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar.
| |
Collapse
|
24
|
Chen S, Xu H, Guo C, Liu Z, Han X. Editorial: The role of multi-omics variants in tumor immunity and immunotherapy. Front Immunol 2022; 13:1098825. [PMID: 36524118 PMCID: PMC9745164 DOI: 10.3389/fimmu.2022.1098825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 11/21/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- Shuang Chen
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China,Center of Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China,Medical School of Zhengzhou University, Zhengzhou, Henan, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China,Interventional Institute of Zhengzhou University, Zhengzhou, Henan, China,Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, China
| | - Chunguang Guo
- Department of Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China,Interventional Institute of Zhengzhou University, Zhengzhou, Henan, China,Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, China,*Correspondence: Xinwei Han, ; Zaoqu Liu,
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China,Interventional Institute of Zhengzhou University, Zhengzhou, Henan, China,Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, China,*Correspondence: Xinwei Han, ; Zaoqu Liu,
| |
Collapse
|
25
|
Habibi M, Taheri G. A new machine learning method for cancer mutation analysis. PLoS Comput Biol 2022; 18:e1010332. [PMID: 36251702 PMCID: PMC9612828 DOI: 10.1371/journal.pcbi.1010332] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 10/27/2022] [Accepted: 10/05/2022] [Indexed: 11/23/2022] Open
Abstract
It is complicated to identify cancer-causing mutations. The recurrence of a mutation in patients remains one of the most reliable features of mutation driver status. However, some mutations are more likely to happen than others for various reasons. Different sequencing analysis has revealed that cancer driver genes operate across complex pathways and networks, with mutations often arising in a mutually exclusive pattern. Genes with low-frequency mutations are understudied as cancer-related genes, especially in the context of networks. Here we propose a machine learning method to study the functionality of mutually exclusive genes in the networks derived from mutation associations, gene-gene interactions, and graph clustering. These networks have indicated critical biological components in the essential pathways, especially those mutated at low frequency. Studying the network and not just the impact of a single gene significantly increases the statistical power of clinical analysis. The proposed method identified important driver genes with different frequencies. We studied the function and the associated pathways in which the candidate driver genes participate. By introducing lower-frequency genes, we recognized less studied cancer-related pathways. We also proposed a novel clustering method to specify driver modules. We evaluated each driver module with different criteria, including the terms of biological processes and the number of simultaneous mutations in each cancer. Materials and implementations are available at: https://github.com/MahnazHabibi/MutationAnalysis.
Collapse
Affiliation(s)
- Mahnaz Habibi
- Department of Mathematics, Qazvin Branch, Islamic Azad University, Qazvin, Iran
| | - Golnaz Taheri
- Department of Electrical Engineering and Computer Science, KTH Royal Institute of Technology, Stockholm, Sweden
- Science for Life Laboratory, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
26
|
Raufaste-Cazavieille V, Santiago R, Droit A. Multi-omics analysis: Paving the path toward achieving precision medicine in cancer treatment and immuno-oncology. Front Mol Biosci 2022; 9:962743. [PMID: 36304921 PMCID: PMC9595279 DOI: 10.3389/fmolb.2022.962743] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 09/21/2022] [Indexed: 11/13/2022] Open
Abstract
The acceleration of large-scale sequencing and the progress in high-throughput computational analyses, defined as omics, was a hallmark for the comprehension of the biological processes in human health and diseases. In cancerology, the omics approach, initiated by genomics and transcriptomics studies, has revealed an incredible complexity with unsuspected molecular diversity within a same tumor type as well as spatial and temporal heterogeneity of tumors. The integration of multiple biological layers of omics studies brought oncology to a new paradigm, from tumor site classification to pan-cancer molecular classification, offering new therapeutic opportunities for precision medicine. In this review, we will provide a comprehensive overview of the latest innovations for multi-omics integration in oncology and summarize the largest multi-omics dataset available for adult and pediatric cancers. We will present multi-omics techniques for characterizing cancer biology and show how multi-omics data can be combined with clinical data for the identification of prognostic and treatment-specific biomarkers, opening the way to personalized therapy. To conclude, we will detail the newest strategies for dissecting the tumor immune environment and host–tumor interaction. We will explore the advances in immunomics and microbiomics for biomarker identification to guide therapeutic decision in immuno-oncology.
Collapse
Affiliation(s)
| | - Raoul Santiago
- CHU de Québec Research Center, Université Laval, Québec, QC, Canada
- Division of Pediatric Hematology-Oncology, Centre Hospitalier Universitaire de L’Université Laval, Charles Bruneau Cancer Center, Québec, QC, Canada
- *Correspondence: Raoul Santiago, ; Arnaud Droit,
| | - Arnaud Droit
- CHU de Québec Research Center, Université Laval, Québec, QC, Canada
- *Correspondence: Raoul Santiago, ; Arnaud Droit,
| |
Collapse
|
27
|
Liu Z, Han Y, Dang Q, Xu H, Zhang Y, Duo M, Lv J, Li H, Kong Y, Han X. Roles of circulating tumor DNA in PD-1/PD-L1 immune checkpoint Inhibitors: Current evidence and future directions. Int Immunopharmacol 2022; 111:109173. [PMID: 35998502 DOI: 10.1016/j.intimp.2022.109173] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/11/2022] [Accepted: 08/14/2022] [Indexed: 12/18/2022]
Abstract
Circulating tumor DNA (ctDNA) sequencing holds considerable promise for early diagnosis and detection of surveillance and minimal residual disease. Blood ctDNA monitors specific cancers by detecting the alterations found in cancer cells, such as apoptosis and necrosis. Due to the short half-life, ctDNA reflects the actual burden of other treatments on tumors. In addition, ctDNA might be preferable to monitor tumor development and treatment compared with invasive tissue biopsy. ctDNA-based liquid biopsy brings remarkable strength to targeted therapy and precision medicine. Notably, multiple ctDNA analysis platforms have been broadly applied in clinical immunotherapy. Through targeted sequencing, early variations in ctDNA could predict response to immune checkpoint inhibitor (ICI). Several studies have demonstrated a correlation between ctDNA kinetics and anti-PD1 antibodies. The need for further research and development remains, although this biomarker holds significant prospects for early cancer detection. This review focuses on describing the basis of ctDNA and its current utilities in oncology and immunotherapy, either for clinical management or early detection, highlighting its advantages and inherent limitations.
Collapse
Affiliation(s)
- Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China; Interventional Institute of Zhengzhou University, Zhengzhou, Henan 450052, China; Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan 450052, China.
| | - Yilin Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Qin Dang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Mengjie Duo
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Jinxiang Lv
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Huanyun Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Ying Kong
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China; Interventional Institute of Zhengzhou University, Zhengzhou, Henan 450052, China; Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan 450052, China.
| |
Collapse
|
28
|
Jiang C, Zhao L, Xin B, Ma G, Wang X, Song S. 18F-FDG PET/CT radiomic analysis for classifying and predicting microvascular invasion in hepatocellular carcinoma and intrahepatic cholangiocarcinoma. Quant Imaging Med Surg 2022; 12:4135-4150. [PMID: 35919043 PMCID: PMC9338369 DOI: 10.21037/qims-21-1167] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 06/06/2022] [Indexed: 12/24/2022]
Abstract
Background Microvascular invasion (MVI) is a critical risk factor for early recurrence of hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC). The aim of this study was to explore the contribution of 18F-fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) radiomic features for the preoperative prediction of HCC and ICC classification and MVI. Methods In this retrospective study, 127 (HCC: ICC =76:51) patients with suspected MVI accompanied by either HCC or ICC were included (In HCC group, MVI positive: negative =46:30 in ICC group, MVI positive: negative =31:20). Results-driven feature engineering workflow was used to select the most predictive feature combinations. The prediction model was based on supervised machine learning classifier. Ten-fold cross validation on training cohort and independent test cohort were constructed to ensure stability and generalization ability of models. Results For HCC and ICC classification, radiomics predictors composed of two PET and one CT feature achieved area under the curve (AUC) of 0.86 (accuracy, sensitivity, specificity was 0.82, 0.78, 0.88, respectively) on test cohort. For MVI prediction, in HCC group, our MVI prediction model achieved AUC of 0.88 (accuracy, sensitivity, specificity was 0.78, 0.88, 0.60 respectively) with three PET features associated with tumor stage on test cohort. In ICC group, the phenotype composed of two PET features and carbohydrate antigen 19-9 (CA19-9) achieved AUC of 0.90 (accuracy, sensitivity, specificity was 0.77, 0.75, 0.80, respectively). Conclusions 18F-FDG PET/CT radiomic features integrating clinical factors have potential in HCC and ICC classification and MVI prediction, while PET features have dominant predictive power in model performance. The prediction model has value in providing a non-invasive biomarker for an earlier indication and comprehensive quantification of primary liver cancers.
