1
|
Kaiglová A, Hockicková P, Bárdyová Z, Reháková R, Melnikov K, Kucharíková S. The chemotactic response of Caenorhabditis elegans represents a promising tool for the early detection of cancer. Discov Oncol 2024; 15:817. [PMID: 39707061 DOI: 10.1007/s12672-024-01721-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 12/18/2024] [Indexed: 12/23/2024] Open
Abstract
The nematode Caenorhabditis elegans, with its highly sensitive olfactory system, has emerged as a promising tool for testing chemotaxis. In the field of cancer diagnostics, there is a growing interest in the development of non-invasive screening methods for the detection of volatile organic compounds in a patient's urine. The objective of this study was to contribute to the existing body of knowledge by evaluating the ability of a Caenorhabditis elegans-based chemotaxis assay to discriminate between urine samples from healthy individuals and patients diagnosed with breast or colon cancer. Following synchronization of the developmental stages of C. elegans, nematodes were exposed to the urine of cancer patients and healthy individuals. Subsequently, chemotactic indices were calculated for each urine sample. Our results demonstrated a statistically significant difference in the chemotactic response of C. elegans to urine samples from cancer patients compared to healthy volunteers (p < 0.001). Furthermore, the test demonstrated promising diagnostic utility, with a sensitivity of 96%, a specificity of 62%, and a detection rate of 73% among patients with breast cancer and a sensitivity of 100%, a specificity of 62%, and a detection rate of 72% among those with colon cancer. Our findings expand on previous observations, confirming the remarkable sensitivity of C. elegans hermaphrodites to discriminating cancer-related volatile organic compounds in urine samples.
Collapse
Affiliation(s)
- Alžbeta Kaiglová
- Department of Laboratory Medicine, Faculty of Health Care and Social Work, Trnava University in Trnava, Univerzitné Námestie 1, 91843, Trnava, Slovakia
| | - Patrícia Hockicková
- Department of Laboratory Medicine, Faculty of Health Care and Social Work, Trnava University in Trnava, Univerzitné Námestie 1, 91843, Trnava, Slovakia
| | - Zuzana Bárdyová
- Department of Laboratory Medicine, Faculty of Health Care and Social Work, Trnava University in Trnava, Univerzitné Námestie 1, 91843, Trnava, Slovakia
| | - Radka Reháková
- Department of Laboratory Medicine, Faculty of Health Care and Social Work, Trnava University in Trnava, Univerzitné Námestie 1, 91843, Trnava, Slovakia
| | - Kamila Melnikov
- Department of Laboratory Medicine, Faculty of Health Care and Social Work, Trnava University in Trnava, Univerzitné Námestie 1, 91843, Trnava, Slovakia
| | - Soňa Kucharíková
- Department of Laboratory Medicine, Faculty of Health Care and Social Work, Trnava University in Trnava, Univerzitné Námestie 1, 91843, Trnava, Slovakia.
| |
Collapse
|
2
|
Schauer C, Teng A, Signal V, Stanley J, Mules TC, Koea J, Inns SJ. Translating evidence into action: overcoming barriers to gastric cancer prevention in Aotearoa. J R Soc N Z 2024:1-19. [DOI: 10.1080/03036758.2024.2427818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 11/06/2024] [Indexed: 01/06/2025]
Affiliation(s)
- Cameron Schauer
- Department of Gastroenterology, Health New Zealand Te Whatu Ora, Waitematā, University of Auckland, Auckland, New Zealand
| | - Andrea Teng
- Department of Public Health, University of Otago, Wellington, New Zealand
| | - Virgina Signal
- Department of Public Health, University of Otago, Wellington, New Zealand
| | - James Stanley
- Department of Public Health, University of Otago, Wellington, New Zealand
| | - Thomas C Mules
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Jonathan Koea
- Department of Surgery, University of Auckland, Auckland, New Zealand
| | - Stephen J. Inns
- Wellington, Health New Zealand Te Whatu Ora, Capital Coast and Hutt Valley, University of Otago, Dunedin, New Zealand
| |
Collapse
|
3
|
Venturini J, Massaro G, Lavacchi D, Rossini D, Pillozzi S, Caliman E, Pellegrini E, Antonuzzo L. The emerging HER2 landscape in colorectal cancer: the key to unveil the future treatment algorithm? Crit Rev Oncol Hematol 2024; 204:104515. [PMID: 39304034 DOI: 10.1016/j.critrevonc.2024.104515] [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: 02/02/2024] [Revised: 09/03/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024] Open
Abstract
Colorectal cancer (CRC) represents a global health threat, standing as the second leading cause of cancer-related death worldwide. Targeted therapies brought new hope for the metastatic stage, which historically bore a very poor prognosis. Human epidermal growth receptor 2 (HER2) overexpression concerns about 5 % of the metastatic CRC (mCRC) patients, including both gene amplifications and point mutations. Albeit its controversial prognostic role, preclinical and clinical data indicate HER2 as a negative predictive biomarker of response to anti-EGFR therapies. Tissue and plasma-based NGS testing, could permit a precise identification of this resistance mechanism both at baseline and during treatment, thus guiding decision-making. Furthermore, promising results come from completed and ongoing randomized trials, testing HER2 as an actionable target. In this review, we discuss the available evidence on HER2 targeting in advanced CRC, analyzing its possible future role in the treatment algorithm.
