51
|
Giraudeau M, Vincze O, Dupont SM, Sepp T, Baines C, Lemaitre JF, Lemberger K, Gentès S, Boddy A, Dujon AM, Bramwell G, Harris V, Ujvari B, Alix-Panabières C, Lair S, Sayag D, Conde DA, Colchero F, Harrison TM, Pavard S, Padilla-Morales B, Chevallier D, Hamede R, Roche B, Malkocs T, Aktipis AC, Maley C, DeGregori J, Loc’h GL, Thomas F. Approaches and methods to study wildlife cancer. J Anim Ecol 2024; 93:1410-1428. [PMID: 39189422 PMCID: PMC11745198 DOI: 10.1111/1365-2656.14144] [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: 12/05/2023] [Accepted: 04/23/2024] [Indexed: 08/28/2024]
Abstract
The last few years have seen a surge of interest from field ecologists and evolutionary biologists to study neoplasia and cancer in wildlife. This contributes to the One Health Approach, which investigates health issues at the intersection of people, wild and domestic animals, together with their changing environments. Nonetheless, the emerging field of wildlife cancer is currently constrained by methodological limitations in detecting cancer using non-invasive sampling. In addition, the suspected differential susceptibility and resistance of species to cancer often make the choice of a unique model species difficult for field biologists. Here, we provide an overview of the importance of pursuing the study of cancer in non-model organisms and we review the currently available methods to detect, measure and quantify cancer in the wild, as well as the methodological limitations to be overcome to develop novel approaches inspired by diagnostic techniques used in human medicine. The methodology we propose here will help understand and hopefully fight this major disease by generating general knowledge about cancer, variation in its rates, tumour-suppressor mechanisms across species as well as its link to life history and physiological characters. Moreover, this is expected to provide key information about cancer in wildlife, which is a top priority due to the accelerated anthropogenic change in the past decades that might favour cancer progression in wild populations.
Collapse
Affiliation(s)
- Mathieu Giraudeau
- Littoral Environnement et Sociétés (LIENSs), UMR 7266 CNRS-La Rochelle Université, La Rochelle, France
| | - Orsolya Vincze
- Littoral Environnement et Sociétés (LIENSs), UMR 7266 CNRS-La Rochelle Université, La Rochelle, France
- ImmunoConcEpT, CNRS UMR 5164, University of Bordeaux, Bordeaux, France
- Hungarian Department of Biology and Ecology, Evolutionary Ecology Group, Babeş-Bolyai University, Cluj-Napoca, Romania
- HUN-REN-DE Conservation Biology Research Group, Debrecen, Hungary
| | - Sophie M. Dupont
- Littoral Environnement et Sociétés (LIENSs), UMR 7266 CNRS-La Rochelle Université, La Rochelle, France
- Laboratoire de Biologie des ORganismes et Ecosystèmes Aquatiques (BOREA), FRE 2030, Muséum National d’Histoire Naturelle, CNRS, IRD, Sorbonne Université, Université de Caen Normandie, Université des Antilles, Paris, France
| | - Tuul Sepp
- Institute of Ecology and Earth Sciences, University of Tartu, Tartu, Estonia
| | - Ciara Baines
- Department of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Jean-Francois Lemaitre
- Laboratoire de Biométrie et Biologie Évolutive, CNRS, UMR5558, Université Lyon 1, Villeurbanne, France
| | | | - Sophie Gentès
- Littoral Environnement et Sociétés (LIENSs), UMR 7266 CNRS-La Rochelle Université, La Rochelle, France
| | - Amy Boddy
- Department of Anthropology, University of California Santa Barbara, Santa Barbara, California, USA
| | - Antoine M. Dujon
- School of Life and Environmental Sciences, Deakin University, Waurn Ponds, Victoria, Australia
- CREEC/CANECEV, MIVEGEC, Unité Mixte de Recherches, IRD 224–CNRS5290–Université de Montpellier, Montpellier, France
| | - Georgina Bramwell
- School of Life and Environmental Sciences, Deakin University, Waurn Ponds, Victoria, Australia
| | - Valerie Harris
- Arizona Cancer Evolution Center, Biodesign Institute, Arizona State University, Tempe, Arizona, USA
| | - Beata Ujvari
- School of Life and Environmental Sciences, Deakin University, Waurn Ponds, Victoria, Australia
- Centre de Recherches Ecologiques et Evolutives sur le Cancer, Montpellier, France
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, Montpellier, France
| | - Stephane Lair
- Faculté de médecine vétérinaire, Canadian Wildlife Health Cooperative/Centre québécois sur la santé des animaux sauvages, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - David Sayag
- ONCOnseil—Unité d’expertise en oncologie vétérinaire, Toulouse, France
| | - Dalia A. Conde
- Department of Biology, University of Southern Denmark, Odense M, Denmark
- Interdisciplinary Centre on Population Dynamics, University of Southern Denmark, Odense M, Denmark
| | - Fernando Colchero
- Interdisciplinary Centre on Population Dynamics, University of Southern Denmark, Odense M, Denmark
- Department of Primate Behavior and Evolution, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
- Department of Mathematics and Computer Sciences, University of Southern Denmark, Odense M, Denmark
| | - Tara M. Harrison
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Samuel Pavard
- Unité Eco-Anthropologie (EA), Muséum National d’Histoire Naturelle, CNRS 7206, Université Paris Cité, Paris, France
| | - Benjamin Padilla-Morales
- Department of Biology and Biochemistry, Milner Centre for Evolution, University of Bath, Bath, UK
| | - Damien Chevallier
- Laboratoire de Biologie des ORganismes et Ecosystèmes Aquatiques (BOREA), FRE 2030, Muséum National d’Histoire Naturelle, CNRS, IRD, Sorbonne Université, Université de Caen Normandie, Université des Antilles, Paris, France
| | - Rodrigo Hamede
- Centre de Recherches Ecologiques et Evolutives sur le Cancer, Montpellier, France
- School of Natural Sciences, University of Tasmania, Hobart, Tasmania, Australia
| | - Benjamin Roche
- CREEC/CANECEV, MIVEGEC, Unité Mixte de Recherches, IRD 224–CNRS5290–Université de Montpellier, Montpellier, France
- Centre de Recherche en Écologie et Évolution de la Santé (CREES), Montpellier, France
- Departamento de Etología, Fauna Silvestre y Animales de Laboratorio, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| | - Tamas Malkocs
- Littoral Environnement et Sociétés (LIENSs), UMR 7266 CNRS-La Rochelle Université, La Rochelle, France
- Univ Brest, CNRS, IRD, Ifremer, LEMAR, IUEM, Plouzane, France
| | - Athena C. Aktipis
- Arizona Cancer Evolution Center, Biodesign Institute, Arizona State University, Tempe, Arizona, USA
- Department of Psychology, Arizona State University, Tempe, Arizona, USA
| | - Carlo Maley
- Arizona Cancer Evolution Center, Biodesign Institute, Arizona State University, Tempe, Arizona, USA
| | - James DeGregori
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | | | - Frédéric Thomas
- CREEC/CANECEV, MIVEGEC, Unité Mixte de Recherches, IRD 224–CNRS5290–Université de Montpellier, Montpellier, France
- Centre de Recherche en Écologie et Évolution de la Santé (CREES), Montpellier, France
| |
Collapse
|
52
|
Chen J, Liu X, Zhang Z, Su R, Geng Y, Guo Y, Zhang Y, Su M. Early Diagnostic Markers for Esophageal Squamous Cell Carcinoma: Copy Number Alteration Gene Identification and cfDNA Detection. J Transl Med 2024; 104:102127. [PMID: 39182610 DOI: 10.1016/j.labinv.2024.102127] [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: 05/25/2024] [Revised: 08/17/2024] [Accepted: 08/19/2024] [Indexed: 08/27/2024] Open
Abstract
The high mortality rate of esophageal squamous cell carcinoma (ESCC) is exacerbated by the absence of early diagnostic markers. The pronounced heterogeneity of mutations in ESCC renders copy number alterations (CNAs) more prevalent among patients. The identification of CNA genes within esophageal squamous dysplasia (ESD), a precancerous stage of ESCC, is crucial for advancing early detection efforts. Utilization of liquid biopsies via droplet-based digital PCR (ddPCR) offers a novel strategy for detecting incipient tumor traces. This study undertook a thorough investigation of CNA profiles across ESCC development stages, integrating data from existing databases and prior investigations to pinpoint and confirm CNA markers conducive to early detection of ESCC. Targeted sequencing was employed to select potential early detection genes, followed by the establishment of prediction models for ESCC early detection using ddPCR. Our analysis revealed widespread CNAs during the ESD stage, mirroring the CNA landscape observed in ESCC. A total of 40 CNA genes were identified as highly frequent in both ESCC and ESD lesions, through a comprehensive gene-level CNA analysis encompassing ESD and ESCC tissues, ESCC cell lines, and pan-cancer data sets. Subsequent validation of 5 candidate markers via ddPCR underscored the efficacy of combined predictive models encompassing PIK3CA, SOX2, EGFR, MYC, and CCND1 in early ESCC screening, as evidenced by the area-under-the-curve values exceeding 0.92 (P < .0001) across various detection contexts. The findings highlighted the significant utility of CNA genes in the early screening of ESCC, presenting robust models that could facilitate early detection, broad-scale population screening, and adjunctive diagnosis.
Collapse
Affiliation(s)
- Jiamin Chen
- Department of Pathology, Institute of Clinical Pathology, Shantou University Medical College, Shantou, China
| | - Xi Liu
- Department of Pathology, Institute of Clinical Pathology, Shantou University Medical College, Shantou, China
| | - Zhihua Zhang
- Department of Pathology, Institute of Clinical Pathology, Shantou University Medical College, Shantou, China
| | - Ruibing Su
- Department of Pathology, Institute of Clinical Pathology, Shantou University Medical College, Shantou, China; Department of Cardiology, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Yiqun Geng
- Department of Molecular Pathology, Shantou University Medical College, Shantou, China
| | - Yi Guo
- Department of Endoscopy, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Yimin Zhang
- Clinical Research Center, Shantou Central Hospital, Shantou, China
| | - Min Su
- Department of Pathology, Institute of Clinical Pathology, Shantou University Medical College, Shantou, China.
| |
Collapse
|
53
|
Chen T, Wang M, Chen Y, Cao Y, Liu Y. Advances in predictive biomarkers associated with immunotherapy in extensive-stage small cell lung cancer. Cell Biosci 2024; 14:117. [PMID: 39267195 PMCID: PMC11391723 DOI: 10.1186/s13578-024-01283-9] [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: 05/06/2024] [Accepted: 08/05/2024] [Indexed: 09/14/2024] Open
Abstract
Small cell lung cancer (SCLC) is a highly malignant and poor-prognosis cancer, with most cases diagnosed at the extensive stage (ES). Amidst a landscape marked by limited progress in treatment modalities for ES-SCLC over the past few decades, the integration of immune checkpoint inhibitors (ICIs) with platinum-based chemotherapy has provided a milestone approach for improving prognosis, emerging as the new standard for initial therapy in ES-SCLC. However, only a minority of SCLC patients can benefit from ICIs, which frequently come with varying degrees of immune-related adverse events (irAEs). Therefore, it is crucial to investigate predictive biomarkers to screen potential beneficiaries of ICIs, mitigate the risk of side effects, and improve treatment precision. This review summarized potential biomarkers for predicting ICI response in ES-SCLC, with a primary focus on markers sourced from tumor tissue or peripheral blood samples. The former mainly included PD-L1 expression, tumor mutational burden (TMB), along with cellular or molecular components related to the tumor microenvironment (TME) and antigen presentation machinery (APM), molecular subtypes of SCLC, and inflammatory gene expression profiles. Circulating biomarkers predominantly comprised circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), cytokines, plasma autoantibodies, inflammation-related parameters, and blood TMB. We synthesized and analyzed the research progress of these potential markers. Notably, investigations into PD-L1 expression and TMB have been the most extensive, exhibiting preliminary predictive efficacy in salvage immunotherapy; however, consistent conclusions have yet to be reached across studies. Additionally, novel predictive markers developed based on TME composition, APM, transcriptomic and genomic features provide promising tools for precision immunotherapy. Circulating biomarkers offer the advantages of convenience, non-invasiveness, and a comprehensive reflection of tumor molecular characteristics. They may serve as alternative options for predicting immunotherapy efficacy in SCLC. However, there is a scarcity of studies, and the significant heterogeneity in research findings warrants attention.
Collapse
Affiliation(s)
- Tong Chen
- 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, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Mingzhao Wang
- 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, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Yanchao Chen
- 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, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Yang Cao
- 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, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Yutao Liu
- 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, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
54
|
Demirbüken SE, Öztürk E, Güngör MA, Garipcan B, Kuralay F. Modified Au:Fe-Ni magnetic micromotors improve drug delivery and diagnosis in MCF-7 cells and spheroids. Colloids Surf B Biointerfaces 2024; 241:114019. [PMID: 38897023 DOI: 10.1016/j.colsurfb.2024.114019] [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: 02/29/2024] [Revised: 05/26/2024] [Accepted: 06/06/2024] [Indexed: 06/21/2024]
Abstract
Nano/micromotors hold immense potential for revolutionizing drug delivery and detection systems, especially in the realm of cancer diagnosis and treatment, owing to their distinctive features, including precise propulsion, maneuverability, and meticulously designed surface modifications. In this study, we explore the capabilities of modified and magnetically driven micromotors as active drug delivery systems within 2D and 3D cell culture environments and cancer diagnosis. We synthesized gold (Au) and iron-nickel (Fe-Ni) metallic-based magnetic micromotors (Au:Fe-Ni MMs) through electrochemical methods, equipping them with functionalities for controlled doxorubicin (DOX) release and cancer cell recognition. In 2D and spheroids of MCF-7 adenocarcinoma cells, the Au segment of these micromotors was utilized to help DOX loading through poly(sodium-4-styrenesulfonate) (PSS) functionalization, and the attachment of antiHER2 antibodies for specific recognition. This innovative approach enabled controlled drug release within the cancerous microenvironment, coupled with magnetic (Fe-Ni) propulsion for biocompatible drug delivery to MCF-7 cells. Furthermore, antiHER2 immobilized Au:Fe-Ni MMs effectively interacted with receptors, capitalizing on the overexpression of HER2 antigens on MCF-7 cells. Encouraging outcomes were observed, particularly in spheroid models, underscoring the remarkable potential of these multifunctional micromotors for advancing intelligent drug delivery methodologies and diagnostic purposes.
Collapse
Affiliation(s)
| | - Elif Öztürk
- Department of Chemistry, Faculty of Sciences, Hacettepe University, Ankara 06800, Turkey
| | - Mustafa Ali Güngör
- Department of Chemistry, Faculty of Sciences, Hacettepe University, Ankara 06800, Turkey; Department of Chemistry, Polatlı Faculty of Arts and Sciences, Ankara Hacı Bayram Veli University, Polatlı, Ankara 06900, Turkey
| | - Bora Garipcan
- Institute of Biomedical Engineering, Bogazici University, Istanbul 34684, Turkey.
| | - Filiz Kuralay
- Department of Chemistry, Faculty of Sciences, Hacettepe University, Ankara 06800, Turkey.
| |
Collapse
|
55
|
Lin H, Cao XX. Current State of Targeted Therapy in Adult Langerhans Cell Histiocytosis and Erdheim-Chester Disease. Target Oncol 2024; 19:691-703. [PMID: 38990463 DOI: 10.1007/s11523-024-01080-x] [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] [Accepted: 06/25/2024] [Indexed: 07/12/2024]
Abstract
The mitogen-activated protein kinase (MAPK) pathway is a key driver in many histiocytic disorders, including Langerhans cell histiocytosis (LCH) and Erdheim-Chester disease (ECD). This has led to successful and promising treatment with targeted therapies, including BRAF inhibitors and MEK inhibitors. Additional novel inhibitors have also demonstrated encouraging results. Nevertheless, there are several problems concerning targeted therapy that need to be addressed. These include, among others, incomplete responsiveness and the emergence of resistance to BRAF inhibition as observed in other BRAF-mutant malignancies. Drug resistance and relapse after treatment interruption remain problems with current targeted therapies. Targeted therapy does not seem to eradicate the mutated clone, leading to inevitable relapes, which is a huge challenge for the future. More fundamental research and clinical trials are needed to address these issues and to develop improved targeted therapies that can overcome resistance and achieve long-lasting remissions.