Collapse
Affiliation(s)
- Chunjuan Jiang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
| | - Liwei Zhao
- School of Computer Science, The University of Sydney, Sydney, NSW, Australia
| | - Bowen Xin
- School of Computer Science, The University of Sydney, Sydney, NSW, Australia
| | - Guang Ma
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
| | - Xiuying Wang
- School of Computer Science, The University of Sydney, Sydney, NSW, Australia
| | - Shaoli Song
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
| |
Collapse
|
29
|
Mousavi SM, Amin Mahdian SM, Ebrahimi MS, Taghizadieh M, Vosough M, Sadri Nahand J, Hosseindoost S, Vousooghi N, Javar HA, Larijani B, Hadjighassem MR, Rahimian N, Hamblin MR, Mirzaei H. Microfluidics for detection of exosomes and microRNAs in cancer: State of the art. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 28:758-791. [PMID: 35664698 PMCID: PMC9130092 DOI: 10.1016/j.omtn.2022.04.011] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Exosomes are small extracellular vesicles with sizes ranging from 30-150 nanometers that contain proteins, lipids, mRNAs, microRNAs, and double-stranded DNA derived from the cells of origin. Exosomes can be taken up by target cells, acting as a means of cell-to-cell communication. The discovery of these vesicles in body fluids and their participation in cell communication has led to major breakthroughs in diagnosis, prognosis, and treatment of several conditions (e.g., cancer). However, conventional isolation and evaluation of exosomes and their microRNA content suffers from high cost, lengthy processes, difficult standardization, low purity, and poor yield. The emergence of microfluidics devices with increased efficiency in sieving, trapping, and immunological separation of small volumes could provide improved detection and monitoring of exosomes involved in cancer. Microfluidics techniques hold promise for advances in development of diagnostic and prognostic devices. This review covers ongoing research on microfluidics devices for detection of microRNAs and exosomes as biomarkers and their translation to point-of-care and clinical applications.
Collapse
Affiliation(s)
- Seyed Mojtaba Mousavi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mohammad Amin Mahdian
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Saeid Ebrahimi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Taghizadieh
- Department of Pathology, School of Medicine, Center for Women’s Health Research Zahra, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran
| | - Javid Sadri Nahand
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saereh Hosseindoost
- Pain Research Center, Neuroscience Institute, Tehran University of Medical Science, Tehran, Iran
| | - Nasim Vousooghi
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Cognitive and Behavioral Sciences, Tehran University of Medical Sciences, Tehran, Iran
- Iranian National Center for Addiction Studies (INCAS), Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Akbari Javar
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmoud Reza Hadjighassem
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Brain and Spinal Cord Research Center, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Neda Rahimian
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
30
|
Teng J, Zhou K, Lv D, Wu C, Feng H. Case Report: PTEN Mutation Induced by anti-PD-1 Therapy in Stage IV Lung Adenocarcinoma. Front Pharmacol 2022; 13:714408. [PMID: 35677433 PMCID: PMC9168362 DOI: 10.3389/fphar.2022.714408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 05/06/2022] [Indexed: 01/05/2023] Open
Abstract
Lung cancer is the most common solid tumor in the worldwide. Targeted therapy and immunotherapy are important treatment options in advanced non-small cell lung cancer (NSCLC). The association of PTEN mutation and tumor immunotherapy is less established for patients with NSCLC. We present the case of an Asian woman diagnosed with stage IV lung adenocarcinoma harboring an ERBB2 mutation. She received Nivolumab treatment when her disease progresses after previous chemotherapy and Afatinib treatment. However, the patient did not response to Nivolumab. PTEN mutation was detected by next-generation sequencing (NGS) after treatment with Nivolumab. PTEN, a secondary mutation, may be served as a biomarker of resistance to anti-PD-1 immunotherapy in lung adenocarcinoma. The relationship between PTEN mutation and immunotherapy is complex and needs further study.
Collapse
Affiliation(s)
- Junjie Teng
- Cancer Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.,Department of Radiotherapy, The Third Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Kai Zhou
- Department of Radiotherapy, The Third Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Dongxiao Lv
- Department of Radiotherapy, The Third Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Changshun Wu
- Department of Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Hong Feng
- Department of Radiotherapy, The Third Affiliated Hospital of Shandong First Medical University, Jinan, China
| |
Collapse
|
31
|
Khouja HI, Ashankyty IM, Bajrai LH, Kumar PKP, Kamal MA, Firoz A, Mobashir M. Multi-staged gene expression profiling reveals potential genes and the critical pathways in kidney cancer. Sci Rep 2022; 12:7240. [PMID: 35508649 PMCID: PMC9065671 DOI: 10.1038/s41598-022-11143-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 10/11/2021] [Indexed: 02/05/2023] Open
Abstract
Cancer is among the highly complex disease and renal cell carcinoma is the sixth-leading cause of cancer death. In order to understand complex diseases such as cancer, diabetes and kidney diseases, high-throughput data are generated at large scale and it has helped in the research and diagnostic advancement. However, to unravel the meaningful information from such large datasets for comprehensive and minute understanding of cell phenotypes and disease pathophysiology remains a trivial challenge and also the molecular events leading to disease onset and progression are not well understood. With this goal, we have collected gene expression datasets from publicly available dataset which are for two different stages (I and II) for renal cell carcinoma and furthermore, the TCGA and cBioPortal database have been utilized for clinical relevance understanding. In this work, we have applied computational approach to unravel the differentially expressed genes, their networks for the enriched pathways. Based on our results, we conclude that among the most dominantly altered pathways for renal cell carcinoma, are PI3K-Akt, Foxo, endocytosis, MAPK, Tight junction, cytokine-cytokine receptor interaction pathways and the major source of alteration for these pathways are MAP3K13, CHAF1A, FDX1, ARHGAP26, ITGBL1, C10orf118, MTO1, LAMP2, STAMBP, DLC1, NSMAF, YY1, TPGS2, SCARB2, PRSS23, SYNJ1, CNPPD1, PPP2R5E. In terms of clinical significance, there are large number of differentially expressed genes which appears to be playing critical roles in survival.