Collapse
Affiliation(s)
- Jacopo Venturini
- Clinical Oncology Unit, Careggi University Hospital, Florence 50134, Italy
| | - Giulia Massaro
- Clinical Oncology Unit, Careggi University Hospital, Florence 50134, Italy
| | - Daniele Lavacchi
- Clinical Oncology Unit, Careggi University Hospital, Florence 50134, Italy
| | - Daniele Rossini
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy; Department of Health Science, University of Florence, Florence 50139, Italy
| | - Serena Pillozzi
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy; Medical Oncology Unit, Careggi University Hospital, Florence 50134, Italy
| | - Enrico Caliman
- Clinical Oncology Unit, Careggi University Hospital, Florence 50134, Italy
| | - Elisa Pellegrini
- Medical Oncology Unit, Careggi University Hospital, Florence 50134, Italy
| | - Lorenzo Antonuzzo
- Clinical Oncology Unit, Careggi University Hospital, Florence 50134, Italy; Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy; Medical Oncology Unit, Careggi University Hospital, Florence 50134, Italy.
| |
Collapse
|
4
|
Bai L, Guan Y, Zhang Y, Gu J, Ni B, Zhang HY, Aimaiti M, Wang S, Yue B, Xia X, Zhang Z. Effectiveness of peritoneal lavage fluid circulating tumour cells and circulating tumour DNA in the prediction of metachronous peritoneal metastasis of gastric cancer (pT4NxM0/pT1-3N+M0) after radical resection: protocol of a prospective single-centre clinical study. BMJ Open 2024; 14:e083659. [PMID: 39353699 PMCID: PMC11448147 DOI: 10.1136/bmjopen-2023-083659] [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: 12/24/2023] [Accepted: 09/10/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) is the fourth leading cause of cancer mortality worldwide. Peritoneal metastasis (PM) is a significant cause of death in patients with GC, and presents a major challenge in clinical diagnosis and treatment. Predicting the occurrence of PM in high-risk patients, and diagnosing and treating PM in advance to improve patient survival, remains an unsolved problem in clinical practice. Given the low positive rate of cytology and difficulty in diagnosing occult PM, new molecular markers and detection technologies for early diagnosis require urgent validation. The primary objective of this study is to observe and evaluate the predictive effect of intraoperative peritoneal lavage fluid (PLF) circulating tumour cells (CTC) and circulating tumour DNA (ctDNA) levels in patients with pT4NxM0/pT1-3N+M0 GC on metachronous PM after R0 resection. METHODS AND ANALYSIS This prospective single-centre clinical study is conducted at Renji Hospital, Shanghai Jiao Tong University School of Medicine. In this study, 200 cases of patients with pT4NxM0/pT1-3N+M0 gastric adenocarcinoma older than 18 years will be screened. Participants will undergo intraoperative PLF CTC and ctDNA testing and will be followed up for 2 years, with imaging assessments performed every 3-6 months until PM occurrs. The primary outcome is the incidence of PM 1 year after surgery, which will be estimated using Clopper-Pearson method, with 95% CIs calculated and compared between groups. Secondary outcome include the incidence of PM 2 years after surgery, overall survival and disease progression. Data will be analysed using the Kaplan-Meier method and the log-rank test. ETHICS AND COMMUNICATION Informed consent has been obtained from all subjects. This protocol has been approved by the Ethics Committee of Renji Hospital, Shanghai Jiao Tong University School of Medicine (LY2023-142-B). The findings will be disseminated through peer-reviewed manuscripts, reports and presentations. TRIAL REGISTRATION NUMBER ChiCTR2300074910.