Collapse
Affiliation(s)
- He Lin
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China
| | - Xin-Xin Cao
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China.
| |
Collapse
|
56
|
Yin L, Duan A, Zhang W, Li B, Zhao T, Xu X, Yang L, Nian B, Lu K, Chen S, Li Z, Liu J, Duan Q, Liu D, Chen H, Cui L, Chang Y, Kuang Y, Zhang D, Wang X, Zhang Y. Identification of whole-genome mutations and structural variations of bile cell-free DNA in cholangiocarcinoma. Genomics 2024; 116:110916. [PMID: 39147332 DOI: 10.1016/j.ygeno.2024.110916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/17/2024] [Accepted: 08/02/2024] [Indexed: 08/17/2024]
Abstract
Bile cell-free DNA (cfDNA) has been reported as a promising liquid biopsy tool for cholangiocarcinoma (CCA), however, the whole-genome mutation landscape and structural variants (SVs) of bile cfDNA remains unknown. Here we performed whole-genome sequencing on bile cfDNA and analyzed the correlation between mutation characteristics of bile cfDNA and clinical prognosis. TP53 and KRAS were the most frequently mutated genes, and the RTK/RAS, homologous recombination (HR), and HIPPO were top three pathways containing most gene mutations. Ten overlapping putative driver genes were found in bile cfDNA and tumor tissue. SVs such as chromothripsis and kataegis were identified. Moreover, the hazard ratio of HR pathway mutations were 15.77 (95% CI: 1.571-158.4), patients with HR pathway mutations in bile cfDNA exhibited poorer overall survival (P = 0.0049). Our study suggests that bile cfDNA contains genome mutations and SVs, and HR pathway mutations in bile cfDNA can predict poor outcomes of CCA patients.
Collapse
Affiliation(s)
- Lei Yin
- Second Department of Biliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Anqi Duan
- Second Department of Biliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Wei Zhang
- Department of Clinical and Translational Medicine, 3D Medicines Inc., Shanghai, China
| | - Bin Li
- Department of Clinical and Translational Medicine, 3D Medicines Inc., Shanghai, China
| | - Teng Zhao
- Second Department of Biliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Xiaoya Xu
- Department of Clinical and Translational Medicine, 3D Medicines Inc., Shanghai, China
| | - Lixue Yang
- Second Department of Biliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Baoning Nian
- Department of Clinical and Translational Medicine, 3D Medicines Inc., Shanghai, China
| | - Kai Lu
- Second Department of Biliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Sheng Chen
- Department of Clinical and Translational Medicine, 3D Medicines Inc., Shanghai, China
| | - Zhikuan Li
- Department of Clinical and Translational Medicine, 3D Medicines Inc., Shanghai, China
| | - Jian Liu
- Second Department of Biliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Qiaonan Duan
- Department of Clinical and Translational Medicine, 3D Medicines Inc., Shanghai, China
| | - Dongyu Liu
- Department of Clinical and Translational Medicine, 3D Medicines Inc., Shanghai, China
| | - Hao Chen
- Department of Clinical and Translational Medicine, 3D Medicines Inc., Shanghai, China
| | - Longjiu Cui
- Second Department of Biliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Yanxin Chang
- Second Department of Biliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Yue Kuang
- Second Department of Biliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Dadong Zhang
- Department of Clinical and Translational Medicine, 3D Medicines Inc., Shanghai, China.
| | - Xiang Wang
- Second Department of Biliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University (Second Military Medical University), Shanghai, China.
| | - Yongjie Zhang
- Second Department of Biliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University (Second Military Medical University), Shanghai, China.
| |
Collapse
|
57
|
Park W, Gwack J, Park J. Implementing Massive Parallel Sequencing into Biliary Samples Obtained through Endoscopic Retrograde Cholangiopancreatography for Diagnosing Malignant Bile Duct Strictures. Int J Mol Sci 2024; 25:9461. [PMID: 39273408 PMCID: PMC11395203 DOI: 10.3390/ijms25179461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 09/15/2024] Open
Abstract
Despite advancements in radiologic, laboratory, and pathological evaluations, differentiating between benign and malignant bile duct strictures remains a diagnostic challenge. Recent developments in massive parallel sequencing (MPS) have introduced new opportunities for early cancer detection and management, but these techniques have not yet been rigorously applied to biliary samples. We prospectively evaluated the Oncomine Comprehensive Assay (OCA) and the Oncomine Pan-Cancer Cell-Free Assay (OPCCFA) using biliary brush cytology and bile fluid obtained via endoscopic retrograde cholangiopancreatography from patients with bile duct strictures. The diagnostic performance of MPS testing was assessed and compared to the pathological findings of biliary brush cytology and primary tissue. Mutations in TP53, BRAF, CTNNB1, SMAD4, and K-/N-RAS identified in biliary brush cytology samples were also detected in the corresponding bile fluid samples from patients with extrahepatic cholangiocarcinoma. These mutations were also identified in the bile fluid samples, but with variant allele frequencies lower than those in the corresponding biliary brush cytology samples. In control patients diagnosed with gallstones, neither the biliary brush cytology samples nor the bile fluid samples showed any pathogenic mutations classified as tier 1 or 2. Our study represents a prospective investigation into the role of MPS-based molecular testing in evaluating bile duct strictures. MPS-based molecular testing shows promise in identifying actionable genomic alterations, potentially enabling the stratification of patients for targeted chemotherapeutic treatments. Future research should focus on integrating OCA and OPCCFA testing, as well as similar MPS-based assays, into existing surveillance and management protocols for patients with bile duct strictures.
Collapse
Affiliation(s)
- Wonsuk Park
- Division of Gastroenterology, Department of Internal Medicine, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jin Gwack
- Department of Preventive Medicine, Jeonbuk National University Medical School, Jeonju 54907, Republic of Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea
| | - Joonhong Park
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea
- Department of Laboratory Medicine, Jeonbuk National University Medical School and Hospital, Jeonju 54907, Republic of Korea
| |
Collapse
|
58
|
Rocchetti F, Tenore G, Macali F, Vicidomini T, Podda GM, Fantozzi PJ, Silvestri V, Porzio V, Valentini V, Ottini L, Richetta AG, Valentini V, Della Monaca M, Grenga C, Polimeni A, Romeo U. Expression Analysis of Circulating microRNAs in Saliva and Plasma for the Identification of Clinically Relevant Biomarkers for Oral Squamous Cell Carcinoma and Oral Potentially Malignant Disorders. Cancers (Basel) 2024; 16:2990. [PMID: 39272848 PMCID: PMC11394426 DOI: 10.3390/cancers16172990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/10/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
This study aims to evaluate the expression of salivary and plasmatic miRNAs as diagnostic biomarkers in patients with oral squamous cell carcinoma (OSCC) and oral potentially malignant disorders (OPMDs). A total of 25 patients were divided into three groups, according to their diagnosis: OSCC patients (n = 14); OPMDs patients (n = 6); and healthy controls (n = 5). At the time at diagnosis/enrolment, patients underwent salivary and plasmatic collection. The expression of miRNA -21, -31, -138, -145, -184, and -424 were evaluated by real-time PCR. An F-test and ANOVA test were performed to evaluate the miRNA levels (significance at p < 0.05). By comparing miRNA expression levels from saliva, a statistically significant difference emerged in the expression of miR-138 and miR-424 between the three groups (p < 0.05). In particular, these two miRNAs showed decreased expression levels in saliva samples from OSCC and OPMD patients compared to those from healthy controls. On the other hand, miRNA expression levels in plasma were low in all the groups, and no statistically significant differences were found. Overall, our results showed that liquid biopsy from saliva may be a useful tool for the identification of diagnostic molecular biomarkers in OSCC and OPMDs.
Collapse
Affiliation(s)
- Federica Rocchetti
- Department of Oral and Maxillofacial Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Gianluca Tenore
- Department of Oral and Maxillofacial Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Federica Macali
- Department of Oral and Maxillofacial Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Teresa Vicidomini
- Department of Oral and Maxillofacial Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Gian Marco Podda
- Department of Oral and Maxillofacial Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Paolo Junior Fantozzi
- Department of Oral and Maxillofacial Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Valentina Silvestri
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Virginia Porzio
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Virginia Valentini
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Laura Ottini
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Antonio Giovanni Richetta
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, 00161 Rome, Italy
| | - Valentino Valentini
- Department of Oral and Maxillofacial Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Marco Della Monaca
- Department of Oral and Maxillofacial Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Camilla Grenga
- Department of Oral and Maxillofacial Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Antonella Polimeni
- Department of Oral and Maxillofacial Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Umberto Romeo
- Department of Oral and Maxillofacial Sciences, Sapienza University of Rome, 00161 Rome, Italy
| |
Collapse
|
59
|
Karp JM, Modrek AS, Ezhilarasan R, Zhang ZY, Ding Y, Graciani M, Sahimi A, Silvestro M, Chen T, Li S, Wong KK, Ramkhelawon B, Bhat KP, Sulman EP. Deconvolution of the tumor-educated platelet transcriptome reveals activated platelet and inflammatory cell transcript signatures. JCI Insight 2024; 9:e178719. [PMID: 39190500 PMCID: PMC11466191 DOI: 10.1172/jci.insight.178719] [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: 12/21/2023] [Accepted: 08/13/2024] [Indexed: 08/29/2024] Open
Abstract
Tumor-educated platelets (TEPs) are a potential method of liquid biopsy for the diagnosis and monitoring of cancer. However, the mechanism underlying tumor education of platelets is not known, and transcripts associated with TEPs are often not tumor-associated transcripts. We demonstrated that direct tumor transfer of transcripts to circulating platelets is an unlikely source of the TEP signal. We used CDSeq, a latent Dirichlet allocation algorithm, to deconvolute the TEP signal in blood samples from patients with glioblastoma. We demonstrated that a substantial proportion of transcripts in the platelet transcriptome are derived from nonplatelet cells, and the use of this algorithm allows the removal of contaminant transcripts. Furthermore, we used the results of this algorithm to demonstrate that TEPs represent a subset of more activated platelets, which also contain transcripts normally associated with nonplatelet inflammatory cells, suggesting that these inflammatory cells, possibly in the tumor microenvironment, transfer transcripts to platelets that are then found in circulation. Our analysis suggests a useful and efficient method of processing TEP transcriptomic data to enable the isolation of a unique TEP signal associated with specific tumors.
Collapse
Affiliation(s)
- Jerome M. Karp
- Department of Radiation Oncology, NYU Grossman School of Medicine, New York, New York, USA
| | - Aram S. Modrek
- Department of Radiation Oncology, Keck School of Medicine of University of Southern California, Los Angeles, California, USA
| | - Ravesanker Ezhilarasan
- Department of Radiation Oncology, NYU Grossman School of Medicine, New York, New York, USA
| | - Ze-Yan Zhang
- Department of Radiation Oncology, NYU Grossman School of Medicine, New York, New York, USA
| | - Yingwen Ding
- Department of Radiation Oncology, NYU Grossman School of Medicine, New York, New York, USA
| | - Melanie Graciani
- Department of Radiation Oncology, NYU Grossman School of Medicine, New York, New York, USA
| | - Ali Sahimi
- Department of Radiation Oncology, NYU Grossman School of Medicine, New York, New York, USA
| | | | - Ting Chen
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, New York, USA
| | - Shuai Li
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, New York, USA
| | - Kwok-Kin Wong
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, New York, USA
| | | | | | - Erik P. Sulman
- Department of Radiation Oncology, NYU Grossman School of Medicine, New York, New York, USA
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
60
|
Pataer P, Zhang P, Li Z. Single Methylation Sensitive Restriction Endonuclease-Based Cascade Exponential Amplification Assay for Visual Detection of DNA Methylation at Single-Molecule Level. Anal Chem 2024; 96:13335-13343. [PMID: 39093040 DOI: 10.1021/acs.analchem.4c03638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Function as a potential cancer biomarker, DNA methylation shows great significance in cancer diagnosis, prognosis, and treatment monitoring. While the lack of an ultrasensitive, specific, and accurate method at the single-molecule level hinders the analysis of the exceedingly low levels of DNA methylation. Herein, based on the outstanding recognition and digestion ability of methylation-sensitive restriction endonuclease (MSRE), we established a single MSRE-based cascade exponential amplification method, which requires only two ingeniously designed primers and only one recognition site of MSRE for the detection of DNA methylation. Differentiated by MSRE digestion, the cleaved unmethylated DNA is too short to induce any amplification reactions, while methylated DNA remains intact to trigger cascade exponential amplification and the subsequent CRISPR/Cas12a system. By integrating the two exponential amplification reactions, as low as 1 aM methylated DNA can be accurately detected, which corresponds to 6 molecules in a 10 μL system, indicating that our method is more sensitive than single amplification-based methods with the ability to detect DNA methylation at the single-molecule level. In addition, 0.1% methylated DNA can be effectively distinguished from large amounts of unmethylated DNA. Our method is further introduced to exploit the expression difference of DNA methylation among normal cells and cancer cells. Moreover, the visual detection of DNA methylation is also realized by the full hybridization between amplification products and the crRNA of CRISPR/Cas12a. Therefore, the proposed method has great potential to be a promising and robust bisulfite-free method for the detection of DNA methylation at the single-molecule level, which is of great importance for early diagnosis of cancer.
Collapse
Affiliation(s)
- Parezhati Pataer
- Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, 30 Xueyuan Road, Haidian District, Beijing 100083, China
| | - Pengbo Zhang
- Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, 30 Xueyuan Road, Haidian District, Beijing 100083, China
| | - Zhengping Li
- Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, 30 Xueyuan Road, Haidian District, Beijing 100083, China
| |
Collapse
|
61
|
Chung J, Xiao S, Gao Y, Soung YH. Recent Technologies towards Diagnostic and Therapeutic Applications of Circulating Nucleic Acids in Colorectal Cancers. Int J Mol Sci 2024; 25:8703. [PMID: 39201393 PMCID: PMC11354501 DOI: 10.3390/ijms25168703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 07/30/2024] [Accepted: 08/05/2024] [Indexed: 09/02/2024] Open
Abstract
Liquid biopsy has emerged as a promising noninvasive approach for colorectal cancer (CRC) management. This review focuses on technologies detecting circulating nucleic acids, specifically circulating tumor DNA (ctDNA) and circulating RNA (cfRNA), as CRC biomarkers. Recent advancements in molecular technologies have enabled sensitive and specific detection of tumor-derived genetic material in bodily fluids. These include quantitative real-time PCR, digital PCR, next-generation sequencing (NGS), and emerging nanotechnology-based methods. For ctDNA analysis, techniques such as BEAMing and droplet digital PCR offer high sensitivity in detecting rare mutant alleles, while NGS approaches provide comprehensive genomic profiling. cfRNA detection primarily utilizes qRT-PCR arrays, microarray platforms, and RNA sequencing for profiling circulating microRNAs and discovering novel RNA biomarkers. These technologies show potential in early CRC detection, treatment response monitoring, minimal residual disease assessment, and tumor evolution tracking. However, challenges remain in standardizing procedures, optimizing detection limits, and establishing clinical utility across disease stages. This review summarizes current circulating nucleic acid detection technologies, their CRC applications, and discusses future directions for clinical implementation.