Collapse
Affiliation(s)
- Hamed Ishaq Khouja
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Ibraheem Mohammed Ashankyty
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Leena Hussein Bajrai
- Special Infectious Agents Unit-BSL3, King Fahad Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Biochemistry Department, Sciences College, King Abdulaziz University, Jeddah, Saudi Arabia
| | - P K Praveen Kumar
- Department of Biotechnology, Sri Venkateswara College of Engineering, Sriperumbudur, 602105, India
| | - Mohammad Amjad Kamal
- West China School of Nursing/Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- King Fahd Medical Research Center, King Abdulaziz University, P. O. Box 80216, Jeddah, 21589, Saudi Arabia
- Enzymoics, Novel Global Community Educational Foundation, 7 Peterlee Place, Hebersham, NSW, 2770, Australia
| | - Ahmad Firoz
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Mohammad Mobashir
- SciLifeLab, Department of Oncology and Pathology, Karolinska Institutet, Box 1031, 171 21, Stockholm, Sweden.
| |
Collapse
|
32
|
Clinical Impact of High Throughput Sequencing on Liquid Biopsy in Advanced Solid Cancer. Curr Oncol 2022; 29:1902-1918. [PMID: 35323355 PMCID: PMC8947301 DOI: 10.3390/curroncol29030155] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/03/2022] [Accepted: 03/08/2022] [Indexed: 12/31/2022] Open
Abstract
Background: Cancer therapies targeting actionable molecular alterations (AMA) have developed, but the clinical routine impact of high-throughput molecular profiling remains unclear. We present a monocentric experience of molecular profiling based on liquid biopsy in patients with cancer. Methods: Patients included had solid cancer and underwent cfDNA genomic profiling with FoudationOne Liquid CDx (F1LCDx) test, analyzing 324 genes. Primary endpoint was to describe patients with an AMA for whom clinical decisions were impacted by F1LCDx test results. Results: 191 patients were included, mostly with lung cancer (46%). An AMA was found in 52%. The most common molecular alterations were: TP53 (52%), KRAS (14%) and DNMT3 (11%). The most common AMA were: CHEK2 (10%), PIK3CA (9%), ATM (7%). There was no difference in progression-free survival (2.66 months vs. 3.81 months, p = 0.17), overall survival (5.3 months vs. 7.1 months, p = 0.64), or PFS2/PFS1 ratio ≥ 1.3 (20% vs. 24%, p = 0.72) between patients receiving a molecularly matched therapy (MMT) or a non-MMT, respectively. Patients with a MMT had an overall response rate of 19% and a disease control of 32%. Conclusions: Routine cfDNA molecular profiling is feasible and can lead to the access of targeted therapies. However, no notable benefit in patient’s outcomes was shown in this unselected pan-cancer study.
Collapse
|
33
|
Mota A, Oltra SS, Selenica P, Moiola CP, Casas-Arozamena C, López-Gil C, Diaz E, Gatius S, Ruiz-Miro M, Calvo A, Rojo-Sebastián A, Hurtado P, Piñeiro R, Colas E, Gil-Moreno A, Reis-Filho JS, Muinelo-Romay L, Abal M, Matias-Guiu X, Weigelt B, Moreno-Bueno G. Intratumor genetic heterogeneity and clonal evolution to decode endometrial cancer progression. Oncogene 2022; 41:1835-1850. [PMID: 35145232 PMCID: PMC8956509 DOI: 10.1038/s41388-022-02221-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 01/12/2022] [Accepted: 01/27/2022] [Indexed: 12/12/2022]
Abstract
Analyzing different tumor regions by next generation sequencing allows the assessment of intratumor genetic heterogeneity (ITGH), a phenomenon that has been studied widely in some tumor types but has been less well explored in endometrial carcinoma (EC). In this study, we sought to characterize the spatial and temporal heterogeneity of 9 different ECs using whole-exome sequencing, and by performing targeted sequencing validation of the 42 primary tumor regions and 30 metastatic samples analyzed. In addition, copy number alterations of serous carcinomas were assessed by comparative genomic hybridization arrays. From the somatic mutations, identified by whole-exome sequencing, 532 were validated by targeted sequencing. Based on these data, the phylogenetic tree reconstructed for each case allowed us to establish the tumors’ evolution and correlate this to tumor progression, prognosis, and the presence of recurrent disease. Moreover, we studied the genetic landscape of an ambiguous EC and the molecular profile obtained was used to guide the selection of a potential personalized therapy for this patient, which was subsequently validated by preclinical testing in patient-derived xenograft models. Overall, our study reveals the impact of analyzing different tumor regions to decipher the ITGH in ECs, which could help make the best treatment decision.
Collapse
Affiliation(s)
- Alba Mota
- MD Anderson International Foundation, 28033, Madrid, Spain.,Biochemistry Department, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas 'Alberto Sols' (CSIC-UAM), IdiPaz, 28029, Madrid, Spain
| | - Sara S Oltra
- MD Anderson International Foundation, 28033, Madrid, Spain.,Biochemistry Department, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas 'Alberto Sols' (CSIC-UAM), IdiPaz, 28029, Madrid, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029, Madrid, Spain
| | - Pier Selenica
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Cristian P Moiola
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029, Madrid, Spain.,Biomedical Research Group in Gynecology, Vall Hebron Institute of Research, Universitat Autònoma de Barcelona, 08035, Barcelona, Spain
| | - Carlos Casas-Arozamena
- Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago de Compostela (IDIS), University Hospital of Santiago de Compostela (SERGAS), Trav. Choupana s/n, 15706, Santiago de Compostela, Spain
| | - Carlos López-Gil
- Biomedical Research Group in Gynecology, Vall Hebron Institute of Research, Universitat Autònoma de Barcelona, 08035, Barcelona, Spain
| | - Eva Diaz
- MD Anderson International Foundation, 28033, Madrid, Spain
| | - Sonia Gatius
- Department of Pathology, Hospital U Arnau de Vilanova, University of Lleida, IRBLLEIDA, Lleida, Spain
| | | | - Ana Calvo
- Department of Gynecology, Hospital U Arnau de Vilanova, IRBLLEIDA, Lleida, Spain
| | - Alejandro Rojo-Sebastián
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029, Madrid, Spain.,Biomedical Research Group in Gynecology, Vall Hebron Institute of Research, Universitat Autònoma de Barcelona, 08035, Barcelona, Spain.,MD Anderson Cancer Center, Madrid, Spain
| | - Pablo Hurtado
- Roche-Chus Joint Unit, Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago de Compostela (IDIS), Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
| | - Roberto Piñeiro
- Roche-Chus Joint Unit, Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago de Compostela (IDIS), Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
| | - Eva Colas
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029, Madrid, Spain.,Biomedical Research Group in Gynecology, Vall Hebron Institute of Research, Universitat Autònoma de Barcelona, 08035, Barcelona, Spain
| | - Antonio Gil-Moreno
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029, Madrid, Spain.,Biomedical Research Group in Gynecology, Vall Hebron Institute of Research, Universitat Autònoma de Barcelona, 08035, Barcelona, Spain.,Gynaecological Department, Vall Hebron University Hospital, 08035, Barcelona, Spain
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Laura Muinelo-Romay
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029, Madrid, Spain.,Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago de Compostela (IDIS), University Hospital of Santiago de Compostela (SERGAS), Trav. Choupana s/n, 15706, Santiago de Compostela, Spain
| | - Miguel Abal
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029, Madrid, Spain.,Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago de Compostela (IDIS), University Hospital of Santiago de Compostela (SERGAS), Trav. Choupana s/n, 15706, Santiago de Compostela, Spain
| | - Xavier Matias-Guiu
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029, Madrid, Spain.,Department of Pathology, Hospital U Arnau de Vilanova, University of Lleida, IRBLLEIDA, Lleida, Spain.,Departments of Pathology, Hospital U. de Bellvitge, Universities of Lleida and Barcelona, IDIBELL Lleida and Barcelona, Spain
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Gema Moreno-Bueno
- MD Anderson International Foundation, 28033, Madrid, Spain. .,Biochemistry Department, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas 'Alberto Sols' (CSIC-UAM), IdiPaz, 28029, Madrid, Spain. .,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029, Madrid, Spain.
| |
Collapse
|
34
|
Kashyap A, Rapsomaniki MA, Barros V, Fomitcheva-Khartchenko A, Martinelli AL, Rodriguez AF, Gabrani M, Rosen-Zvi M, Kaigala G. Quantification of tumor heterogeneity: from data acquisition to metric generation. Trends Biotechnol 2021; 40:647-676. [PMID: 34972597 DOI: 10.1016/j.tibtech.2021.11.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/26/2021] [Accepted: 11/29/2021] [Indexed: 01/18/2023]
Abstract
Tumors are unique and complex ecosystems, in which heterogeneous cell subpopulations with variable molecular profiles, aggressiveness, and proliferation potential coexist and interact. Understanding how heterogeneity influences tumor progression has important clinical implications for improving diagnosis, prognosis, and treatment response prediction. Several recent innovations in data acquisition methods and computational metrics have enabled the quantification of spatiotemporal heterogeneity across different scales of tumor organization. Here, we summarize the most promising efforts from a common experimental and computational perspective, discussing their advantages, shortcomings, and challenges. With personalized medicine entering a new era of unprecedented opportunities, our vision is that of future workflows integrating across modalities, scales, and dimensions to capture intricate aspects of the tumor ecosystem and to open new avenues for improved patient care.
Collapse
Affiliation(s)
- Aditya Kashyap
- IBM Research Europe -Säumerstrasse 4, Rüschlikon CH-8803, Zurich, Switzerland
| | | | - Vesna Barros
- Department of Healthcare Informatics, IBM Research, IBM R&D Labs, University of Haifa Campus, Mount Carmel, Haifa, 3498825, Israel; The Hebrew University, The Edmond J. Safra Campus - Givat Ram, Jerusalem, 9190401, Israel
| | - Anna Fomitcheva-Khartchenko
- IBM Research Europe -Säumerstrasse 4, Rüschlikon CH-8803, Zurich, Switzerland; Eidgenössische Technische Hochschule (ETH-Zurich), Vladimir-Prelog-Weg 1-5/10, 8099 Zurich, Switzerland
| | | | | | - Maria Gabrani
- IBM Research Europe -Säumerstrasse 4, Rüschlikon CH-8803, Zurich, Switzerland
| | - Michal Rosen-Zvi
- Department of Healthcare Informatics, IBM Research, IBM R&D Labs, University of Haifa Campus, Mount Carmel, Haifa, 3498825, Israel; The Hebrew University, The Edmond J. Safra Campus - Givat Ram, Jerusalem, 9190401, Israel
| | - Govind Kaigala
- IBM Research Europe -Säumerstrasse 4, Rüschlikon CH-8803, Zurich, Switzerland.
| |
Collapse
|
35
|
Chen C, Wang T, Yang M, Song J, Huang M, Bai Y, Su H. Genomic Profiling of Blood-Derived Circulating Tumor DNA from Patients with Advanced Biliary Tract Cancer. Pathol Oncol Res 2021; 27:1609879. [PMID: 34720757 PMCID: PMC8553707 DOI: 10.3389/pore.2021.1609879] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 09/30/2021] [Indexed: 01/10/2023]
Abstract
Background: Biliary tract cancer is a highly lethal malignancy with poor clinical outcome. Accumulating evidence indicates targeted therapeutics may provide new hope for improving treatment response in BTC, hence better understanding the genomic profile is particularly important. Since tumor tissue may not be available for some patients, a complementary method is urgently needed. Circulating tumor DNA (ctDNA) provides a noninvasive means for detecting genomic alterations, and has been regarded as a promising tool to guide clinical therapies. Methods: Next-generation sequencing of 150 cancer-related genes was used to detect gene alterations in blood-derived ctDNA from 154 Chinese patients with BTC. Genomic alterations were analyzed and compared with an internal tissue genomic database and TCGA database. Results: 94.8% patients had at least one change detected in their ctDNA. The median maximum somatic allele frequency was 6.47% (ranging 0.1-34.8%). TP53 and KRAS were the most often mutated genes. The frequencies of single nucleotide variation in commonly mutated genes in ctDNA were similar to those detected in tissue samples, TP53 (35.1 vs. 40.4%) and KRAS (20.1 vs. 22.6%). Pathway analysis revealed that mutated genes were mapped to several key pathways including PI3K-Akt, p53, ErbB and Ras signaling pathway. In addition, patients harboring LRP1B, TP53, and ErbB family mutations presented significantly higher tumor mutation burden. Conclusions: These findings demonstrated that ctDNA testing by NGS was feasible in revealing genomic changes and could be a viable alternative to tissue biopsy in patients with metastatic BTC.
Collapse
Affiliation(s)
- Chen Chen
- Hepatobiliary Surgery, Hunan Provincial People’s Hospital (The First Affiliate Hospital of Hunan Normal University), Changsha, China
| | - Tao Wang
- Hepatobiliary Surgery, Hunan Provincial People’s Hospital (The First Affiliate Hospital of Hunan Normal University), Changsha, China
| | - Mengmei Yang
- The Medical Department, 3D Medicines Inc., Shanghai, China
| | - Jia Song
- The Medical Department, 3D Medicines Inc., Shanghai, China
| | - Mengli Huang
- The Medical Department, 3D Medicines Inc., Shanghai, China
| | - Yuezong Bai
- The Medical Department, 3D Medicines Inc., Shanghai, China
| | - Hao Su
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
36
|
Gong XQ, Tao YY, Wu YK, Liu N, Yu X, Wang R, Zheng J, Liu N, Huang XH, Li JD, Yang G, Wei XQ, Yang L, Zhang XM. Progress of MRI Radiomics in Hepatocellular Carcinoma. Front Oncol 2021; 11:698373. [PMID: 34616673 PMCID: PMC8488263 DOI: 10.3389/fonc.2021.698373] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 08/31/2021] [Indexed: 02/05/2023] Open
Abstract
Background Hepatocellular carcinoma (HCC) is the sixth most common cancer in the world and the third leading cause of cancer-related death. Although the diagnostic scheme of HCC is currently undergoing refinement, the prognosis of HCC is still not satisfactory. In addition to certain factors, such as tumor size and number and vascular invasion displayed on traditional imaging, some histopathological features and gene expression parameters are also important for the prognosis of HCC patients. However, most parameters are based on postoperative pathological examinations, which cannot help with preoperative decision-making. As a new field, radiomics extracts high-throughput imaging data from different types of images to build models and predict clinical outcomes noninvasively before surgery, rendering it a powerful aid for making personalized treatment decisions preoperatively. Objective This study reviewed the workflow of radiomics and the research progress on magnetic resonance imaging (MRI) radiomics in the diagnosis and treatment of HCC. Methods A literature review was conducted by searching PubMed for search of relevant peer-reviewed articles published from May 2017 to June 2021.The search keywords included HCC, MRI, radiomics, deep learning, artificial intelligence, machine learning, neural network, texture analysis, diagnosis, histopathology, microvascular invasion, surgical resection, radiofrequency, recurrence, relapse, transarterial chemoembolization, targeted therapy, immunotherapy, therapeutic response, and prognosis. Results Radiomics features on MRI can be used as biomarkers to determine the differential diagnosis, histological grade, microvascular invasion status, gene expression status, local and systemic therapeutic responses, and prognosis of HCC patients. Conclusion Radiomics is a promising new imaging method. MRI radiomics has high application value in the diagnosis and treatment of HCC.
Collapse
Affiliation(s)
- Xue-Qin Gong
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Medical Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yun-Yun Tao
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Medical Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yao-Kun Wu
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Medical Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Ning Liu
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Medical Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xi Yu
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Medical Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Ran Wang
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Medical Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Jing Zheng
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Medical Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Nian Liu
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Medical Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xiao-Hua Huang
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Medical Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Jing-Dong Li
- Department of Hepatocellular Surgery, Institute of Hepato-Biliary-Intestinal Disease, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Gang Yang
- Department of Hepatocellular Surgery, Institute of Hepato-Biliary-Intestinal Disease, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xiao-Qin Wei
- School of Medical Imaging, North Sichuan Medical College, Nanchong, China
| | - Lin Yang
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Medical Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xiao-Ming Zhang
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Medical Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| |
Collapse
|
37
|
Wu W, Liu Y, Zeng S, Han Y, Shen H. Intratumor heterogeneity: the hidden barrier to immunotherapy against MSI tumors from the perspective of IFN-γ signaling and tumor-infiltrating lymphocytes. J Hematol Oncol 2021; 14:160. [PMID: 34620200 PMCID: PMC8499512 DOI: 10.1186/s13045-021-01166-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 09/07/2021] [Indexed: 12/15/2022] Open
Abstract
In this era of precision medicine, with the help of biomarkers, immunotherapy has significantly improved prognosis of many patients with malignant tumor. Deficient mismatch repair (dMMR)/microsatellite instability (MSI) status is used as a biomarker in clinical practice to predict favorable response to immunotherapy and prognosis. MSI is an important characteristic which facilitates mutation and improves the likelihood of a favorable response to immunotherapy. However, many patients with dMMR/MSI still respond poorly to immunotherapies, which partly results from intratumor heterogeneity propelled by dMMR/MSI. In this review, we discuss how dMMR/MSI facilitates mutations in tumor cells and generates intratumor heterogeneity, especially through type II interferon (IFN-γ) signaling and tumor-infiltrating lymphocytes (TILs). We discuss the mechanism of immunotherapy from the perspective of dMMR/MSI, molecular pathways and TILs, and we discuss how intratumor heterogeneity hinders the therapeutic effect of immunotherapy. Finally, we summarize present techniques and strategies to look at the tumor as a whole to design personalized regimes and achieve favorable prognosis.