Collapse
Affiliation(s)
| | | | | | - Young Hwa Soung
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA; (J.C.); (S.X.); (Y.G.)
| |
Collapse
|
62
|
Suri S, Boora GS, Kaur R, Chauhan A, Ghoshal S, Pal A. Recent advances in minimally invasive biomarkers of OSCC: from generalized to personalized approach. FRONTIERS IN ORAL HEALTH 2024; 5:1426507. [PMID: 39157206 PMCID: PMC11327221 DOI: 10.3389/froh.2024.1426507] [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: 05/01/2024] [Accepted: 07/16/2024] [Indexed: 08/20/2024] Open
Abstract
Oral cancer is the 6th most common type of cancer worldwide, and oral squamous cell carcinoma (OSCC) accounts for >90% of oral cancers. It is a major health problem, particularly in low- and middle-income countries (LMICs), due to both its high incidence and significant mortality and morbidity. Despite being a global burden, and even with the significant advancement in the management of OSCC, the overall outcome of the disease is still abysmal. With the advent of time, advanced diagnostic and treatment approaches have come into practice, but the burden of the disease has not improved significantly. Major reasons attributed to the poor outcome are delay in diagnosis, locoregional recurrence and resistance to the currently available treatment regimen. In this review, we have highlighted the existing challenges in the diagnosis and have emphasized the advancements in minimally invasive biomarkers. Additionally, the importance of collaborative multidimensional approaches involving clinicians and researchers has been discussed, as well as the need to redefine and establish better utility and management of existing diagnostic and treatment protocols along with the minimally invasive/non-invasive biomarkers.
Collapse
Affiliation(s)
- Smriti Suri
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh,India
| | - Geeta S. Boora
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh,India
| | - Rajandeep Kaur
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh,India
| | - Anshika Chauhan
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh,India
| | - Sushmita Ghoshal
- Department of Radiotherapy, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Arnab Pal
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh,India
| |
Collapse
|
63
|
Cao L, Yang W, Zhao X, Chen Z. Diagnostic and prognostic value of circulating tumor cells in renal cell cancer: A systematic review and meta-analysis. Asian J Surg 2024; 47:3425-3434. [PMID: 38378410 DOI: 10.1016/j.asjsur.2024.02.002] [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: 03/31/2023] [Revised: 01/11/2024] [Accepted: 02/07/2024] [Indexed: 02/22/2024] Open
Abstract
Renal cell carcinoma (RCC) is a type of tumor with high morbidity and recurrence rates. The application of circulating tumor cells (CTCs) in RCC remains controversial. Hence, we performed a meta-analysis to elucidate the diagnostic and prognostic value of CTCs in RCC. To obtain a precise conclusion, a systematic search was conducted in Pubmed, Cochrane Database, Embase and Web of Science up to Dec 01, 2022. We also further identified the references in relevant studies. The diagnostic accuracy variables (sensitivity, specificity) and odds ratios (ORs) with corresponding 95% confidence intervals (CIs) were used to access precise of CTCs and relationship between CTCs and disease stages, respectively. Heterogeneity test, sensitivity analysis, meta-regression and publishing bias were also applied. A total of 12 studies involving 767 patients were considered to be included in the final meta-analysis. The results revealed that the overall sensitivity, specificity of CTC detection in RCC were 45% (95%CI, 32-60%) and 99% (95%CI, 97-100%), respectively. In subgroup analysis, diagnostic sensitivity of CTCs in clear cell renal cell carcinoma (ccRCC) (69%, 95%CI; 50-88%) was significantly higher than other RCC subtypes (34%, 95%CI; 21-48%) (p<0.05). Meanwhile, advanced diseases (stage III-IV) were more likely to find CTCs than localized diseases (stage I-II) (OR, 2.29; 95%CI, 1.37-3.83; p = 0.002). This systematic review and meta-analysis demonstrated that CTC detection could be considered as a promising auxiliary diagnostic and staging method for RCC, especially ccRCC subtype. Meanwhile, the presence of cytokeratin-positive CTCs is highly likely associated with increased risk of poor prognosis in RCC.
Collapse
Affiliation(s)
- Liang Cao
- Department of Urology, The First People's Hospital of Neijiang, No. 1866 Han'an Avenue, Shizhong, Neijiang, 641000, China.
| | - Wenming Yang
- Department of Pediatric Urology, West China Second University Hospital, Sichuan University, No. 20 Section 3 of Renmin South Road, Chengdu, 610041, China
| | - Xiang Zhao
- Department of Otolaryngology, The First People's Hospital of Neijiang, No. 1866 Han'an Avenue, Shizhong, Neijiang, 641000, China
| | - Zhibin Chen
- Department of Urology, The First People's Hospital of Neijiang, No. 1866 Han'an Avenue, Shizhong, Neijiang, 641000, China
| |
Collapse
|
64
|
Hou Y, Meng X, Zhou X. Systematically Evaluating Cell-Free DNA Fragmentation Patterns for Cancer Diagnosis and Enhanced Cancer Detection via Integrating Multiple Fragmentation Patterns. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308243. [PMID: 38881520 PMCID: PMC11321639 DOI: 10.1002/advs.202308243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 04/12/2024] [Indexed: 06/18/2024]
Abstract
Cell-free DNA (cfDNA) fragmentation patterns have immense potential for early cancer detection. However, the definition of fragmentation varies, ranging from the entire genome to specific genomic regions. These patterns have not been systematically compared, impeding broader research and practical implementation. Here, 1382 plasma cfDNA sequencing samples from 8 cancer types are collected. Considering that cfDNA within open chromatin regions is more susceptible to fragmentation, 10 fragmentation patterns within open chromatin regions as features and employed machine learning techniques to evaluate their performance are examined. All fragmentation patterns demonstrated discernible classification capabilities, with the end motif showing the highest diagnostic value for cross-validation. Combining cross and independent validation results revealed that fragmentation patterns that incorporated both fragment length and coverage information exhibited robust predictive capacities. Despite their diagnostic potential, the predictive power of these fragmentation patterns is unstable. To address this limitation, an ensemble classifier via integrating all fragmentation patterns is developed, which demonstrated notable improvements in cancer detection and tissue-of-origin determination. Further functional bioinformatics investigations on significant feature intervals in the model revealed its impressive ability to identify critical regulatory regions involved in cancer pathogenesis.
Collapse
Affiliation(s)
- Yuying Hou
- Hubei Key Laboratory of Agricultural BioinformaticsCollege of InformaticsHuazhong Agricultural UniversityWuhan430070China
| | - Xiang‐Yu Meng
- Hubei Key Laboratory of Agricultural BioinformaticsCollege of InformaticsHuazhong Agricultural UniversityWuhan430070China
- Health Science CenterHubei Minzu UniversityEnshi445000China
- Hubei Provincial Clinical Medical Research Center for NephropathyHubei Minzu UniversityEnshi445000China
| | - Xionghui Zhou
- Hubei Key Laboratory of Agricultural BioinformaticsCollege of InformaticsHuazhong Agricultural UniversityWuhan430070China
- Key Laboratory of Smart Farming for Agricultural AnimalsMinistry of Agriculture and Rural AffairsWuhan430070China
| |
Collapse
|
65
|
Kim SH, Weigelt B. Circulating cell-free (cf)DNA analysis: Current technologies and applications in gynecologic cancer. Gynecol Oncol Rep 2024; 54:101431. [PMID: 38947418 PMCID: PMC11211888 DOI: 10.1016/j.gore.2024.101431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/04/2024] [Accepted: 06/09/2024] [Indexed: 07/02/2024] Open
Abstract
Cell-free DNA (cfDNA) analysis has several promising clinical applications in the management of cancer patients, with clinical validity established in different types of solid tumors (e.g., lung, breast, and colon cancer). Cancers harbor unique genetic alterations that can be detected in the plasma and other bodily fluids of cancer patients, constituting an alternate source of tumor-derived DNA. Technologic advances and wide-spread availability of next-generation sequencing (NGS) have made sequencing analysis of circulating tumor DNA (ctDNA) possible, employing both off-the-shelf and personalized tumor-informed panels. Tumor size, disease burden and high-grade histologic types have been shown to correlate with ctDNA levels across multiple solid cancer types. Detection of tumor-derived genetic alterations in plasma-derived cfDNA can facilitate diagnosis, guide treatment selection, and serve as a biomarker for treatment response and prognostication. Molecular residual disease (MRD) is at the forefront of cfDNA analysis, with implications in treatment de-escalation/ escalation in the neoadjuvant and adjuvant settings. The development of cfDNA analysis in early detection of cancers is under active investigation. Proof-of-principles studies in gynecologic cancers have demonstrated feasibility and potential for innovation in cancers lacking specific biomarkers, including the tracking of human papillomavirus (HPV) cfDNA in patients with cervical cancer. In this review, we outline the assays currently available for cfDNA sequencing/ ctDNA detection, the role of cfDNA analysis in clinical decision-making and the current status and potential clinical uses of cfDNA research in gynecologic cancers.
Collapse
Affiliation(s)
- Sarah H Kim
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Britta Weigelt
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
66
|
Save T, Neal RD, Round T. A multi-cancer early detection (MCED) test: clinical update for GPs. Br J Gen Pract 2024; 74:380-382. [PMID: 39054076 PMCID: PMC11299699 DOI: 10.3399/bjgp24x739113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024] Open
Affiliation(s)
- Tanvi Save
- School of Life Course & Population Sciences, King's College London, London
| | - Richard D Neal
- APEx (Exeter Collaboration for Academic Primary Care), Faculty of Health & Life Sciences, University of Exeter, Exeter
| | - Thomas Round
- School of Life Course & Population Sciences, King's College London, London
| |
Collapse
|
67
|
Pallocca M, Betti M, Baldinelli S, Palombo R, Bucci G, Mazzarella L, Tonon G, Ciliberto G. Clinical bioinformatics desiderata for molecular tumor boards. Brief Bioinform 2024; 25:bbae447. [PMID: 39297878 PMCID: PMC11411775 DOI: 10.1093/bib/bbae447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 06/28/2024] [Accepted: 08/30/2024] [Indexed: 09/26/2024] Open
Abstract
Clinical Bioinformatics is a knowledge framework required to interpret data of medical interest via computational methods. This area became of dramatic importance in precision oncology, fueled by cancer genomic profiling: most definitions of Molecular Tumor Boards require the presence of bioinformaticians. However, all available literature remained rather vague on what are the specific needs in terms of digital tools and expertise to tackle and interpret genomics data to assign novel targeted or biomarker-driven targeted therapies to cancer patients. To fill this gap, in this article, we present a catalog of software families and human skills required for the tumor board bioinformatician, with specific examples of real-world applications associated with each element presented.
Collapse
Affiliation(s)
- Matteo Pallocca
- Institute of Experimental Endocrinology and Oncology, National Research Council, Via Sergio Pansini, 5, 80131 Naples, Italy
| | - Martina Betti
- Biostatistics, Bioinformatics and Clinical Trial Center, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53, 00144 Rome, Italy
| | - Sara Baldinelli
- Biostatistics, Bioinformatics and Clinical Trial Center, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53, 00144 Rome, Italy
| | - Ramona Palombo
- Institute of Experimental Endocrinology and Oncology, National Research Council, Via Sergio Pansini, 5, 80131 Naples, Italy
| | - Gabriele Bucci
- Center for OMICS Sciences, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132 Milan, Italy
| | - Luca Mazzarella
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, IRCCS IEO - European Institute of Oncology, Via Adamello 16, 20139 Milan, Italy
- Department of Experimental Oncology, IRCCS IEO - European Institute of Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Giovanni Tonon
- Functional Genomics of Cancer Unit, Division of Experimental Oncology, and Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Via Olgettina, 60, 20132 Milan, Italy
| | - Gennaro Ciliberto
- Scientific Direction, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53, 00144 Rome, Italy
| |
Collapse
|
68
|
Liu L, Xiong Y, Zheng Z, Huang L, Song J, Lin Q, Tang B, Wong KC. AutoCancer as an automated multimodal framework for early cancer detection. iScience 2024; 27:110183. [PMID: 38989460 PMCID: PMC11233972 DOI: 10.1016/j.isci.2024.110183] [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: 12/22/2023] [Revised: 03/21/2024] [Accepted: 06/01/2024] [Indexed: 07/12/2024] Open
Abstract
Current studies in early cancer detection based on liquid biopsy data often rely on off-the-shelf models and face challenges with heterogeneous data, as well as manually designed data preprocessing pipelines with different parameter settings. To address those challenges, we present AutoCancer, an automated, multimodal, and interpretable transformer-based framework. This framework integrates feature selection, neural architecture search, and hyperparameter optimization into a unified optimization problem with Bayesian optimization. Comprehensive experiments demonstrate that AutoCancer achieves accurate performance in specific cancer types and pan-cancer analysis, outperforming existing methods across three cohorts. We further demonstrated the interpretability of AutoCancer by identifying key gene mutations associated with non-small cell lung cancer to pinpoint crucial factors at different stages and subtypes. The robustness of AutoCancer, coupled with its strong interpretability, underscores its potential for clinical applications in early cancer detection.
Collapse
Affiliation(s)
- Linjing Liu
- Department of Computer Science, City University of Hong Kong, Kowloon Tong, Hong Kong SAR
| | - Ying Xiong
- Department of Computer Science, Harbin Institute of Technology (Shenzhen), Shenzhen, China
| | - Zetian Zheng
- Department of Computer Science, City University of Hong Kong, Kowloon Tong, Hong Kong SAR
| | - Lei Huang
- Department of Computer Science, City University of Hong Kong, Kowloon Tong, Hong Kong SAR
| | - Jiangning Song
- Monash Biomedicine Discovery Institute and Monash Data Futures Institute, Monash University, Melbourne, VIC 3800, Australia
| | - Qiuzhen Lin
- College of Computer Science and Software Engineering, Shenzhen University, Shenzhen, China
| | - Buzhou Tang
- Department of Computer Science, Harbin Institute of Technology (Shenzhen), Shenzhen, China
| | - Ka-Chun Wong
- Department of Computer Science, City University of Hong Kong, Kowloon Tong, Hong Kong SAR
| |
Collapse
|
69
|
Li G, Zhang S, Zou Y, Ai H, Zheng X, Qian K, Lei C, Fu W. The therapeutic potential of exosomes in immunotherapy. Front Immunol 2024; 15:1424081. [PMID: 39040108 PMCID: PMC11260647 DOI: 10.3389/fimmu.2024.1424081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 06/24/2024] [Indexed: 07/24/2024] Open
Abstract
Exosomes are found in various tissues of the body and carry abundant contents including nucleic acids, proteins, and metabolites, which continuously flow between cells of various tissues and mediate important intercellular communication. In addition, exosomes from different cellular sources possess different physiopathological immunomodulatory effects, which are closely related to the immune regeneration of normal or abnormal organs and tissues. Here, we focus on the mechanistic interactions between exosomes and the human immune system, introduce the immuno-regenerative therapeutic potential of exosomes in common clinical immune-related diseases, such as infectious diseases, autoimmune diseases, and tumors, and reveal the safety and efficacy of exosomes as a novel cell-free immune regenerative therapy.