Collapse
Affiliation(s)
- Wantao Wu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China, 410008
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China, 410008
| | - Yihan Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China, 410008
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China, 410008
| | - Shan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China, 410008.
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China, 410008.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China.
| | - Ying Han
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China, 410008.
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China, 410008.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China.
| | - Hong Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China, 410008.
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China, 410008.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China.
| |
Collapse
|
38
|
Vendramin R, Litchfield K, Swanton C. Cancer evolution: Darwin and beyond. EMBO J 2021; 40:e108389. [PMID: 34459009 PMCID: PMC8441388 DOI: 10.15252/embj.2021108389] [Citation(s) in RCA: 120] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/04/2021] [Accepted: 06/25/2021] [Indexed: 12/16/2022] Open
Abstract
Clinical and laboratory studies over recent decades have established branched evolution as a feature of cancer. However, while grounded in somatic selection, several lines of evidence suggest a Darwinian model alone is insufficient to fully explain cancer evolution. First, the role of macroevolutionary events in tumour initiation and progression contradicts Darwin's central thesis of gradualism. Whole-genome doubling, chromosomal chromoplexy and chromothripsis represent examples of single catastrophic events which can drive tumour evolution. Second, neutral evolution can play a role in some tumours, indicating that selection is not always driving evolution. Third, increasing appreciation of the role of the ageing soma has led to recent generalised theories of age-dependent carcinogenesis. Here, we review these concepts and others, which collectively argue for a model of cancer evolution which extends beyond Darwin. We also highlight clinical opportunities which can be grasped through targeting cancer vulnerabilities arising from non-Darwinian patterns of evolution.
Collapse
Affiliation(s)
- Roberto Vendramin
- Cancer Research UK Lung Cancer Centre of ExcellenceUniversity College London Cancer InstituteLondonUK
| | - Kevin Litchfield
- Cancer Research UK Lung Cancer Centre of ExcellenceUniversity College London Cancer InstituteLondonUK
| | - Charles Swanton
- Cancer Research UK Lung Cancer Centre of ExcellenceUniversity College London Cancer InstituteLondonUK
- Cancer Evolution and Genome Instability LaboratoryThe Francis Crick InstituteLondonUK
| |
Collapse
|
39
|
Li S, Ke L, Meng X, Zhou H, Zhang X, Wu H, Yu J, Zhang H. Next Generation Sequencing in the Management of Leptomeningeal Metastases of Non-Small Cell Lung Cancer: A Case Report and Literature Review. Recent Pat Anticancer Drug Discov 2021; 16:108-116. [PMID: 33245275 DOI: 10.2174/1574892815666201127114224] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/15/2020] [Accepted: 11/10/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Diagnosis of Leptomeningeal Metastases (LM) from Non-Small Cell Lung Cancer (NSCLC) is usually based on clinical symptoms, Cerebral-Spinal Fluid (CSF) cytology, and neuro-imaging. However, early diagnosis of LM in NSCLC is challenging due to the low sensitivity of these approaches. The Next-Generation Sequencing (NGS) using CSF could help improve the diagnosis of LM and guide its treatment options. CASE PRESENTATION We report a 39-year-old male NSCLC patient with negative molecular testing results in the lung cancer tissue sample. The patient developed symptoms of LM with the negative CSF cytology and MRI; however, the NGS analysis of CSF revealed an EGFR exon 19 del mutation. The patient attained 6 months of Progression-Free Survival (PFS) by treating with erlotinib and anlotinib before the neurological symptoms appeared again. EGFR Thr790Met was positive in the CSF but negative in his plasma. The patient was then treated with osimertinib therapy and the response was maintained for more than 1 year. RESULTS & DISCUSSION This case is the first study reporting the clinical benefit of using the combination of erlotinib and anlotinib for the treatment of LM with the EGFR 19 del, osimertinib with EGFR T790M mutation in CSF, but negative gene mutation in the blood or lung tumor biopsy specimens. Our results support that genetic analysis should be performed with CSF samples in all cases of suspected LM when the results of testing for EGFR/ALK/ROS1 mutation in blood samples or tumor biopsy specimens are negative, as these patients could benefit from treatment of TKIs in a poor prognostic setting. CONCLUSION In parallel to current patents, NGS could be applied as a novel strategy in the managing of NSCLC patients with LM.
Collapse
Affiliation(s)
- Shuo Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Linping Ke
- Department of Clinical Medicine, Weifang Medical University, Weifang, Shandong, China
| | - Xue Meng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Haiyan Zhou
- Department of Medical Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Xiqin Zhang
- Department of Medical Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Huaguo Wu
- Department of Head and Neck Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Jinming Yu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Hui Zhang
- Department of Medical Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Science, Jinan, Shandong, China
| |
Collapse
|
40
|
Abstract
BACKGROUND Radiomics is an emerging field that extracts high-throughput imaging data from different types of images to build models and predict clinical outcomes ina non-invasive manner. This field currently is in the initial growth phase and lacks standardized evaluation criteria but remains a very promising tool for the future todevelop suitable biomarkers for diagnosis, prognosis, and treatment response assessments. The analysis of hepatocellular carcinoma by radiomics will contribute toearly diagnosis and treatment of tumors and improve survival and cure rates. AIM Herein, we aimed to provide an up-to-date overview of the principles of radiomics specifically regarding hepatocellular carcinoma (HCC) and discuss the current challenges and future advancements of radiomics for HCC.
Collapse
Affiliation(s)
- Aysegul Sagir Kahraman
- Department of Radiology, Turgut Ozal Medical Center, Inonu University School of Medicine, 244280, Malatya, Turkey.
- Liver Transplantation Institute, Inonu University, 244280, Malatya, Turkey.
| |
Collapse
|
41
|
Dentro SC, Leshchiner I, Haase K, Tarabichi M, Wintersinger J, Deshwar AG, Yu K, Rubanova Y, Macintyre G, Demeulemeester J, Vázquez-García I, Kleinheinz K, Livitz DG, Malikic S, Donmez N, Sengupta S, Anur P, Jolly C, Cmero M, Rosebrock D, Schumacher SE, Fan Y, Fittall M, Drews RM, Yao X, Watkins TBK, Lee J, Schlesner M, Zhu H, Adams DJ, McGranahan N, Swanton C, Getz G, Boutros PC, Imielinski M, Beroukhim R, Sahinalp SC, Ji Y, Peifer M, Martincorena I, Markowetz F, Mustonen V, Yuan K, Gerstung M, Spellman PT, Wang W, Morris QD, Wedge DC, Van Loo P. Characterizing genetic intra-tumor heterogeneity across 2,658 human cancer genomes. Cell 2021; 184:2239-2254.e39. [PMID: 33831375 PMCID: PMC8054914 DOI: 10.1016/j.cell.2021.03.009] [Citation(s) in RCA: 256] [Impact Index Per Article: 85.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 09/21/2020] [Accepted: 03/03/2021] [Indexed: 02/07/2023]
Abstract
Intra-tumor heterogeneity (ITH) is a mechanism of therapeutic resistance and therefore an important clinical challenge. However, the extent, origin, and drivers of ITH across cancer types are poorly understood. To address this, we extensively characterize ITH across whole-genome sequences of 2,658 cancer samples spanning 38 cancer types. Nearly all informative samples (95.1%) contain evidence of distinct subclonal expansions with frequent branching relationships between subclones. We observe positive selection of subclonal driver mutations across most cancer types and identify cancer type-specific subclonal patterns of driver gene mutations, fusions, structural variants, and copy number alterations as well as dynamic changes in mutational processes between subclonal expansions. Our results underline the importance of ITH and its drivers in tumor evolution and provide a pan-cancer resource of comprehensively annotated subclonal events from whole-genome sequencing data.
Collapse
Affiliation(s)
- Stefan C Dentro
- Cancer Genomics Laboratory, The Francis Crick Institute, London NW1 1AT, UK; Wellcome Trust Sanger Institute, Cambridge CB10 1SA, UK; Big Data Institute, University of Oxford, Oxford OX3 7LF, UK
| | | | - Kerstin Haase
- Cancer Genomics Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Maxime Tarabichi
- Cancer Genomics Laboratory, The Francis Crick Institute, London NW1 1AT, UK; Wellcome Trust Sanger Institute, Cambridge CB10 1SA, UK
| | - Jeff Wintersinger
- University of Toronto, Toronto, ON M5S 3E1, Canada; Vector Institute, Toronto, ON M5G 1L7, Canada
| | - Amit G Deshwar
- University of Toronto, Toronto, ON M5S 3E1, Canada; Vector Institute, Toronto, ON M5G 1L7, Canada
| | - Kaixian Yu
- The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yulia Rubanova
- University of Toronto, Toronto, ON M5S 3E1, Canada; Vector Institute, Toronto, ON M5G 1L7, Canada
| | - Geoff Macintyre
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | - Jonas Demeulemeester
- Cancer Genomics Laboratory, The Francis Crick Institute, London NW1 1AT, UK; Department of Human Genetics, University of Leuven, 3000 Leuven, Belgium
| | - Ignacio Vázquez-García
- Wellcome Trust Sanger Institute, Cambridge CB10 1SA, UK; University of Cambridge, Cambridge CB2 0QQ, UK; Computational Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Irving Institute for Cancer Dynamics, Columbia University, New York, NY 10027, USA
| | - Kortine Kleinheinz
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Heidelberg University, 69120 Heidelberg, Germany
| | | | - Salem Malikic
- Cancer Data Science Laboratory, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Nilgun Donmez
- Simon Fraser University, Burnaby, BC V5A 1S6, Canada; Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
| | | | - Pavana Anur
- Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR 97231, USA
| | - Clemency Jolly
- Cancer Genomics Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Marek Cmero
- University of Melbourne, Melbourne, VIC 3010, Australia; Walter + Eliza Hall Institute, Melbourne, VIC 3000, Australia
| | | | | | - Yu Fan
- The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Matthew Fittall
- Cancer Genomics Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Ruben M Drews
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | - Xiaotong Yao
- Weill Cornell Medicine, New York, NY 10065, USA; New York Genome Center, New York, NY 10013, USA
| | - Thomas B K Watkins
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Juhee Lee
- University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | | | - Hongtu Zhu
- The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - David J Adams
- Wellcome Trust Sanger Institute, Cambridge CB10 1SA, UK
| | - Nicholas McGranahan
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London WC1E 6BT, UK; Cancer Genome Evolution Research Group, University College London Cancer Institute, London WC1E 6DD, UK
| | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London NW1 1AT, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London WC1E 6BT, UK; Department of Medical Oncology, University College London Hospitals, London NW1 2BU, UK
| | - Gad Getz
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Massachusetts General Hospital Center for Cancer Research, Charlestown, MA 02129, USA; Massachusetts General Hospital, Department of Pathology, Boston, MA 02114, USA; Harvard Medical School, Boston, MA 02215, USA
| | - Paul C Boutros
- University of Toronto, Toronto, ON M5S 3E1, Canada; Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada; University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Marcin Imielinski
- Weill Cornell Medicine, New York, NY 10065, USA; New York Genome Center, New York, NY 10013, USA
| | - Rameen Beroukhim
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - S Cenk Sahinalp
- Cancer Data Science Laboratory, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Yuan Ji
- NorthShore University HealthSystem, Evanston, IL 60201, USA; The University of Chicago, Chicago, IL 60637, USA
| | - Martin Peifer
- Department of Translational Genomics, Center for Integrated Oncology Cologne-Bonn, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | | | - Florian Markowetz
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | - Ville Mustonen
- Organismal and Evolutionary Biology Research Programme, Department of Computer Science, Institute of Biotechnology, University of Helsinki, 00014 Helsinki, Finland
| | - Ke Yuan
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK; School of Computing Science, University of Glasgow, Glasgow G12 8RZ, UK
| | - Moritz Gerstung
- Wellcome Trust Sanger Institute, Cambridge CB10 1SA, UK; European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge CB10 1SD, UK; European Molecular Biology Laboratory, Genome Biology Unit, 69117 Heidelberg, Germany
| | - Paul T Spellman
- Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR 97231, USA
| | - Wenyi Wang
- The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Quaid D Morris
- University of Toronto, Toronto, ON M5S 3E1, Canada; Vector Institute, Toronto, ON M5G 1L7, Canada; Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada; Computational and Systems Biology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - David C Wedge
- Big Data Institute, University of Oxford, Oxford OX3 7LF, UK; Oxford NIHR Biomedical Research Centre, Oxford OX4 2PG, UK; Manchester Cancer Research Centre, University of Manchester, Manchester M20 4GJ, UK
| | - Peter Van Loo
- Cancer Genomics Laboratory, The Francis Crick Institute, London NW1 1AT, UK.
| |
Collapse
|
42
|
Leers MPG. Circulating tumor DNA and their added value in molecular oncology. Clin Chem Lab Med 2021; 58:152-161. [PMID: 31490771 DOI: 10.1515/cclm-2019-0436] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 08/06/2019] [Indexed: 12/14/2022]
Abstract
New methods for molecular diagnosis are now available in oncology thanks to the discovery of circulating tumor DNA molecules in the plasma of cancer patients. By utilizing blood samples, rather than traditional tissue sampling, clinical practice is on the verge of new discoveries from the analysis of cell-free DNA (cfDNA). The method, known as a "liquid biopsy", consists of analyzing therapeutic targets and drug-resistant conferring gene mutations in circulating tumor cells (CTC) and cell-free circulating tumor DNA (ctDNA). These are subsequently released from primary tumors and metastatic deposits into the peripheral blood. The advantages of the method can be observed in the diagnosis, but also in the choice of treatment for solid tumors (e.g. non-small cell lung carcinomas [NSCLC]). In order to interpret the results, an understanding of the biological characteristics of circulating tumor DNA is required. Currently there is no consensus as to how a liquid biopsy should be conducted. In this review, we will assess the pros of ctDNA as analytes in peripheral blood samples and its impact on clinical applications in solid tumors and hematological malignancies. We will also address practical issues facing clinical implementation, such as pre-analytical factors. Moreover, we will emphasize the open questions that remain when considering the current state of personalized medicine and targeted therapy.
Collapse
Affiliation(s)
- Math P G Leers
- Department of Clinical Chemistry and Hematology, Zuyderland Medical Center Sittard-Geleen, Dr. H. Van der Hoffplein 1, P.O. Box 5500, 6130 MB Sittard, The Netherlands
| |
Collapse
|
43
|
Rodríguez J, Avila J, Rolfo C, Ruíz-Patiño A, Russo A, Ricaurte L, Ordóñez-Reyes C, Arrieta O, Zatarain-Barrón ZL, Recondo G, Cardona AF. When Tissue is an Issue the Liquid Biopsy is Nonissue: A Review. Oncol Ther 2021; 9:89-110. [PMID: 33689160 PMCID: PMC8140006 DOI: 10.1007/s40487-021-00144-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 02/11/2021] [Indexed: 02/07/2023] Open
Abstract
Precision medicine has impacted the field of medical oncology by introducing personalized therapies, improving all measurable outcomes. This field, in turn, has expanded to obtaining and analyzing a vast and ever-increasing amount of genomic information. One technique currently applied is the liquid biopsy, which consists of detecting and isolating DNA and exosomes in cancer patients. Newly developed techniques have made it possible to use the liquid biopsy in a wide range of settings. However, challenges regarding the validation of its clinical utility exist because of a lack of standardization across different techniques and tumor types, confounder genomic information, lack of appropriate clinical trial designs, and a non-measured, and therefore not estimated, economic impact on population health. Nowadays, liquid biopsy is not routinely used, but ongoing research is increasing its popularity, and a new era in oncology is developing. Therefore, it is essential to have an in-depth understanding of the liquid biopsy technique. In this review, we summarize the leading techniques and liquid biopsy applications in cancer.