Collapse
Affiliation(s)
- Guangyao Li
- Department of Biophysics, College of Basic Medical Sciences, Naval Medical University (Second Military Medical University), Shanghai, China
- Department of Biomedical Engineering, College of Basic Medical Sciences, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Shuyi Zhang
- Department of Biophysics, College of Basic Medical Sciences, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Yitan Zou
- Department of Biomedical Engineering, College of Basic Medical Sciences, Naval Medical University (Second Military Medical University), Shanghai, China
- Department of Respiratory and Critical Care Medicine, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Hongru Ai
- Department of Biomedical Engineering, College of Basic Medical Sciences, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Xinya Zheng
- Department of Biomedical Engineering, College of Basic Medical Sciences, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Kewen Qian
- Department of Biomedical Engineering, College of Basic Medical Sciences, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Changhai Lei
- Department of Biophysics, College of Basic Medical Sciences, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Wenyan Fu
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
70
|
Shi J, Li R, Wang Y, Zhang C, Lyu X, Wan Y, Yu Z. Detection of lung cancer through SERS analysis of serum. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 314:124189. [PMID: 38569385 DOI: 10.1016/j.saa.2024.124189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/11/2024] [Accepted: 03/24/2024] [Indexed: 04/05/2024]
Abstract
Early detection and postoperative assessment are crucial for improving overall survival among lung cancer patients. Here, we report a non-invasive technique that integrates Raman spectroscopy with machine learning for the detection of lung cancer. The study encompassed 88 postoperative lung cancer patients, 73 non-surgical lung cancer patients, and 68 healthy subjects. The primary aim was to explore variations in serum metabolism across these cohorts. Comparative analysis of average Raman spectra was conducted, while principal component analysis was employed for data visualization. Subsequently, the augmented dataset was used to train convolutional neural networks (CNN) and Resnet models, leading to the development of a diagnostic framework. The CNN model exhibited superior performance, as verified by the receiver operating characteristic curve. Notably, postoperative patients demonstrated an increased likelihood of recurrence, emphasizing the crucial need for continuous postoperative monitoring. In summary, the integration of Raman spectroscopy with CNN-based classification shows potential for early detection and postoperative assessment of lung cancer.
Collapse
Affiliation(s)
- Jiamin Shi
- Department of Thoracic Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Shenyang 110042, People's Republic of China; School of Physics, Dalian University of Technology, Dalian, 116023, People's Republic of China
| | - Rui Li
- Department of Thoracic Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Shenyang 110042, People's Republic of China; School of Physics, Dalian University of Technology, Dalian, 116023, People's Republic of China; State Key Laboratory of Fine Chemicals, Frontier Science Center for Smart Materials, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, People's Republic of China
| | - Yuchen Wang
- Department of Thoracic Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Shenyang 110042, People's Republic of China; School of Physics, Dalian University of Technology, Dalian, 116023, People's Republic of China
| | - Chenlei Zhang
- Department of Thoracic Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Shenyang 110042, People's Republic of China
| | - Xiaohong Lyu
- Department of Radiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121000, People's Republic of China
| | - Yuan Wan
- The Pq Laboratory of BiomeDx/Rx, Department of Biomedical Engineering, Binghamton University, Vestal, 13850 NY, USA
| | - Zhanwu Yu
- Department of Thoracic Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Shenyang 110042, People's Republic of China.
| |
Collapse
|
71
|
Chen Z, Tian Z, Wang Z, Wang Z, Liu W, Gu Q, Liu S, Wu Y. A Portable Device for in Situ Noninvasive Monitoring of Cell Secretions and Communications with Fluorescence and Nanochannel Electrochemistry. Anal Chem 2024; 96:9218-9227. [PMID: 38781682 DOI: 10.1021/acs.analchem.4c01380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
In situ monitoring of cell secretions and communications plays a fundamental role in screening of disease diagnostic biomarkers and drugs. Quantitative detection of cell secretions and monitoring of intercellular communication have been separately reported, which often rely on target labeling or complex pretreatment steps, inevitably causing damage to the target. Simultaneous in situ noninvasive detection of cell secretions and monitoring of intercellular communication are challenging and have never been reported. Herein, we smartly developed a portable device for in situ label-free monitoring of cell secretions and communications with fluorescence and ion-transport-based nanochannel electrochemistry. Based on the dual signal mode, a series of nonelectroactive secretions were sensitively and accurately quantified. The detection limits for VEGF, MUC1, and ATP were 3.84 pg/mL, 32.7 pg/mL, and 47.4 fM (3σ/S), which were 1/3.9, 1/1.1, and 1/41 of those of commercial ELISA kits, respectively. More interestingly, under the released secretions, the gradual opening of the nanochannel connected the two cells in the left and right chambers of the device; thus, the secretion mediated intercellular communication can be monitored. The proposed platform may provide a promising tool for understanding the mechanism of intercellular communication and discovering new therapeutic targets.
Collapse
Affiliation(s)
- Zixuan Chen
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Zhaoyan Tian
- State Key Laboratory for Macromolecule Drugs and Large-Scale Manufacturing, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252059, China
| | - Zhi Wang
- Wuxi Institute of Inspection, Testing and Certification, Wuxi 214125, China
| | - Zhaohan Wang
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Weiwei Liu
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Qinglin Gu
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Songqin Liu
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Yafeng Wu
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| |
Collapse
|
72
|
Nickel B, Heiss R, Shih P, Gram EG, Copp T, Taba M, Moynihan R, Zadro J. Social Media Promotion of Health Tests With Potential for Overdiagnosis or Overuse: Protocol for a Content Analysis. JMIR Res Protoc 2024; 13:e56899. [PMID: 38833693 PMCID: PMC11185923 DOI: 10.2196/56899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/19/2024] [Accepted: 05/02/2024] [Indexed: 06/06/2024] Open
Abstract
BACKGROUND In recent years, social media have emerged as important spaces for commercial marketing of health tests, which can be used for the screening and diagnosis of otherwise generally healthy people. However, little is known about how health tests are promoted on social media, whether the information provided is accurate and balanced, and if there is transparency around conflicts of interest. OBJECTIVE This study aims to understand and quantify how social media is being used to discuss or promote health tests with the potential for overdiagnosis or overuse to generally healthy people. METHODS Content analysis of social media posts on the anti-Mullerian hormone test, whole-body magnetic resonance imaging scan, multicancer early detection, testosterone test, and gut microbe test from influential international social media accounts on Instagram and TikTok. The 5 tests have been identified as having the following criteria: (1) there are evidence-based concerns about overdiagnosis or overuse, (2) there is evidence or concerns that the results of tests do not lead to improved health outcomes for generally healthy people and may cause harm or waste, and (3) the tests are being promoted on social media to generally healthy people. English language text-only posts, images, infographics, articles, recorded videos including reels, and audio-only posts are included. Posts from accounts with <1000 followers as well as stories, live videos, and non-English posts are excluded. Using keywords related to the test, the top posts were searched and screened until there were 100 eligible posts from each platform for each test (total of 1000 posts). Data from the caption, video, and on-screen text are being summarized and extracted into a Microsoft Excel (Microsoft Corporation) spreadsheet and included in the analysis. The analysis will take a combined inductive approach when generating key themes and a deductive approach using a prespecified framework. Quantitative data will be analyzed in Stata SE (version 18.0; Stata Corp). RESULTS Data on Instagram and TikTok have been searched and screened. Analysis has now commenced. The findings will be disseminated via publications in peer-reviewed international medical journals and will also be presented at national and international conferences in late 2024 and 2025. CONCLUSIONS This study will contribute to the limited evidence base on the nature of the relationship between social media and the problems of overdiagnosis and overuse of health care services. This understanding is essential to develop strategies to mitigate potential harm and plan solutions, with the aim of helping to protect members of the public from being marketed low-value tests, becoming patients unnecessarily, and taking resources away from genuine needs within the health system. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) DERR1-10.2196/56899.
Collapse
Affiliation(s)
- Brooke Nickel
- Sydney School of Public Health, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Raffael Heiss
- Center for Social & Health Innovation, Management Centre Innsbruk, Innsbruk, Austria
| | - Patti Shih
- Australian Centre for Health Engagement Evidence and Values, School of Health and Society, University of Wollongong, Wollongong, Australia
| | - Emma Grundtvig Gram
- Center for General Practice, Department of Public Health, University of Copenhagen, Denmark, Australia
| | - Tessa Copp
- Sydney School of Public Health, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Melody Taba
- Sydney School of Public Health, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Ray Moynihan
- Institute for Evidence-Based Healthcare, Faculty of Health Sciences and Medicine, Bond University, Gold Coast, Australia
| | - Joshua Zadro
- Sydney School of Public Health, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| |
Collapse
|
73
|
Suartz CV, Martinez LM, Cordeiro MD, Botelho LAA, Gallutti FP, Mota JM, Leite KRM, Toren P, Nahas WC, Ribeiro-Filho LA. Honing the Hunt: A Comprehensive Review of Cell-free Tumor DNA to Predict Neoadjuvant Therapy Efficacy in Bladder Cancer. Clin Genitourin Cancer 2024; 22:102087. [PMID: 38688207 DOI: 10.1016/j.clgc.2024.102087] [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: 12/16/2023] [Accepted: 03/29/2024] [Indexed: 05/02/2024]
Abstract
OBJECTIVE To provide an updated view on the role of cell-free DNA as a predictor of pathological response to neoadjuvant therapy in patients with muscle-invasive bladder cancer. METHODS A systematic review was conducted from September 2023 to October 2023. Selected studies from the MEDLINE and clinical trial databases were critically analyzed regarding the clinical efficacy of cell-free DNA as a predictive instrument after neoadjuvant therapy in bladder cancer. The methodological quality assessment was based on the QUADAS-2 tool. RESULTS In this systematic review, we analyzed 5 studies encompassing a cumulative patient cohort of 780 individuals diagnosed with muscle-invasive bladder cancer, with a median follow-up ranging from 6 to 23 months. Among these studies, 4 primarily focused on detecting and analyzing circulating tumor DNA in plasma, while 1 study uniquely utilized cell-free tumor DNA in urine samples. The diagnostic accuracy of cell-free DNA in plasma ranges from 79% to 100%, indicating a variable yet significant predictive capability. In contrast, the study utilizing urinary cell-free DNA demonstrated an accuracy of 81% in predicting treatment response post-neoadjuvant chemotherapy. CONCLUSION Cell-free DNA is emerging as a valuable biomarker for predicting response to neoadjuvant chemotherapy in patients with muscle-invasive bladder tumors.
Collapse
Affiliation(s)
- Caio V Suartz
- Division of Urology, Institute of Cancer of São Paulo, University of São Paulo, Brazil; Division of Urology, Department of Surgery, CHU de Québec - Université Laval, Quebec City, QC, Canada.
| | - Lucas Motta Martinez
- Division of Urology, Institute of Cancer of São Paulo, University of São Paulo, Brazil
| | - Maurício D Cordeiro
- Division of Urology, Institute of Cancer of São Paulo, University of São Paulo, Brazil
| | - Luiz A A Botelho
- Division of Urology, Institute of Cancer of São Paulo, University of São Paulo, Brazil
| | - Fábio P Gallutti
- Division of Urology, Institute of Cancer of São Paulo, University of São Paulo, Brazil
| | - José M Mota
- Genitourinary Medical Oncology Service, Institute of Cancer of São Paulo, University of São Paulo, Brazil
| | - Katia R M Leite
- Laboratory of Medical Investigation (LIM55), Urology Department, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Paul Toren
- Division of Urology, Department of Surgery, CHU de Québec - Université Laval, Quebec City, QC, Canada
| | - William C Nahas
- Division of Urology, Institute of Cancer of São Paulo, University of São Paulo, Brazil
| | | |
Collapse
|
74
|
Edsjö A, Russnes HG, Lehtiö J, Tamborero D, Hovig E, Stenzinger A, Rosenquist R. High-throughput molecular assays for inclusion in personalised oncology trials - State-of-the-art and beyond. J Intern Med 2024; 295:785-803. [PMID: 38698538 DOI: 10.1111/joim.13785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
In the last decades, the development of high-throughput molecular assays has revolutionised cancer diagnostics, paving the way for the concept of personalised cancer medicine. This progress has been driven by the introduction of such technologies through biomarker-driven oncology trials. In this review, strengths and limitations of various state-of-the-art sequencing technologies, including gene panel sequencing (DNA and RNA), whole-exome/whole-genome sequencing and whole-transcriptome sequencing, are explored, focusing on their ability to identify clinically relevant biomarkers with diagnostic, prognostic and/or predictive impact. This includes the need to assess complex biomarkers, for example microsatellite instability, tumour mutation burden and homologous recombination deficiency, to identify patients suitable for specific therapies, including immunotherapy. Furthermore, the crucial role of biomarker analysis and multidisciplinary molecular tumour boards in selecting patients for trial inclusion is discussed in relation to various trial concepts, including drug repurposing. Recognising that today's exploratory techniques will evolve into tomorrow's routine diagnostics and clinical study inclusion assays, the importance of emerging technologies for multimodal diagnostics, such as proteomics and in vivo drug sensitivity testing, is also discussed. In addition, key regulatory aspects and the importance of patient engagement in all phases of a clinical trial are described. Finally, we propose a set of recommendations for consideration when planning a new precision cancer medicine trial.
Collapse
Affiliation(s)
- Anders Edsjö
- Department of Clinical Genetics, Pathology and Molecular Diagnostics, Office for Medical Services, Region Skåne, Lund, Sweden
- Division of Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Hege G Russnes
- Department of Pathology, Oslo University Hospital, Oslo, Norway
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Janne Lehtiö
- Department of Oncology and Pathology, Karolinska Institutet, Science for Life Laboratory, Stockholm, Sweden
- Cancer genomics and proteomics, Karolinska University Hospital, Solna, Sweden
| | - David Tamborero
- Department of Oncology and Pathology, Karolinska Institutet, Science for Life Laboratory, Stockholm, Sweden
| | - Eivind Hovig
- Center for Bioinformatics, Department of Informatics, University of Oslo, Oslo, Norway
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Albrecht Stenzinger
- Institute of Pathology, Division of Molecular Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Richard Rosenquist
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Clinical Genetics and Genomics, Karolinska University Hospital, Solna, Sweden
| |
Collapse
|
75
|
Wu X, Liu H, Chen J, Tao L, Zhou S, Shen X. Aptamer biorecognition and gold nanoshell-mediated fluorescence quenching for glycoprotein analysis and breast cancer diagnosis. Microchem J 2024; 201:110719. [DOI: 10.1016/j.microc.2024.110719] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2025]
|
76
|
Chen Y, Wang X, Luo S, Dai C, Wu Y, Zhao J, Liu W, Kong D, Yang Y, Geng L, Liu Y, Wei D. Electrically Oriented Antibodies on Transistor for Monitoring Several Copies of Methylated DNA. Anal Chem 2024; 96:8300-8307. [PMID: 38747393 DOI: 10.1021/acs.analchem.3c04670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
An antibody transistor is a promising biosensing platform for the diagnosis and monitoring of various diseases. Nevertheless, the low concentration and short half-life of biomarkers require biodetection at the trace-molecule level, which remains a challenge for existing antibody transistors. Herein, we demonstrate a graphene field-effect transistor (gFET) with electrically oriented antibody probes (EOA-gFET) for monitoring several copies of methylated DNA. The electric field confines the orientation of antibody probes on graphene and diminishes the distance between graphene and methylated DNAs captured by antibodies, generating more induced charges on graphene and amplifying the electric signal. EOA-gFET realizes a limit of detection (LoD) of ∼0.12 copy μL-1, reaching the lowest LoD reported before. EOA-gFET shows a distinguishable signal for liver cancer clinical serum samples within ∼6 min, which proves its potential as a powerful tool for disease screening and diagnosis.