Collapse
Affiliation(s)
- July Rodríguez
- Foundation for Clinical and Applied Cancer Research (FICMAC), Bogota, Colombia
- Molecular Oncology and Biology Systems Research Group (Fox-G/ONCOLGroup), Universidad El Bosque, Bogota, Colombia
| | - Jenny Avila
- Foundation for Clinical and Applied Cancer Research (FICMAC), Bogota, Colombia
- Molecular Oncology and Biology Systems Research Group (Fox-G/ONCOLGroup), Universidad El Bosque, Bogota, Colombia
| | - Christian Rolfo
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alejandro Ruíz-Patiño
- Foundation for Clinical and Applied Cancer Research (FICMAC), Bogota, Colombia
- Molecular Oncology and Biology Systems Research Group (Fox-G/ONCOLGroup), Universidad El Bosque, Bogota, Colombia
| | - Alessandro Russo
- Medical Oncology Unit A.O. Papardo and Department of Human Pathology, University of Messina, Messina, Italy
| | - Luisa Ricaurte
- Foundation for Clinical and Applied Cancer Research (FICMAC), Bogota, Colombia
- Pathology Department, Mayo Clinic, Rochester, MN, USA
| | | | - Oscar Arrieta
- Thoracic Oncology Unit, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
| | | | - Gonzalo Recondo
- Thoracic Oncology Section, Centro de Educación Médica e Investigaciones Clínicas (CEMIC), Buenos Aires, Argentina
| | - Andrés F Cardona
- Foundation for Clinical and Applied Cancer Research (FICMAC), Bogota, Colombia.
- Molecular Oncology and Biology Systems Research Group (Fox-G/ONCOLGroup), Universidad El Bosque, Bogota, Colombia.
- Clinical and Traslational Oncology Group, Clinica del Country, Bogota, Colombia.
| |
Collapse
|
44
|
Moon SW, Son HJ, Mo HY, Choi EJ, Yoo NJ, Lee SH. Mutation and expression alterations of histone methylation-related NSD2, KDM2B and SETMAR genes in colon cancers. Pathol Res Pract 2021; 219:153354. [PMID: 33621919 DOI: 10.1016/j.prp.2021.153354] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/20/2021] [Accepted: 01/25/2021] [Indexed: 12/22/2022]
Abstract
Epigenetic dysregulation is a hallmark of cancers, and examples of its cancer-associated expression and mutation alterations are rapidly growing. Histone methylation, a process by which methyl groups are transferred to amino acids of histone proteins, is crucial for the epigenetic gene regulation. NSD2 (nuclear receptor-binding SET domain protein 2) and SETMAR are epigenetic regulators for histone methylation. KDM2B, also known as FBXL10, is a histone demethylase that targets histone methylation processes. They are known to be altered in many cancers, but somatic frameshift mutation and expression of these genes remain undetermined in many other subsets of cancers, including high microsatellite instability (MSI-H) colon cancer (CC). In this study, we analyzed mononucleotide repeats in coding sequences of NSD2, KDM2B and SETMAR genes, and found frameshift mutations in 10 %, 2 % and 1 % of CCs with MSI-H, respectively. Of note, there was no frameshift mutation of these genes in microsatellite stable (MSS) CCs. In addition, we discovered that 2 and 2 of 16 CRCs (12.5 % and 12.5 %) harbored intratumoral heterogeneity (ITH) of the NSD2 and KDM2B frameshift mutations, respectively. In the immunohistochemistry for NSD2, intensity of NSD2 immunostaining in MSI-H CC is decreased compared to that in MSS. These results suggest that NSD2 might be altered at multiple levels (frameshift mutation, mutational ITH and expression) in MSI-H CCs, and could be related to MSI-H cancer pathogenesis.
Collapse
Affiliation(s)
- Seong Won Moon
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea
| | - Hyun Ji Son
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea; Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea
| | - Ha Yoon Mo
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea
| | - Eun Ji Choi
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea
| | - Nam Jin Yoo
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea
| | - Sug Hyung Lee
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea; Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea.
| |
Collapse
|
45
|
Use of Gefitinib in EGFR-Amplified Refractory Solid Tumors: An Open-Label, Single-Arm, Single-Center Prospective Pilot Study. Target Oncol 2020; 15:185-192. [PMID: 32107712 DOI: 10.1007/s11523-020-00706-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Treatment options for patients with chemotherapy-refractory solid tumors are limited. OBJECTIVE We conducted an open-label, single-arm, single-center phase II trial to evaluate the efficacy and safety of gefitinib in patients with chemotherapy-refractory solid tumors and EGFR amplification or sensitivity to an EGFR inhibitor identified through a drug-screening platform with patient-derived tumor cells (PDCs). PATIENTS AND METHODS EGFR amplification was detected by targeted sequencing. Sensitivity to an EGFR inhibitor was established in chemical screening using PDCs. Gefitinib (250 mg daily) was administered continuously in 28-day cycles until the occurrence of disease progression, unacceptable toxicity, or death due to any cause. The primary endpoint was the objective response rate (ORR). RESULTS In total, 15 patients were assigned to the present study. The most common tumor type was glioblastoma multiforme (n = 9, 60%), followed by gastric cancer (n = 3, 20%), anal squamous cancer, rectal cancer, and sarcoma (each n = 1, 6.7%). Among 13 evaluable patients, one patient had a partial response and five had stable disease, with an ORR of 7.7% and a disease control rate of 46.1%. The median progression-free survival was 2.1 months (95% confidence interval [CI] 0.77-3.43). The most common adverse events were diarrhea (26.7%) and skin rash (26.7%). CONCLUSION Gefitinib demonstrated modest anti-tumor activity and a manageable safety profile in chemotherapy-refractory solid tumors with EGFR amplification or sensitivity to an EGFR inhibitor identified through a drug-screening platform with PDCs. CLINICALTRIALS. GOV IDENTIFIER NCT02447419.
Collapse
|
46
|
Wang H, Wang T, Zhao X, Wu H, You M, Sun Z, Mao F. AI-Driver: an ensemble method for identifying driver mutations in personal cancer genomes. NAR Genom Bioinform 2020; 2:lqaa084. [PMID: 33575629 PMCID: PMC7671397 DOI: 10.1093/nargab/lqaa084] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 09/22/2020] [Accepted: 09/30/2020] [Indexed: 01/02/2023] Open
Abstract
The current challenge in cancer research is to increase the resolution of driver prediction from gene-level to mutation-level, which is more closely aligned with the goal of precision cancer medicine. Improved methods to distinguish drivers from passengers are urgently needed to dig out driver mutations from increasing exome sequencing studies. Here, we developed an ensemble method, AI-Driver (AI-based driver classifier, https://github.com/hatchetProject/AI-Driver), to predict the driver status of somatic missense mutations based on 23 pathogenicity features. AI-Driver has the best overall performance compared with any individual tool and two cancer-specific driver predicting methods. We demonstrate the superior and stable performance of our model using four independent benchmarks. We provide pre-computed AI-Driver scores for all possible human missense variants (http://aidriver.maolab.org/) to identify driver mutations in the sea of somatic mutations discovered by personal cancer sequencing. We believe that AI-Driver together with pre-computed database will play vital important roles in the human cancer studies, such as identification of driver mutation in personal cancer genomes, discovery of targeting sites for cancer therapeutic treatments and prediction of tumor biomarkers for early diagnosis by liquid biopsy.