Collapse
Affiliation(s)
- Yiheng Chen
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200433, China
- Laboratory of Molecular Materials and Devices, Department of Material Science, Fudan University, Shanghai 200433, China
| | - Xuejun Wang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200433, China
- Laboratory of Molecular Materials and Devices, Department of Material Science, Fudan University, Shanghai 200433, China
| | - Shi Luo
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200433, China
- Laboratory of Molecular Materials and Devices, Department of Material Science, Fudan University, Shanghai 200433, China
| | - Changhao Dai
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200433, China
- Laboratory of Molecular Materials and Devices, Department of Material Science, Fudan University, Shanghai 200433, China
| | - Yungen Wu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200433, China
- Laboratory of Molecular Materials and Devices, Department of Material Science, Fudan University, Shanghai 200433, China
| | - Junhong Zhao
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200433, China
- Laboratory of Molecular Materials and Devices, Department of Material Science, Fudan University, Shanghai 200433, China
| | - Wentao Liu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200433, China
- Laboratory of Molecular Materials and Devices, Department of Material Science, Fudan University, Shanghai 200433, China
| | - Derong Kong
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200433, China
- Laboratory of Molecular Materials and Devices, Department of Material Science, Fudan University, Shanghai 200433, China
| | - Yuetong Yang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200433, China
- Laboratory of Molecular Materials and Devices, Department of Material Science, Fudan University, Shanghai 200433, China
| | - Li Geng
- Department of Special Treatment, Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China
| | - Yunqi Liu
- Laboratory of Molecular Materials and Devices, Department of Material Science, Fudan University, Shanghai 200433, China
- Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Dacheng Wei
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai 200433, China
- Laboratory of Molecular Materials and Devices, Department of Material Science, Fudan University, Shanghai 200433, China
| |
Collapse
|
77
|
Rakshit I, Mandal S, Pal S, Bhattacharjee P. Advancements in bladder cancer detection: a comprehensive review on liquid biopsy and cell-free DNA analysis. THE NUCLEUS 2024. [DOI: 10.1007/s13237-024-00494-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 05/04/2024] [Indexed: 01/06/2025] Open
|
78
|
Rosas-Alonso R, Colmenarejo-Fernández J, Pernía O, Burdiel M, Rodríguez-Antolín C, Losantos-García I, Rubio T, Moreno-Velasco R, Esteban-Rodríguez I, Martínez-Marín V, Yubero P, Costa-Fraga N, Díaz-Lagares A, López-López R, Díaz-Martin E, García JF, Sánchez CV, Gandía-González ML, Moreno-Bueno G, de Castro J, de Cáceres II. Evaluation of the clinical use of MGMT methylation in extracellular vesicle-based liquid biopsy as a tool for glioblastoma patient management. Sci Rep 2024; 14:11398. [PMID: 38762534 PMCID: PMC11102540 DOI: 10.1038/s41598-024-62061-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 05/13/2024] [Indexed: 05/20/2024] Open
Abstract
Glioblastoma (GB) is a devastating tumor of the central nervous system characterized by a poor prognosis. One of the best-established predictive biomarker in IDH-wildtype GB is O6-methylguanine-DNA methyltransferase (MGMT) methylation (mMGMT), which is associated with improved treatment response and survival. However, current efforts to monitor GB patients through mMGMT detection have proven unsuccessful. Small extracellular vesicles (sEVs) hold potential as a key element that could revolutionize clinical practice by offering new possibilities for liquid biopsy. This study aimed to determine the utility of sEV-based liquid biopsy as a predictive biomarker and disease monitoring tool in patients with IDH-wildtype GB. Our findings show consistent results with tissue-based analysis, achieving a remarkable sensitivity of 85.7% for detecting mMGMT in liquid biopsy, the highest reported to date. Moreover, we suggested that liquid biopsy assessment of sEV-DNA could be a powerful tool for monitoring disease progression in IDH-wildtype GB patients. This study highlights the critical significance of overcoming molecular underdetection, which can lead to missed treatment opportunities and misdiagnoses, possibly resulting in ineffective therapies. The outcomes of our research significantly contribute to the field of sEV-DNA-based liquid biopsy, providing valuable insights into tumor tissue heterogeneity and establishing it as a promising tool for detecting GB biomarkers. These results have substantial implications for advancing predictive and therapeutic approaches in the context of GB and warrant further exploration and validation in clinical settings.
Collapse
Affiliation(s)
- Rocío Rosas-Alonso
- Cancer Epigenetics Laboratory, INGEMM, La Paz University Hospital, Paseo La Castellana 261, Edificio Bloque Quirúrgico Planta-2, 28046, Madrid, Spain.
- Biomarkers and Experimental Therapeutics in Cancer, IdiPAZ, Madrid, Spain.
| | - Julian Colmenarejo-Fernández
- Cancer Epigenetics Laboratory, INGEMM, La Paz University Hospital, Paseo La Castellana 261, Edificio Bloque Quirúrgico Planta-2, 28046, Madrid, Spain
- Biomarkers and Experimental Therapeutics in Cancer, IdiPAZ, Madrid, Spain
| | - Olga Pernía
- Cancer Epigenetics Laboratory, INGEMM, La Paz University Hospital, Paseo La Castellana 261, Edificio Bloque Quirúrgico Planta-2, 28046, Madrid, Spain
- Biomarkers and Experimental Therapeutics in Cancer, IdiPAZ, Madrid, Spain
| | - Miranda Burdiel
- Cancer Epigenetics Laboratory, INGEMM, La Paz University Hospital, Paseo La Castellana 261, Edificio Bloque Quirúrgico Planta-2, 28046, Madrid, Spain
- Biomarkers and Experimental Therapeutics in Cancer, IdiPAZ, Madrid, Spain
| | - Carlos Rodríguez-Antolín
- Cancer Epigenetics Laboratory, INGEMM, La Paz University Hospital, Paseo La Castellana 261, Edificio Bloque Quirúrgico Planta-2, 28046, Madrid, Spain
- Biomarkers and Experimental Therapeutics in Cancer, IdiPAZ, Madrid, Spain
| | | | - Tania Rubio
- Cancer Epigenetics Laboratory, INGEMM, La Paz University Hospital, Paseo La Castellana 261, Edificio Bloque Quirúrgico Planta-2, 28046, Madrid, Spain
- Biomarkers and Experimental Therapeutics in Cancer, IdiPAZ, Madrid, Spain
| | - Rocío Moreno-Velasco
- Cancer Epigenetics Laboratory, INGEMM, La Paz University Hospital, Paseo La Castellana 261, Edificio Bloque Quirúrgico Planta-2, 28046, Madrid, Spain
- Biomarkers and Experimental Therapeutics in Cancer, IdiPAZ, Madrid, Spain
| | - Isabel Esteban-Rodríguez
- Biomarkers and Experimental Therapeutics in Cancer, IdiPAZ, Madrid, Spain
- Department of Pathology, La Paz University Hospital, Madrid, Spain
| | | | - Paloma Yubero
- Department of Medical Oncology, La Paz University Hospital, Madrid, Spain
| | - Nicolas Costa-Fraga
- Cancer Epigenomics Laboratory, Epigenomics Unit, Translational Medical Oncology Group (ONCOMET), IDIS, University Clinical Hospital of Santiago (CHUS/SERGAS), Santiago de Compostela, Spain
| | - Angel Díaz-Lagares
- Cancer Epigenomics Laboratory, Epigenomics Unit, Translational Medical Oncology Group (ONCOMET), IDIS, University Clinical Hospital of Santiago (CHUS/SERGAS), Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Rafael López-López
- Cancer Epigenomics Laboratory, Epigenomics Unit, Translational Medical Oncology Group (ONCOMET), IDIS, University Clinical Hospital of Santiago (CHUS/SERGAS), Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
- Department of Medical Oncology, University Hospital Complex of Santiago de Compostela, Santiago de Compostela, Spain
| | | | - Juan F García
- MD Anderson International Foundation, Madrid, Spain
- Department of Pathology, MD Anderson Cancer Center, Madrid, Spain
| | | | | | - Gema Moreno-Bueno
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
- MD Anderson International Foundation, Madrid, Spain
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas 'Alberto Sols' (CSIC-UAM), IdiPAZ, Madrid, Spain
| | - Javier de Castro
- Biomarkers and Experimental Therapeutics in Cancer, IdiPAZ, Madrid, Spain
- Department of Medical Oncology, La Paz University Hospital, Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Inmaculada Ibánez de Cáceres
- Cancer Epigenetics Laboratory, INGEMM, La Paz University Hospital, Paseo La Castellana 261, Edificio Bloque Quirúrgico Planta-2, 28046, Madrid, Spain.
- Biomarkers and Experimental Therapeutics in Cancer, IdiPAZ, Madrid, Spain.
| |
Collapse
|
79
|
Drejeriene I, Cicenas S, Stanciute D, Krasauskas A, Gruode J. Detection of Oncogene Hotspot Mutations in Female NSCLC Tumor DNA and Cell-Free DNA. Cancers (Basel) 2024; 16:1770. [PMID: 38730722 PMCID: PMC11083023 DOI: 10.3390/cancers16091770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/16/2024] [Accepted: 04/21/2024] [Indexed: 05/13/2024] Open
Abstract
Non-small-cell lung cancer (NSCLC) is the most prevalent type of lung cancer, with extensively characterized mutational spectra. Several biomarkers (such as EGFR, BRAF, KRAS gene mutations, etc.) have emerged as predictive and prognostic markers for NSCLC. Unfortunately, the quality of the available tumor biopsy and/or cytology material is not always adequate to perform the necessary molecular testing, prompting the search for alternatives. Cell-free DNA (cfDNA) found in plasma is emerging as a highly promising avenue or a supplementary method for assessing the efficacy of cancer treatments. This is especially valuable in instances where conventional biopsy specimens, like formalin-fixed, paraffin-embedded (FFPE), or freshly frozen tumor tissues prove inadequate for conducting molecular pathology analyses subsequent to the initial diagnostic procedures. By leveraging cfDNA from plasma, clinicians gain an additional tool to gauge the effectiveness of cancer therapies, thereby enhancing their ability to optimize tailored treatment strategies. In this study, 51 Lithuanian females with NSCLC were analyzed, with adenocarcinoma being the predominant pathology diagnosis in 40 cases (78%). Target mutations were identified in 38 out of 51 patients (74.5%) in tumor tissue samples, while in plasma samples, they were identified in only 10 patients' samples (19.6%). Even though we did not have enough voluminous plasma samples in our study, gene mutations were detected in plasma from ten women, three of whom were diagnosed with early stages of lung cancer (stages I and II). For these patients, the following mutations were detected: deletion in exon 19 of the EGFR gene and single nucleotide polymorphisms in the TP53 and MET genes. All other women were diagnosed with stages III or IV of lung cancer. This indicates that the later stages of cancer contribute more cfDNA in plasma, making extraction less complicated.
Collapse
Affiliation(s)
- Ieva Drejeriene
- Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
- Klaipeda University Hospital, 92288 Klaipeda, Lithuania;
| | | | | | - Arnoldas Krasauskas
- Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
- National Cancer Institute, 08406 Vilnius, Lithuania
| | - Jurate Gruode
- Klaipeda University Hospital, 92288 Klaipeda, Lithuania;
- Faculty of Health Science, Klaipeda University, 92294 Klaipeda, Lithuania
| |
Collapse
|
80
|
Conca V, Ciracì P, Boccaccio C, Minelli A, Antoniotti C, Cremolini C. Waiting for the "liquid revolution" in the adjuvant treatment of colon cancer patients: a review of ongoing trials. Cancer Treat Rev 2024; 126:102735. [PMID: 38613871 DOI: 10.1016/j.ctrv.2024.102735] [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: 12/01/2023] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 04/15/2024]
Abstract
Since colon cancer has a high rate of shedding of tumour fragments into the blood, several research efforts are now focused on the investigation of the minimal residual disease through the detection of ctDNA to tailor the adjuvant therapy of colon cancer patients and optimize its cost/effectiveness balance. The negative prognostic impact of detectable ctDNA in patients' blood after radical surgery for colon cancer is well established. Several clinical trials adopting heterogeneous designs and techniques are now ongoing to translate promises into daily practice by answering five general questions: i) is a ctDNA-guided decision making efficacious in the post-operative management of colon cancer patients? ii) are de-escalation strategies possible in ctDNA-negative cases? iii) are escalation strategies useful to improve the prognosis of ctDNA-positive patients? iv) when MRD is identified at the end of the adjuvant chemotherapy, is another post-adjuvant systemic therapy efficacious? v) can we exploit ctDNA technologies in the follow up of colon cancer patients? This review focuses on currently ongoing trials and how their results may affect the ctDNA "liquid revolution" of early colon cancer.
Collapse
Affiliation(s)
- V Conca
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy; Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - P Ciracì
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy; Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - C Boccaccio
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy; Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - A Minelli
- Division of Medical Oncology, Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - C Antoniotti
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy; Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - C Cremolini
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy; Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy.
| |
Collapse
|
81
|
Li M, Xie S, Hou T, Shao T, Kuang J, Liu C, Qu Y, Lu C, Liu J, Liu X, Zhu L, Zhu L. Circulating Tumor DNA Profiling Approach Based on In Silico Background Elimination Guides Chemotherapy in Nasopharyngeal Carcinoma. Clin Pharmacol Ther 2024; 115:993-1006. [PMID: 38037868 DOI: 10.1002/cpt.3125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 11/17/2023] [Indexed: 12/02/2023]
Abstract
Circulating tumor DNA (ctDNA) analysis increasingly provides a promising minimally invasive alternative to tissue biopsies in precision oncology. However, there are no ctDNA analysis approaches available in nasopharyngeal carcinoma (NPC) and current methods of ctDNA mutation profiling have limited resolution because of the high background noise and false-positive rate caused by benign variants in plasma cell-free DNA (cfDNA), majorly generated during clonal hematopoiesis. Although personalized parallel white blood cell genome sequencing suppresses the noise of clonal hematopoiesis variances, the system cost and complexity restrict its extensive application in clinical settings. We developed Matched WBC Genome sequencing Independent CtDNA profiling (MaGIC) approaches, which synergically integrated a ctDNA capturing panel for a hybrid capture cfDNA deep sequencing, in silico background elimination, and a reliable readout measurement. We profiled the ctDNAs of 80 plasma samples from 40 patients with NPC before and during chemotherapy by MaGICs. In addition, the public cfDNA sequencing data and The Cancer Genome Atlas project data were analyzed by MaGICs to evaluate their application in other scenarios of patient classification. The MaGIC version-2 has the ability to predict the chemosensitivity of patients with NPC with high accuracy by utilizing a single sample of liquid biopsy from each patient prior to a standardized treatment regimen. Moreover, both versions of MaGICs are of ideal performance in the diagnosis of patients with prostate cancer by liquid biopsy and prognosis prediction of multiple cancers by tissue biopsy. This study has the potential to enhance the sensitivity and expand the application scope of ctDNA detection, independently of other paired genome sequencing methods. As a result, it might further increase the clinical utilization of liquid biopsy based on ctDNA.
Collapse
Affiliation(s)
- Ming Li
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, China
- Jiuquan Satellite Launch Centre, Jiuquan, China
| | - Sisi Xie
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, China
| | - Tao Hou
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Tong Shao
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, China
| | - Jingyu Kuang
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, China
| | - Chuanyang Liu
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, China
| | - Ying Qu
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, China
| | - Chenyu Lu
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, China
| | - Jiali Liu
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, China
| | - Xianling Liu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Lingyun Zhu
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, China
| | - Lvyun Zhu
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, China
| |
Collapse
|
82
|
Spagnolo CC, Pepe F, Ciappina G, Nucera F, Ruggeri P, Squeri A, Speranza D, Silvestris N, Malapelle U, Santarpia M. Circulating biomarkers as predictors of response to immune checkpoint inhibitors in NSCLC: Are we on the right path? Crit Rev Oncol Hematol 2024; 197:104332. [PMID: 38580184 DOI: 10.1016/j.critrevonc.2024.104332] [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: 02/14/2024] [Accepted: 03/25/2024] [Indexed: 04/07/2024] Open
Abstract
Immune checkpoints inhibitors (ICIs) have markedly improved the therapeutic management of advanced NSCLC and, more recently, they have demonstrated efficacy also in the early-stage disease. Despite better survival outcomes with ICIs compared to standard chemotherapy, a large proportion of patients can derive limited clinical benefit from these agents. So far, few predictive biomarkers, including the programmed death-ligand 1 (PD-L1), have been introduced in clinical practice. Therefore, there is an urgent need to identify novel biomarkers to select patients for immunotherapy, to improve efficacy and avoid unnecessary toxicity. A deeper understanding of the mechanisms involved in antitumor immunity and advances in the field of liquid biopsy have led to the identification of a wide range of circulating biomarkers that could potentially predict response to immunotherapy. Herein, we provide an updated overview of these circulating biomarkers, focusing on emerging data from clinical studies and describing modern technologies used for their detection.