Collapse
Affiliation(s)
- Haoxuan Wang
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China
| | - Tao Wang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410083, China
| | - Xiaolu Zhao
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Honghu Wu
- Department of Science and Technology, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | | | - Zhongsheng Sun
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China
| | - Fengbiao Mao
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
47
|
EGFR and BRAF mutations in inverted sinonasal papilloma - a more complex landscape? Virchows Arch 2020; 478:915-924. [PMID: 33048186 PMCID: PMC8286953 DOI: 10.1007/s00428-020-02945-y] [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: 11/19/2019] [Revised: 09/01/2020] [Accepted: 10/05/2020] [Indexed: 12/23/2022]
Abstract
Inverted (Schneiderian) sinonasal papilloma (ISP) is a neoplasm derived from mucosa of the sinonasal tract characterized by local aggressive growth, a tendency to recur and an association with sinonasal carcinoma. The etiology of ISP remains unclear. Recently, identical mutations in exons 19 and 20 of the oncogene EGFR were reported in ISP and ISP-associated sinonasal carcinoma. Nevertheless, it remains unclear whether recurring ISPs show identical EGFR mutations at different time points or whether these mutations are identical throughout the respective ISP sample. We used Sanger sequencing to test 60 formalin-fixed paraffin embedded ISP samples from 40 patients regarding mutations in exons 19 and 20 of EGFR—together with exon 15 of BRAF. Overall, 32 samples of 22 patients showed a mutation in EGFR exon 20, whereas 28 samples of 18 patients showed none. No mutation in EGFR exon 19 was found in any sample. Four samples of four patients showed a BRAF exon 15 mutation. Interestingly, samples of four patients exhibited genetic heterogeneity, enabling us to report this in ISP for the first time.
Collapse
|
48
|
Extracellular Vesicles as Biomarkers in Cancer Immunotherapy. Cancers (Basel) 2020; 12:cancers12102825. [PMID: 33007968 PMCID: PMC7600903 DOI: 10.3390/cancers12102825] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 09/17/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Extracellular vesicles (EVs) are small particles found throughout the body. EVs are released by living cells and contain cargo representing the cell of origin. In recent years, EVs have gained attention in cancer research. Since the cargo found inside EVs can be traced back to the cell of origin, EVs shed from cancer cells, in particular, may be used to better describe and characterize a patient’s tumor. EVs have been found and isolated from a variety of bodily fluids, including blood, saliva, and amniotic fluid, and therefore offer a non-invasive way of also diagnosing and monitoring patients before, during, and after cancer immunotherapy. The aim of this review article was to summarize some of the recent work conducted in this field and the challenges we face moving forward in utilizing EVs for cancer diagnostic and therapeutic purposes in cancer immunotherapy in the clinical setting. Abstract Extracellular vesicles (EVs), including exosomes and microvesicles, are membrane-bound vesicles secreted by most cell types during both physiologic conditions as well in response to cellular stress. EVs play an important role in intercellular communication and are emerging as key players in tumor immunology. Tumor-derived EVs (TDEs) harbor a diverse array of tumor neoantigens and contain unique molecular signature that is reflective of tumor’s underlying genetic complexity. As such they offer a glimpse into the immune tumor microenvironment (TME) and have the potential to be a novel, minimally invasive biomarker for cancer immunotherapy. Immune checkpoint inhibitors (ICI), such as anti- programmed death-1(PD-1) and its ligand (PD-L1) antibodies, have revolutionized the treatment of a wide variety of solid tumors including head and neck squamous cell carcinoma, urothelial carcinoma, melanoma, non-small cell lung cancer, and others. Typically, an invasive tissue biopsy is required both for histologic diagnosis and next-generation sequencing efforts; the latter have become more widespread in daily clinical practice. There is an unmet need for noninvasive or minimally invasive (e.g., plasma-based) biomarkers both for diagnosis and treatment monitoring. Targeted analysis of EVs in biospecimens, such as plasma and saliva could serve this purpose by potentially obviating the need for tissue sample. In this review, we describe the current challenges of biomarkers in cancer immunotherapy as well as the mechanistic role of TDEs in modulating antitumor immune response.
Collapse
|
49
|
Beyond tissue biopsy: a diagnostic framework to address tumor heterogeneity in lung cancer. Curr Opin Oncol 2020; 32:68-77. [PMID: 31714259 DOI: 10.1097/cco.0000000000000598] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW The objective of this review is to discuss the strength and limitations of tissue and liquid biopsy and functional imaging to capture spatial and temporal tumor heterogeneity either alone or as part of a diagnostic framework in non-small cell lung cancer (NSCLC). RECENT FINDINGS NSCLC displays genetic and phenotypic heterogeneity - a detailed knowledge of which is crucial to personalize treatment. Tissue biopsy often lacks spatial and temporal resolution. Thus, NSCLC needs to be characterized by complementary diagnostic methods to resolve heterogeneity. Liquid biopsy offers detection of tumor biomarkers and for example, the classification and monitoring of EGFR mutations in NSCLC. It allows repeated sampling, and therefore, appears promising to address temporal aspects of tumor heterogeneity. Functional imaging methods and emerging image analytic tools, such as radiomics capture temporal and spatial heterogeneity. Further standardization of radiomics is required to allow introduction into clinical routine. SUMMARY To augment the potential of precision therapy, improved diagnostic characterization of tumors is pivotal. We suggest a comprehensive diagnostic framework combining tissue and liquid biopsy and functional imaging to address the known aspects of spatial and temporal tumor heterogeneity on the example of NSCLC. We envision how this framework might be implemented in clinical practice.
Collapse
|
50
|
Ye P, Cai P, Xie J, Wei Y. The diagnostic accuracy of digital PCR, ARMS and NGS for detecting KRAS mutation in cell-free DNA of patients with colorectal cancer: A protocol for systematic review and meta-analysis. Medicine (Baltimore) 2020; 99:e20708. [PMID: 32590745 PMCID: PMC7328928 DOI: 10.1097/md.0000000000020708] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Cetuximab and panitumumab have been used clinically to treat metastatic colorectal cancer for more than 15 years. Before the treatment is given, it is required to determine the KRAS mutation status since it would lead to drug resistance. Tumor tissue sample is traditionally used for cancer genotyping. In recent years, liquid biopsy sample has been intensively investigated as a surrogate for tumor tissue sample due to its non-invasiveness and better presentation of tumor heterogeneity. The aim of this study is to systematically summarize the accuracy of KRAS mutation measurement in colorectal cancer using cell-free DNA in liquid biopsy samples, with tumor tissue sample as reference (gold standard). METHODS AND ANALYSIS We will search literatures in the following databases: Pubmed, Embase, and Cochrane Library. Systemic review and meta-analysis will be performed to summarize the accuracy of KRAS mutation measurement in colorectal cancer using liquid biopsy sample, and subgroup analysis will be performed on different testing platforms, and on metastatic and non-metastatic colorectal cancer. TIMELINE This study will start on June 1, 2020, and is expected to be finished by November 1, 2020. ETHICS AND DISSEMINATION Ethical approval will not be required since the data obtained and analyzed in this study will not be on individual patients. Study results will be disseminated as an official publication in a peer-reviewed journal.Registration: PROSPERO CRD42020176682.
Collapse
Affiliation(s)
- Peng Ye
- Department of Anatomy and Histology, College of Medicine, Chengdu University
| | - Peiling Cai
- Department of Anatomy and Histology, College of Medicine, Chengdu University
| | - Jing Xie
- Department of Pathology and Clinical Laboratory, Sichuan Provincial Fourth People's Hospital
| | - Yuanyuan Wei
- Department of Physiology, College of Medicine, Chengdu University, Chengdu, China
| |
Collapse
|