Collapse
Affiliation(s)
- Calogera Claudia Spagnolo
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina 98122, Italy
| | - Francesco Pepe
- Department of Public Health, University of Naples Federico II, Via S. Pansini, Naples 80131, Italy
| | - Giuliana Ciappina
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina 98122, Italy
| | - Francesco Nucera
- Respiratory Medicine Unit, Department of Biomedical Sciences, Dentistry and Morphological and Functional Imaging (BIOMORF), University of Messina, Messina 98122, Italy
| | - Paolo Ruggeri
- Respiratory Medicine Unit, Department of Biomedical Sciences, Dentistry and Morphological and Functional Imaging (BIOMORF), University of Messina, Messina 98122, Italy
| | - Andrea Squeri
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina 98122, Italy
| | - Desirèe Speranza
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina 98122, Italy
| | - Nicola Silvestris
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina 98122, Italy
| | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Via S. Pansini, Naples 80131, Italy
| | - Mariacarmela Santarpia
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina 98122, Italy.
| |
Collapse
|
83
|
Eguchi H, Takenaka Y, Tanaka H, Suzuki M, Horie M, Kanai H, Seo Y, Ogawa K, Yachida S, Inohara H. Survival of Metastatic Human Papillomavirus (HPV)-Related Head and Neck Cancer Receiving Platinum-Based Triplet Induction Chemotherapy and Relevance of Circulating Tumor HPV DNA. Cureus 2024; 16:e60547. [PMID: 38887331 PMCID: PMC11181740 DOI: 10.7759/cureus.60547] [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] [Accepted: 05/18/2024] [Indexed: 06/20/2024] Open
Abstract
Objectives We aimed to examine the effectiveness of platinum-based triplet induction chemotherapy in metastatic squamous cell carcinoma of the head and neck (HNSCC) at diagnosis in terms of tumor human papillomavirus (HPV) status and the clinical relevance of circulating tumor HPV DNA (ctHPVDNA) during induction chemotherapy. Methods Twenty-one patients were included. ctHPVDNA was longitudinally quantified using optimized digital PCR in a subset of patients. Results HPV-related HNSCC patients (N=7) had a significantly better response to induction chemotherapy than HPV-unrelated HNSCC patients (N=14) (complete or partial response rate, 100% vs. 36%, P = 0.007). Following induction chemotherapy, more HPV-related HNSCC patients than HPV-unrelated patients received radiotherapy (86% vs. 36%, P = 0.06). With a median follow-up of 26 months in surviving patients, the two-year overall survival was 86% in HPV-related HNSCC patients and 43% in HPV-unrelated HNSCC patients (P = 0.04). In two patients, ctHPVDNA levels drastically decreased after the first cycle of induction chemotherapy but turned to continuous increase after the second cycle, suggesting the acquisition of drug resistance by the end of the second cycle. Radiographic imaging after induction chemotherapy failed to identify the drug resistance. In one patient, ctHPVDNA decreased gradually but remained detectable after induction chemotherapy despite no radiographic residual disease. ctHPVDNA became undetectable during radiotherapy. Conclusion HPV-related HNSCC patients with distant metastasis at diagnosis should be treated definitively. The ctHPVDNA level reflects real-time disease activity. ctHPVDNA monitoring during induction chemotherapy could help the decision-making of the therapeutic strategy.
Collapse
Affiliation(s)
| | | | | | | | | | - Haruka Kanai
- Head and Neck Surgery, Osaka University, Suita, JPN
| | - Yuji Seo
- Radiation Oncology, Osaka University, Suita, JPN
| | | | | | - Hidenori Inohara
- Otorhinolaryngology - Head and Neck Surgery, Osaka University Graduate School of Medicine, Suita, JPN
| |
Collapse
|
84
|
De Sota RE, Quake SR, Sninsky JJ, Toden S. Decoding bioactive signals of the RNA secretome: the cell-free messenger RNA catalogue. Expert Rev Mol Med 2024; 26:e12. [PMID: 38682644 PMCID: PMC11140549 DOI: 10.1017/erm.2024.12] [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: 08/08/2023] [Revised: 01/18/2024] [Accepted: 03/18/2024] [Indexed: 05/01/2024]
Abstract
Despite gene-expression profiling being one of the most common methods to evaluate molecular dysregulation in tissues, the utilization of cell-free messenger RNA (cf-mRNA) as a blood-based non-invasive biomarker analyte has been limited compared to other RNA classes. Recent advancements in low-input RNA-sequencing and normalization techniques, however, have enabled characterization as well as accurate quantification of cf-mRNAs allowing direct pathological insights. The molecular profile of the cell-free transcriptome in multiple diseases has subsequently been characterized including, prenatal diseases, neurological disorders, liver diseases and cancers suggesting this biological compartment may serve as a disease agnostic platform. With mRNAs packaged in a myriad of extracellular vesicles and particles, these signals may be used to develop clinically actionable, non-invasive disease biomarkers. Here, we summarize the recent scientific developments of extracellular mRNA, biology of extracellular mRNA carriers, clinical utility of cf-mRNA as disease biomarkers, as well as proposed functions in cell and tissue pathophysiology.
Collapse
Affiliation(s)
- Rhys E. De Sota
- Superfluid Dx., 259 E Grand Avenue, South San Francisco, CA 94080, USA
| | - Stephen R. Quake
- Department of Bioengineering and Department of Applied Physics, Stanford University, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - John J. Sninsky
- Superfluid Dx., 259 E Grand Avenue, South San Francisco, CA 94080, USA
| | - Shusuke Toden
- Superfluid Dx., 259 E Grand Avenue, South San Francisco, CA 94080, USA
| |
Collapse
|
85
|
Shiota M, Matsubara N, Kato T, Eto M, Osawa T, Abe T, Shinohara N, Nishimoto K, Yasumizu Y, Tanaka N, Oya M, Fujisawa T, Horasawa S, Nakamura Y, Yoshino T, Nonomura N. Genomic profiling and clinical utility of circulating tumor DNA in metastatic prostate cancer: SCRUM-Japan MONSTAR SCREEN project. BJC REPORTS 2024; 2:28. [PMID: 39516321 PMCID: PMC11523993 DOI: 10.1038/s44276-024-00049-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 02/10/2024] [Accepted: 02/18/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Circulating tumor DNA (ctDNA) testing has emerged as a novel tool for cancer precision medicine. This study investigated the genomic profiling and clinical utility of ctDNA in metastatic prostate cancer. METHODS This is a nation-wide prospective observational study. Patients treated with systemic treatment for metastatic castration-sensitive prostate cancer (mCSPC) and metastatic castration-resistant prostate cancer (mCRPC) were included. ctDNA was analyzed using FoundationOne Liquid®CDx at enrollment. In a subset of patients, ctDNA after disease progression and tissue prior to the initiation of treatment were examined using FoundationOne Liquid®CDx and FoundationOne®CDx, respectively. RESULTS The frequency of AR alterations and homologous recombination repair (HRR) defect was higher in mCRPC compared with mCSPC. Tumor mutational burden was correlated between tissue and ctDNA at pre-treatment, as well as ctDNA between at pre-treatment and at post-treatment. Patients with HRR defect were associated with shorter time to castration resistance in androgen deprivation therapy/combined androgen blockade, but not in androgen receptor pathway inhibitor, compared with patients without HRR defect in mCSPC. Time to treatment failure in patients with AR amplification or AR mutation was shorter compared with patients without AR alterations in mCRPC. CONCLUSIONS This study revealed valuable findings for the clinical care of metastatic prostate cancer. Especially, predictive factors such as HRR defect in mCSPC should be validated in the future.
Collapse
Affiliation(s)
- Masaki Shiota
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Kyushu, Japan.
| | - Nobuaki Matsubara
- Department Medical Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Taigo Kato
- Department of Urology, Osaka University Graduate School of Medicine, Yamadaoka, Japan
| | - Masatoshi Eto
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Kyushu, Japan
| | - Takahiro Osawa
- Department of Urology, Graduate School of Medicine Hokkaido University, Hokkaido, Japan
| | - Takashige Abe
- Department of Urology, Graduate School of Medicine Hokkaido University, Hokkaido, Japan
| | - Nobuo Shinohara
- Department of Urology, Graduate School of Medicine Hokkaido University, Hokkaido, Japan
| | - Koshiro Nishimoto
- Department of Uro-Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Yota Yasumizu
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Nobuyuki Tanaka
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Mototsugu Oya
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Takao Fujisawa
- Department Head and Neck Medical Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Satoshi Horasawa
- Translational Research Support Office, National Cancer Center Hospital East, Chiba, Japan
| | - Yoshiaki Nakamura
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Norio Nonomura
- Department of Urology, Osaka University Graduate School of Medicine, Yamadaoka, Japan
| |
Collapse
|
86
|
Gupta S, Singh B, Abhishek R, Gupta S, Sachan M. The emerging role of liquid biopsy in oral squamous cell carcinoma detection: advantages and challenges. Expert Rev Mol Diagn 2024; 24:311-331. [PMID: 38607339 DOI: 10.1080/14737159.2024.2340997] [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: 12/11/2023] [Accepted: 04/05/2024] [Indexed: 04/13/2024]
Abstract
INTRODUCTION Oral Squamous Cell Carcinoma (OSCC), the sixth most widespread malignancy in the world, accounts for 90% of all cases of oral cancer. The primary risk factors are tobacco chewing, alcohol consumption, viral infection, and genetic modifications. OSCC has a high morbidity rate due to the lack of early diagnostic methods. Nowadays, liquid biopsy plays a vital role in the initial diagnosis of oral cancer. ctNAs extracted from saliva and serum/plasma offer meaningful insights into tumor genetics and dynamics. The interplay of these elements in saliva and serum/plasma showcases their significance in advancing noninvasive, effective OSCC detection and monitoring. AREAS COVERED This review mainly focused on the role of liquid biopsy as an emerging point in the diagnosis and prognosis of OSCC and the current advancements and challenges associated with liquid biopsy. EXPERT OPINION Liquid biopsy is regarded as a new, minimally invasive, real-time monitoring tool for cancer diagnosis and prognosis. Many biomolecules found in bodily fluids, including ctDNA, ctRNA, CTCs, and EVs, are significant biomarkers to identify cancer in its early stages. Despite these groundbreaking strides, challenges persist. Standardization of sample collection, isolation, processing, and detection methods is imperative for ensuring result reproducibility across diverse studies.
Collapse
Affiliation(s)
- Sudha Gupta
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, India
| | - Brijesh Singh
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, India
| | - Rajul Abhishek
- Department of Surgical Oncology, Motilal Nehru Medical College, Prayagraj, India
| | - Sameer Gupta
- Department of Surgical Oncology, King George Medical University, Lucknow, India
| | - Manisha Sachan
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, India
| |
Collapse
|
87
|
Ewongwo A, Hui C, Moding EJ. Opportunity in Complexity: Harnessing Molecular Biomarkers and Liquid Biopsies for Personalized Sarcoma Care. Semin Radiat Oncol 2024; 34:195-206. [PMID: 38508784 DOI: 10.1016/j.semradonc.2023.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
Due to their rarity and complexity, sarcomas represent a substantial therapeutic challenge. However, the incredible diversity within and across sarcoma subtypes presents an opportunity for personalized care to maximize efficacy and limit toxicity. A deeper understanding of the molecular alterations that drive sarcoma development and treatment response has paved the way for molecular biomarkers to shape sarcoma treatment. Genetic, transcriptomic, and protein biomarkers have become critical tools for diagnosis, prognostication, and treatment selection in patients with sarcomas. In the future, emerging biomarkers like circulating tumor DNA analysis offer the potential to improve early detection, monitoring response to treatment, and identifying mechanisms of resistance to personalize sarcoma treatment. Here, we review the current state of molecular biomarkers for sarcomas and highlight opportunities and challenges for the implementation of new technologies in the future.
Collapse
Affiliation(s)
- Agnes Ewongwo
- Department of Radiation Oncology, Stanford University, Stanford, CA
| | - Caressa Hui
- Department of Radiation Oncology, Stanford University, Stanford, CA
| | - Everett J Moding
- Department of Radiation Oncology, Stanford University, Stanford, CA.; Stanford Cancer Institute, Stanford University, Stanford, CA..
| |
Collapse
|
88
|
Ando T, Yokoi T, Manri C, Anazawa T, Ishida T. Improvement of limit of detection in primer extension-based multiplexed mutation assay using capillary electrophoresis. ANAL SCI 2024; 40:731-739. [PMID: 38319561 PMCID: PMC10961290 DOI: 10.1007/s44211-024-00508-8] [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: 11/03/2023] [Accepted: 01/04/2024] [Indexed: 02/07/2024]
Abstract
One of the challenges in liquid biopsy for early cancer detection is ascribed to the fact that mutation DNA often represents an extremely small ratio of less than 1% compared to wild-type genes in blood. However, in conventional fragment analysis with capillary electrophoresis (CE), the detectable allele frequency could be about 5%. In this work, we developed an original reagent-based fragment analysis with single base extension (SBE) reactions for cancer-associated mutation assay using a commercially available CE device, and investigated on a possibility of improvement of limit of detection (LOD) for genetic mutation. First, after adjustment of reagent conditions for the SBE reactions, the linear relationship between gene template concentration and fluorescence intensity was obtained from 1 to 100 fmol of target genes. Next, from the results of an experiment to detect mutation EGFR L858R at abundance ratios of mutant type to wild type (100-fmol template) of 0, 1, 5, and 10%, it was shown that the target gene can be detected with LOD of 0.33%. This high sensitivity was realized in part by separating fluorescently labeled substrates into an individual tube for an each-colored SBE reaction. Moreover, mutations EGFR L858R and KRAS G12V were simultaneously detected at sensitivities equivalent to LODs of 0.57 and 0.47%, respectively. These results indicate that < 1% of mutations in multiplex gene mutations can be simultaneously detected, and that possibility suggests that the developed method can be used in clinical practice for detecting cancers.
Collapse
Affiliation(s)
- Takahiro Ando
- Research & Development Group, Hitachi, Ltd., 1-280 Higashi-Koigakubo, Kokubunji-shi, Tokyo, 185-8601, Japan.
| | - Takahide Yokoi
- Research & Development Group, Hitachi, Ltd., 1-280 Higashi-Koigakubo, Kokubunji-shi, Tokyo, 185-8601, Japan
| | - Chihiro Manri
- Research & Development Group, Hitachi, Ltd., 1-280 Higashi-Koigakubo, Kokubunji-shi, Tokyo, 185-8601, Japan
| | - Takashi Anazawa
- Research & Development Group, Hitachi, Ltd., 1-280 Higashi-Koigakubo, Kokubunji-shi, Tokyo, 185-8601, Japan
| | - Takeshi Ishida
- Research & Development Group, Hitachi, Ltd., 1-280 Higashi-Koigakubo, Kokubunji-shi, Tokyo, 185-8601, Japan
| |
Collapse
|
89
|
Allen TA. The Role of Circulating Tumor Cells as a Liquid Biopsy for Cancer: Advances, Biology, Technical Challenges, and Clinical Relevance. Cancers (Basel) 2024; 16:1377. [PMID: 38611055 PMCID: PMC11010957 DOI: 10.3390/cancers16071377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/23/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
Cancer remains a leading cause of mortality worldwide, with metastasis significantly contributing to its lethality. The metastatic spread of tumor cells, primarily through the bloodstream, underscores the importance of circulating tumor cells (CTCs) in oncological research. As a critical component of liquid biopsies, CTCs offer a non-invasive and dynamic window into tumor biology, providing invaluable insights into cancer dissemination, disease progression, and response to treatment. This review article delves into the recent advancements in CTC research, highlighting their emerging role as a biomarker in various cancer types. We explore the latest technologies and methods for CTC isolation and detection, alongside novel approaches to characterizing their biology through genomics, transcriptomics, proteomics, and epigenetic profiling. Additionally, we examine the clinical implementation of these findings, assessing how CTCs are transforming the landscape of cancer diagnosis, prognosis, and management. By offering a comprehensive overview of current developments and potential future directions, this review underscores the significance of CTCs in enhancing our understanding of cancer and in shaping personalized therapeutic strategies, particularly for patients with metastatic disease.
Collapse
|
90
|
Fu X, Luo Z, Deng Y, LaFramboise W, Bartlett D, Schwartz R. Marker selection strategies for circulating tumor DNA guided by phylogenetic inference. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.21.585352. [PMID: 38586041 PMCID: PMC10996527 DOI: 10.1101/2024.03.21.585352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Motivation Blood-based profiling of tumor DNA ("liquid biopsy") has offered great prospects for non-invasive early cancer diagnosis, treatment monitoring, and clinical guidance, but require further advances in computational methods to become a robust quantitative assay of tumor clonal evolution. We propose new methods to better characterize tumor clonal dynamics from circulating tumor DNA (ctDNA), through application to two specific questions: 1) How to apply longitudinal ctDNA data to refine phylogeny models of clonal evolution, and 2) how to quantify changes in clonal frequencies that may be indicative of treatment response or tumor progression. We pose these questions through a probabilistic framework for optimally identifying maximum likelihood markers and applying them to characterizing clonal evolution. Results We first estimate a distribution over plausible clonal lineage models, using bootstrap samples over pre-treatment tissue-based sequence data. We then refine these lineage models and the clonal frequencies they imply over successive longitudinal samples. We use the resulting framework for modeling and refining tree distributions to pose a set of optimization problems to select ctDNA markers to maximize measures of utility capturing ability to solve the two questions of reducing uncertain in phylogeny models or quantifying clonal frequencies given the models. We tested our methods on synthetic data and showed them to be effective at refining distributions of tree models and clonal frequencies so as to minimize measures of tree distance relative to the ground truth. Application of the tree refinement methods to real tumor data further demonstrated their effectiveness in refining a clonal lineage model and assessing its clonal frequencies. The work shows the power of computational methods to improve marker selection, clonal lineage reconstruction, and clonal dynamics profiling for more precise and quantitative assays of tumor progression. Availability https://github.com/CMUSchwartzLab/Mase-phi.git. Contact russells@andrew.cmu.edu.
Collapse
Affiliation(s)
- Xuecong Fu
- Department of Biological Sciences, Carnegie Mellon University, 5000 Forbes Avenue, 15217, Pittsburgh, PA, USA
| | - Zhicheng Luo
- Department of Biological Sciences, Carnegie Mellon University, 5000 Forbes Avenue, 15217, Pittsburgh, PA, USA
| | - Yueqian Deng
- Ray and Stephanie Lane Computational Biology Department, Carnegie Mellon University, 5000 Forbes Avenue, 15217, Pittsburgh, PA, USA
| | - William LaFramboise
- Allegheny Health Network Cancer Institute, Allegheny Health Network, 320 East North Avenue, 15212, Pittsburgh, PA, USA
| | - David Bartlett
- Allegheny Health Network Cancer Institute, Allegheny Health Network, 320 East North Avenue, 15212, Pittsburgh, PA, USA
| | - Russell Schwartz
- Department of Biological Sciences, Carnegie Mellon University, 5000 Forbes Avenue, 15217, Pittsburgh, PA, USA
- Ray and Stephanie Lane Computational Biology Department, Carnegie Mellon University, 5000 Forbes Avenue, 15217, Pittsburgh, PA, USA
| |
Collapse
|
91
|
Jia L, Hu W, Yan X, Shao J, Guo Y, Zhang A, Yu L, Zhou Y, Li Y, Ren L, Dong D. Soluble Periostin is a potential surveillance biomarker for early and long-term response to chemotherapy in advanced breast cancer. Cancer Cell Int 2024; 24:109. [PMID: 38504252 PMCID: PMC10953259 DOI: 10.1186/s12935-024-03298-1] [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: 09/03/2023] [Accepted: 03/08/2024] [Indexed: 03/21/2024] Open
Abstract
BACKGROUND Noninvasive biomarkers for the assessment of response to chemotherapy in advanced breast cancer (BCa) are essential for optimized therapeutic decision-making. We evaluated the potential of soluble Periostin (POSTN) in circulation as a novel biomarker for chemotherapy efficacy monitoring. METHODS Two hundred and thirty-one patients with different stages of BCa were included. Of those patients, 58 patients with inoperable metastatic disease receiving HER2-targeted or non-targeted chemotherapy were enrolled to assess the performances of markers in recapitulating the chemotherapy efficacy assessed by imaging. POSTN, together with CA153 or CEA at different time points (C0, C2, and C4) were determined. RESULTS POSTN levels were significantly associated with tumor volume (P < 0.0001) and TNM stages (P < 0.0001) of BCa. For early monitoring, dynamics of POSTN could recapitulate the chemotherapy efficacy among all molecular subtypes (Cohen's weighted kappa = 0.638, P < 0.0001), much better than that of carcinoembryonic antigen (CEA) and cancer antigen 153 (CA15-3). For early partial response, superior performance of POSTN was observed (Cohen's weighted kappa = 0.827, P < 0.0001) in cases with baseline levels above 17.19 ng/mL. For long-term monitoring, the POSTN response was observed to be strongly consistent with the course of the disease. Moreover, progression free survival analysis showed that patients experienced a significant early decrease of POSTN tended to obtain more benefits from the treatments. CONCLUSIONS The current study suggests that soluble POSTN is an informative serum biomarker to complement the current clinical approaches for early and long-term chemotherapy efficacy monitoring in advanced BCa.
Collapse
Affiliation(s)
- Li Jia
- Department of Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Tianjin's Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Huanhuxi Road, Hexi District, Tianjin, 300060, PR China
| | - Wenwei Hu
- Department of Gastroenterology, Chinese People's Liberation Army Rocket Force Characteristic Medical Center, Beijing, 100088, PR China
| | - Xu Yan
- Department of Anesthesiology, Chinese People's Liberation Army Rocket Force Characteristic Medical Center, Beijing, 100088, PR China
| | - Jie Shao
- Department of Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Tianjin's Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Huanhuxi Road, Hexi District, Tianjin, 300060, PR China
| | - Yuhong Guo
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Tianjin's Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, PR China
| | - Aimin Zhang
- Department of Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Tianjin's Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Huanhuxi Road, Hexi District, Tianjin, 300060, PR China
| | - Lianzi Yu
- Department of Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Tianjin's Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Huanhuxi Road, Hexi District, Tianjin, 300060, PR China
| | - Yunli Zhou
- Department of Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Tianjin's Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Huanhuxi Road, Hexi District, Tianjin, 300060, PR China
| | - Yueguo Li
- Department of Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Tianjin's Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Huanhuxi Road, Hexi District, Tianjin, 300060, PR China.
| | - Li Ren
- Department of Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Tianjin's Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Huanhuxi Road, Hexi District, Tianjin, 300060, PR China.
| | - Dong Dong
- Department of Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Tianjin's Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Huanhuxi Road, Hexi District, Tianjin, 300060, PR China.
| |
Collapse
|
92
|
Kim YJ, Rho WY, Park SM, Jun BH. Optical nanomaterial-based detection of biomarkers in liquid biopsy. J Hematol Oncol 2024; 17:10. [PMID: 38486294 PMCID: PMC10938695 DOI: 10.1186/s13045-024-01531-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 03/02/2024] [Indexed: 03/18/2024] Open
Abstract
Liquid biopsy, which is a minimally invasive procedure as an alternative to tissue biopsy, has been introduced as a new diagnostic/prognostic measure. By screening disease-related markers from the blood or other biofluids, it promises early diagnosis, timely prognostication, and effective treatment of the diseases. However, there will be a long way until its realization due to its conceptual and practical challenges. The biomarkers detected by liquid biopsy, such as circulating tumor cell (CTC) and circulating tumor DNA (ctDNA), are extraordinarily rare and often obscured by an abundance of normal cellular components, necessitating ultra-sensitive and accurate detection methods for the advancement of liquid biopsy techniques. Optical biosensors based on nanomaterials open an important opportunity in liquid biopsy because of their enhanced sensing performance with simple and practical properties. In this review article, we summarized recent innovations in optical nanomaterials to demonstrate the sensitive detection of protein, peptide, ctDNA, miRNA, exosome, and CTCs. Each study prepares the optical nanomaterials with a tailored design to enhance the sensing performance and to meet the requirements of each biomarker. The unique optical characteristics of metallic nanoparticles (NPs), quantum dots, upconversion NPs, silica NPs, polymeric NPs, and carbon nanomaterials are exploited for sensitive detection mechanisms. These recent advances in liquid biopsy using optical nanomaterials give us an opportunity to overcome challenging issues and provide a resource for understanding the unknown characteristics of the biomarkers as well as the mechanism of the disease.
Collapse
Affiliation(s)
- Young Jun Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 05029, Republic of Korea
| | - Won-Yeop Rho
- School of International Engineering and Science, Jeonbuk National University, Chonju, 54896, Republic of Korea
| | - Seung-Min Park
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, 637459, Singapore.
| | - Bong-Hyun Jun
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 05029, Republic of Korea.
| |
Collapse
|
93
|
Zhang S, Ma B, Liu Y, Shen Y, Li D, Liu S, Song F. Predicting locus-specific DNA methylation levels in cancer and paracancer tissues. Epigenomics 2024; 16:549-570. [PMID: 38477028 PMCID: PMC11158003 DOI: 10.2217/epi-2023-0114] [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: 04/03/2023] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Aim: To predict base-resolution DNA methylation in cancerous and paracancerous tissues. Material & methods: We collected six cancer DNA methylation datasets from The Cancer Genome Atlas and five cancer datasets from Gene Expression Omnibus and established machine learning models using paired cancerous and paracancerous tissues. Tenfold cross-validation and independent validation were performed to demonstrate the effectiveness of the proposed method. Results: The developed cross-tissue prediction models can substantially increase the accuracy at more than 68% of CpG sites and contribute to enhancing the statistical power of differential methylation analyses. An XGBoost model leveraging multiple correlating CpGs may elevate the prediction accuracy. Conclusion: This study provides a powerful tool for DNA methylation analysis and has the potential to gain new insights into cancer research from epigenetics.
Collapse
Affiliation(s)
- Shuzheng Zhang
- School of Information Science & Technology, Dalian Maritime University, Dalian, 116026, China
| | - Baoshan Ma
- School of Information Science & Technology, Dalian Maritime University, Dalian, 116026, China
| | - Yu Liu
- School of Information Science & Technology, Dalian Maritime University, Dalian, 116026, China
| | - Yiwen Shen
- School of Information Science & Technology, Dalian Maritime University, Dalian, 116026, China
| | - Di Li
- Department of Neuro Intervention, Dalian Medical University affiliated Dalian Municipal Central Hospital, Dalian, 116033, China
| | - Shuxin Liu
- Department of Nephrology, Dalian Medical University affiliated Dalian Municipal Central Hospital, Dalian, 116033, China
| | - Fengju Song
- Department of Epidemiology & Biostatistics, Key Laboratory of Molecular Cancer Epidemiology, Tianjin, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin, 300060, China
| |
Collapse
|
94
|
Cheng JC, Swarup N, Wong DTW, Chia D. A review on the impact of single-stranded library preparation on plasma cell-free diversity for cancer detection. Front Oncol 2024; 14:1332004. [PMID: 38511142 PMCID: PMC10951391 DOI: 10.3389/fonc.2024.1332004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 02/07/2024] [Indexed: 03/22/2024] Open
Abstract
In clinical oncology, cell-free DNA (cfDNA) has shown immense potential in its ability to noninvasively detect cancer at various stages and monitor the progression of therapy. Despite the rapid improvements in cfDNA liquid biopsy approaches, achieving the required sensitivity to detect rare tumor-derived cfDNA still remains a challenge. For next-generation sequencing, the perceived presentation of cfDNA is strongly linked to the extraction and library preparation protocols. Conventional double-stranded DNA library preparation (dsDNA-LP) focuses on assessing ~167bp double-stranded mononucleosomal (mncfDNA) and its other oligonucleosomal cell-free DNA counterparts in plasma. However, dsDNA-LP methods fail to include short, single-stranded, or nicked DNA in the final library preparation, biasing the representation of the actual cfDNA populations in plasma. The emergence of single-stranded library preparation (ssDNA-LP) strategies over the past decade has now allowed these other populations of cfDNA to be studied from plasma. With the use of ssDNA-LP, single-stranded, nicked, and ultrashort cfDNA can be comprehensively assessed for its molecular characteristics and clinical potential. In this review, we overview the current literature on applications of ssDNA-LP on plasma cfDNA from a potential cancer liquid biopsy perspective. To this end, we discuss the molecular principles of single-stranded DNA adapter ligation, how library preparation contributes to the understanding of native cfDNA characteristics, and the potential for ssDNA-LP to improve the sensitivity of circulating tumor DNA detection. Additionally, we review the current literature on the newly reported species of plasma ultrashort single-stranded cell-free DNA plasma, which appear biologically distinct from mncfDNA. We conclude with a discussion of future perspectives of ssDNA-LP for liquid biopsy endeavors.
Collapse
Affiliation(s)
- Jordan C. Cheng
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
- Stanford Cancer Institute, Stanford University, Stanford, CA, United States
| | - Neeti Swarup
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - David T. W. Wong
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - David Chia
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
95
|
Sacino AN, Chen H, Sahgal A, Bettegowda C, Rhines LD, Maralani P, Redmond KJ. Stereotactic body radiation therapy for spinal metastases: A new standard of care. Neuro Oncol 2024; 26:S76-S87. [PMID: 38437670 PMCID: PMC10911798 DOI: 10.1093/neuonc/noad225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024] Open
Abstract
Advancements in systemic therapies for patients with metastatic cancer have improved overall survival and, hence, the number of patients living with spinal metastases. As a result, the need for more versatile and personalized treatments for spinal metastases to optimize long-term pain and local control has become increasingly important. Stereotactic body radiation therapy (SBRT) has been developed to meet this need by providing precise and conformal delivery of ablative high-dose-per-fraction radiation in few fractions while minimizing risk of toxicity. Additionally, advances in minimally invasive surgical techniques have also greatly improved care for patients with epidural disease and/or unstable spines, which may then be combined with SBRT for durable local control. In this review, we highlight the indications and controversies of SBRT along with new surgical techniques for the treatment of spinal metastases.
Collapse
Affiliation(s)
- Amanda N Sacino
- Department of Neurosurgery, John Hopkins University, Baltimore, Maryland, USA
| | - Hanbo Chen
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Arjun Sahgal
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Chetan Bettegowda
- Department of Neurosurgery, John Hopkins University, Baltimore, Maryland, USA
| | - Laurence D Rhines
- Department of Neurosurgery, MD Anderson Cancer Center, Houston, Texas, USA
| | - Pejman Maralani
- Department of Medical Imaging, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Kristin J Redmond
- Department of Radiation and Molecular Oncology, John Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
96
|
Lippert J, Smith G, Appenzeller S, Landwehr LS, Prete A, Steinhauer S, Asia M, Urlaub H, Elhassan YS, Kircher S, Arlt W, Fassnacht M, Altieri B, Ronchi CL. Circulating cell-free DNA-based biomarkers for prognostication and disease monitoring in adrenocortical carcinoma. Eur J Endocrinol 2024; 190:234-247. [PMID: 38451242 DOI: 10.1093/ejendo/lvae022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 01/11/2024] [Accepted: 02/16/2024] [Indexed: 03/08/2024]
Abstract
OBJECTIVE Adrenocortical carcinoma (ACC) is a rare aggressive cancer with heterogeneous behaviour. Disease surveillance relies on frequent imaging, which comes with significant radiation exposure. The aim of the study was to investigate the role of circulating cell-free DNA (ccfDNA)-related biomarkers (BMs) for prognostication and monitoring of ACC. DESIGN AND METHODS We investigated 34 patients with ACC and 23 healthy subjects (HSs) as controls. Circulating cell-free DNA was extracted by commercial kits and ccfDNA concentrations were quantified by fluorimeter (BM1). Targeted sequencing was performed using a customized panel of 27 ACC-specific genes. Leucocyte DNA was used to discriminate somatic variants (BM2), while tumour DNA was sequenced in 22/34 cases for comparison. Serial ccfDNA samples were collected during follow-up in 19 ACC patients (median period 9 months) and analysed in relationship with standard radiological imaging. RESULTS Circulating cell-free DNA concentrations were higher in ACC than HS (mean ± SD, 1.15 ± 1.56 vs 0.05 ± 0.05 ng/µL, P < .0001), 96% of them being above the cut-off of 0.146 ng/µL (mean HS + 2 SD, positive BM1). At ccfDNA sequencing, 47% of ACC showed at least 1 somatic mutation (positive BM2). A combined ccfDNA-BM score was strongly associated with both progression-free and overall survival (hazard ratio [HR] = 2.63; 95% CI, 1.13-6.13; P = .010, and HR = 5.98; 95% CI, 2.29-15.6; P = .0001, respectively). During disease monitoring, positive BM2 showed the best specificity (100%) and sensitivity (67%) to detect ACC recurrence or progress compared with BM1. CONCLUSION ccfDNA-related BMs are frequently detected in ACC patients and represent a promising, minimally invasive tool to predict clinical outcome and complement surveillance imaging. Our findings will be validated in a larger cohort of ACCs with long-term follow-up.
Collapse
Affiliation(s)
- Juliane Lippert
- Division of Endocrinology and Diabetes, Department of Medicine, University Hospital, University of Wuerzburg, 97080 Wuerzburg, Germany
- Institute of Human Genetics, University of Wuerzburg, 97070 Wuerzburg, Germany
| | - Gabrielle Smith
- Institute of Metabolism and System Research, University of Birmingham, B152TT Birmingham, United Kingdom
| | - Silke Appenzeller
- Core Unit Bioinformatics, Comprehensive Cancer Center Mainfranken, University of Wuerzburg, 97070 Wuerzburg, Germany
| | - Laura-Sophie Landwehr
- Division of Endocrinology and Diabetes, Department of Medicine, University Hospital, University of Wuerzburg, 97080 Wuerzburg, Germany
| | - Alessandro Prete
- Institute of Metabolism and System Research, University of Birmingham, B152TT Birmingham, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism (CEDAM), Birmingham Health Partners, B152TT Birmingham, United Kingdom
- NIHR Birmingham Biomedical Research Centre, University of Birmingham, University Hospitals Birmingham NHS Foundation Trust, B152GW Birmingham, United Kingdom
| | - Sonja Steinhauer
- Division of Endocrinology and Diabetes, Department of Medicine, University Hospital, University of Wuerzburg, 97080 Wuerzburg, Germany
| | - Miriam Asia
- Endocrine Department, Queen Elizabeth Hospital Birmingham NHS Trust, B152GW Birmingham, United Kingdom
| | - Hanna Urlaub
- Division of Endocrinology and Diabetes, Department of Medicine, University Hospital, University of Wuerzburg, 97080 Wuerzburg, Germany
| | - Yasir S Elhassan
- Institute of Metabolism and System Research, University of Birmingham, B152TT Birmingham, United Kingdom
- Endocrine Department, Queen Elizabeth Hospital Birmingham NHS Trust, B152GW Birmingham, United Kingdom
| | - Stefan Kircher
- Department of Pathology, University of Wuerzburg, 97080 Wuerzburg, Germany
| | - Wiebke Arlt
- Institute of Metabolism and System Research, University of Birmingham, B152TT Birmingham, United Kingdom
- MRC Laboratory of Medical Sciences, W120TN London, United Kingdom
| | - Martin Fassnacht
- Division of Endocrinology and Diabetes, Department of Medicine, University Hospital, University of Wuerzburg, 97080 Wuerzburg, Germany
| | - Barbara Altieri
- Division of Endocrinology and Diabetes, Department of Medicine, University Hospital, University of Wuerzburg, 97080 Wuerzburg, Germany
| | - Cristina L Ronchi
- Institute of Metabolism and System Research, University of Birmingham, B152TT Birmingham, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism (CEDAM), Birmingham Health Partners, B152TT Birmingham, United Kingdom
| |
Collapse
|
97
|
Kim R, Mun B, Lim S, Park C, Kim J, Lim J, Jeong H, Son HY, Rho HW, Lim EK, Haam S. Colorimetric Detection of HER2-Overexpressing-Cancer-Derived Exosomes in Mouse Urine Using Magnetic-Polydiacetylene Nanoparticles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307262. [PMID: 37963850 DOI: 10.1002/smll.202307262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/31/2023] [Indexed: 11/16/2023]
Abstract
Breast cancer (BC) is a major global health problem, with ≈20-25% of patients overexpressing human epidermal growth factor receptor 2 (HER2), an aggressive marker, yet access to early detection and treatment varies across countries. A low-cost, equipment-free, and easy-to-use polydiacetylene (PDA)-based colorimetric sensor is developed for HER2-overexpressing cancer detection, designed for use in low- and middle-income countries (LMICs). PDA nanoparticles are first prepared through thin-film hydration. Subsequently, hydrophilic magnetic nanoparticles and HER2 antibodies are sequentially conjugated to them. The synthesized HER2-MPDA can be concentrated and separated by a magnetic field while inheriting the optical characteristics of PDA. The specific binding of HER2 antibody in HER2-MPDA to HER2 receptor in HER2-overexpressing exosomes causes a blue-to-red color change by altering the molecular structure of the PDA backbone. This colorimetric sensor can simultaneously separate and detect HER2-overexpressing exosomes. HER2-MPDA can detect HER2-overexpressing exosomes in the culture medium of HER2-overexpressing BC cells and in mouse urine samples from a HER2-overexpressing BC mouse model. It can selectively isolate and detect only HER2-overexpressing exosomes through magnetic separation, and its detection limit is found to be 8.5 × 108 particles mL-1. This colorimetric sensor can be used for point-of-care diagnosis of HER2-overexpressing BC in LMICs.
Collapse
Affiliation(s)
- Ryunhyung Kim
- Department of Chemical and Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Byeonggeol Mun
- Department of Chemical and Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Seongjae Lim
- Department of Chemical and Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Chaewon Park
- Department of Chemical and Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jinyoung Kim
- Department of Chemical and Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jaewook Lim
- Department of Chemical and Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Hyein Jeong
- Department of Chemical and Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Hye Young Son
- Department of Radiology, College of Medicine, Yonsei University, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Hyun Wook Rho
- Department of Radiology, College of Medicine, Yonsei University, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Eun-Kyung Lim
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
- Department of Nanobiotechnology, KRIBB School of Biotechnology, University of Science and Technology, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34113, Republic of Korea
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Seungjoo Haam
- Department of Chemical and Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| |
Collapse
|
98
|
Lehrich BM, Zhang J, Monga SP, Dhanasekaran R. Battle of the biopsies: Role of tissue and liquid biopsy in hepatocellular carcinoma. J Hepatol 2024; 80:515-530. [PMID: 38104635 PMCID: PMC10923008 DOI: 10.1016/j.jhep.2023.11.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/27/2023] [Accepted: 11/27/2023] [Indexed: 12/19/2023]
Abstract
The diagnosis and management of hepatocellular carcinoma (HCC) have improved significantly in recent years. With the introduction of immunotherapy-based combination therapy, there has been a notable expansion in treatment options for patients with unresectable HCC. Simultaneously, innovative molecular tests for early detection and management of HCC are emerging. This progress prompts a key question: as liquid biopsy techniques rise in prominence, will they replace traditional tissue biopsies, or will both techniques remain relevant? Given the ongoing challenges of early HCC detection, including issues with ultrasound sensitivity, accessibility, and patient adherence to surveillance, the evolution of diagnostic techniques is more relevant than ever. Furthermore, the accurate stratification of HCC is limited by the absence of reliable biomarkers which can predict response to therapies. While the advantages of molecular diagnostics are evident, their potential has not yet been fully harnessed, largely because tissue biopsies are not routinely performed for HCC. Liquid biopsies, analysing components such as circulating tumour cells, DNA, and extracellular vesicles, provide a promising alternative, though they are still associated with challenges related to sensitivity, cost, and accessibility. The early results from multi-analyte liquid biopsy panels are promising and suggest they could play a transformative role in HCC detection and management; however, comprehensive clinical validation is still ongoing. In this review, we explore the challenges and potential of both tissue and liquid biopsy, highlighting that these diagnostic methods, while distinct in their approaches, are set to jointly reshape the future of HCC management.
Collapse
Affiliation(s)
- Brandon M Lehrich
- Department of Pathology and Pittsburgh Liver Institute, University of Pittsburgh, School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Josephine Zhang
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University, Staford, CA, 94303, USA
| | - Satdarshan P Monga
- Department of Pathology and Pittsburgh Liver Institute, University of Pittsburgh, School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.
| | - Renumathy Dhanasekaran
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University, Staford, CA, 94303, USA.
| |
Collapse
|
99
|
Naegele S, Ruiz-Torres DA, Zhao Y, Goss D, Faden DL. Comparing the Diagnostic Performance of Quantitative PCR, Digital Droplet PCR, and Next-Generation Sequencing Liquid Biopsies for Human Papillomavirus-Associated Cancers. J Mol Diagn 2024; 26:179-190. [PMID: 38103593 PMCID: PMC10918646 DOI: 10.1016/j.jmoldx.2023.11.007] [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: 04/28/2023] [Revised: 08/29/2023] [Accepted: 11/13/2023] [Indexed: 12/19/2023] Open
Abstract
Human papillomavirus (HPV)-associated cancers, including oropharyngeal squamous cell carcinoma (HPV + OPSCC), cervical cancer, and squamous cell carcinoma of the anus (HPV + SCCA), release circulating tumor HPV DNA (ctHPVDNA) into the blood. The diagnostic performance of ctHPVDNA detection depends on the approaches used and the individual assay metrics. A comparison of these approaches has not been systematically performed to inform expected performance, which in turn affects clinical interpretation. A meta-analysis was performed using Ovid MEDLINE, Embase, and Web of Science Core Collection databases to assess the diagnostic accuracy of ctHPVDNA detection across cancer anatomic sites, detection platforms, and blood components. The population included patients with HPV + OPSCC, HPV-associated cervical cancer, and HPV + SCCA with pretreatment samples analyzed by quantitative PCR (qPCR), digital droplet PCR (ddPCR), or next-generation sequencing (NGS). Thirty-six studies involving 2986 patients met the inclusion criteria. The sensitivity, specificity, and quality of each study were assessed and pooled for each analysis. The sensitivity of ctHPVDNA detection was greatest with NGS, followed by ddPCR and then qPCR when pooling all studies, whereas specificity was similar (sensitivity: ddPCR > qPCR, P < 0.001; NGS > ddPCR, P = 0.014). ctHPVDNA from OPSCC was more easily detected compared with cervical cancer and SCCA, overall (P = 0.044). In conclusion, detection platform, anatomic site of the cancer, and blood component used affects ctHPVDNA detection and must be considered when interpreting results. Plasma NGS-based testing may be the most sensitive approach for ctHPVDNA overall.
Collapse
Affiliation(s)
- Saskia Naegele
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts
| | - Daniel A Ruiz-Torres
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts
| | - Yan Zhao
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts
| | - Deborah Goss
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts
| | - Daniel L Faden
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts; Broad Institute of MIT and Harvard, Cambridge, Massachusetts.
| |
Collapse
|
100
|
Grossi I, Assoni C, Lorini L, Smussi D, Gurizzan C, Grisanti S, Paderno A, Mattavelli D, Piazza C, Pelisenco IA, De Petro G, Salvi A, Bossi P. Evaluation of DNA methylation levels of SEPT9 and SHOX2 in plasma of patients with head and neck squamous cell carcinoma using droplet digital PCR. Oncol Rep 2024; 51:52. [PMID: 38299234 PMCID: PMC10865173 DOI: 10.3892/or.2024.8711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 12/12/2023] [Indexed: 02/02/2024] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the seventh most commonly diagnosed cancer globally. HNSCC develops from the mucosa of the oral cavity, pharynx and larynx. Methylation levels of septin 9 (SEPT9) and short stature homeobox 2 (SHOX2) genes in circulating cell‑free DNA (ccfDNA) are considered epigenetic biomarkers and have shown predictive value in preliminary reports in HNSCC. Liquid biopsy is a non‑invasive procedure that collects tumor‑derived molecules, including ccfDNA. In the present study, a droplet digital PCR (ddPCR)‑based assay was developed to detect DNA methylation levels of circulating SEPT9 and SHOX2 in the plasma of patients with HNSCC. The assay was first set up using commercial methylated and unmethylated DNA. The dynamic changes in the methylation levels of SEPT9 and SHOX2 were then quantified in 20 patients with HNSCC during follow‑up. The results highlighted: i) The ability of the ddPCR‑based assay to detect very low copies of methylated molecules; ii) the significant decrease in SEPT9 and SHOX2 methylation levels in the plasma of patients with HNSCC at the first time points of follow‑up with respect to T0; iii) a different trend of longitudinally DNA methylation variations in small groups of stratified patients. The absolute and precise quantification of SEPT9 and SHOX2 methylation levels in HNSCC may be useful for studies with translational potential.
Collapse
Affiliation(s)
- Ilaria Grossi
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, I-25123 Brescia, Italy
| | - Claudia Assoni
- Unit of Medical Oncology, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, ASST Spedali Civili of Brescia, University of Brescia, I-25123 Brescia, Italy
| | - Luigi Lorini
- Unit of Medical Oncology, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, ASST Spedali Civili of Brescia, University of Brescia, I-25123 Brescia, Italy
| | - Davide Smussi
- Unit of Medical Oncology, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, ASST Spedali Civili of Brescia, University of Brescia, I-25123 Brescia, Italy
| | - Cristina Gurizzan
- Unit of Medical Oncology, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, ASST Spedali Civili of Brescia, University of Brescia, I-25123 Brescia, Italy
| | - Salvatore Grisanti
- Unit of Medical Oncology, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, ASST Spedali Civili of Brescia, University of Brescia, I-25123 Brescia, Italy
| | - Alberto Paderno
- Unit of Otorhinolaryngology-Head and Neck Surgery, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, ASST Spedali Civili of Brescia, University of Brescia, I-25123 Brescia, Italy
| | - Davide Mattavelli
- Unit of Otorhinolaryngology-Head and Neck Surgery, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, ASST Spedali Civili of Brescia, University of Brescia, I-25123 Brescia, Italy
| | - Cesare Piazza
- Unit of Otorhinolaryngology-Head and Neck Surgery, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, ASST Spedali Civili of Brescia, University of Brescia, I-25123 Brescia, Italy
| | - Iulia Andreea Pelisenco
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, I-25123 Brescia, Italy
| | - Giuseppina De Petro
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, I-25123 Brescia, Italy
| | - Alessandro Salvi
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, I-25123 Brescia, Italy
| | - Paolo Bossi
- Unit of Medical Oncology, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, ASST Spedali Civili of Brescia, University of Brescia, I-25123 Brescia, Italy
| |
Collapse
|