Collapse
Affiliation(s)
- Long Bai
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yujing Guan
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yeqian Zhang
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Jiayi Gu
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Bo Ni
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Hao-yu Zhang
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Muerzhate Aimaiti
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Shuchang Wang
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Ben Yue
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Xiang Xia
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Zizhen Zhang
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
5
|
Ratti M, Orlandi E, Toscani I, Vecchia S, Anselmi E, Hahne JC, Ghidini M, Citterio C. Emerging Therapeutic Targets and Future Directions in Advanced Gastric Cancer: A Comprehensive Review. Cancers (Basel) 2024; 16:2692. [PMID: 39123420 PMCID: PMC11311890 DOI: 10.3390/cancers16152692] [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: 06/28/2024] [Revised: 07/19/2024] [Accepted: 07/21/2024] [Indexed: 08/12/2024] Open
Abstract
Metastatic gastric cancer (GC) still represents a critical clinical challenge, with limited treatment options and a poor prognosis. Most patients are diagnosed at advanced stages, limiting the chances of surgery and cure. The identification of molecular targets and the possibility of combining immune checkpoint inhibitors with chemotherapy have recently reshaped the therapeutic landscape of metastatic gastric cancer. The new classification of gastric cancer, mainly based on immunologic and molecular criteria such as programmed cell death 1 (PD-1), microsatellite instability (MSI), and human epidermal growth factor receptor 2 (HER2), has made it possible to identify and differentiate patients who may benefit from immunotherapy, targeted therapy, or chemotherapy alone. All relevant and available molecular and immunological targets in clinical practice for the systemic treatment of this disease are presented. Particular attention is given to possible future approaches, including circulating tumor DNA (ctDNA) for therapeutic monitoring, new targeting agents against molecular pathways such as fibroblast growth factor receptor (FGFR) and MET, chimeric antigen receptor (CAR)-T cells, and cancer vaccines. This review aims to provide a comprehensive understanding of current targets in advanced gastric cancer and to offer valuable insights into future directions of research and clinical practice in this challenging disease.
Collapse
Affiliation(s)
- Margherita Ratti
- Oncology and Hematology Department, Piacenza General Hospital, Via Taverna 49, 29121 Piacenza, Italy; (E.O.); (I.T.); (S.V.); (E.A.); (C.C.)
| | - Elena Orlandi
- Oncology and Hematology Department, Piacenza General Hospital, Via Taverna 49, 29121 Piacenza, Italy; (E.O.); (I.T.); (S.V.); (E.A.); (C.C.)
| | - Ilaria Toscani
- Oncology and Hematology Department, Piacenza General Hospital, Via Taverna 49, 29121 Piacenza, Italy; (E.O.); (I.T.); (S.V.); (E.A.); (C.C.)
| | - Stefano Vecchia
- Oncology and Hematology Department, Piacenza General Hospital, Via Taverna 49, 29121 Piacenza, Italy; (E.O.); (I.T.); (S.V.); (E.A.); (C.C.)
| | - Elisa Anselmi
- Oncology and Hematology Department, Piacenza General Hospital, Via Taverna 49, 29121 Piacenza, Italy; (E.O.); (I.T.); (S.V.); (E.A.); (C.C.)
| | - Jens Claus Hahne
- Division of Molecular Pathology, The Institute of Cancer Research, Sutton, London SM2 5NG, UK;
| | - Michele Ghidini
- Medical Oncology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Chiara Citterio
- Oncology and Hematology Department, Piacenza General Hospital, Via Taverna 49, 29121 Piacenza, Italy; (E.O.); (I.T.); (S.V.); (E.A.); (C.C.)
| |
Collapse
|
6
|
Shaker F, Razi S, Rezaei N. Circulating miRNA and circulating tumor DNA application as liquid biopsy markers in gastric cancer. Clin Biochem 2024; 129:110767. [PMID: 38705444 DOI: 10.1016/j.clinbiochem.2024.110767] [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: 02/05/2024] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
Liquid biopsy has been investigated as a novel method to overcome the numerous challenges in gastric cancer (GC) management. This non-invasive, feasible, and easy-to-repeat method has been shown to be cost-effective and capable of increasing diagnostic sensitivity and prognostic assessment. Additionally, it is potentially accurate to aid decision-making and personalized treatment planning. MicroRNA (miRNA) and circulating tumor DNA (ctDNA) markers can enhance GC management in various aspects, including diagnosis (mainly earlier diagnosis and the ability to perform population-based screening), prognosis (more precise stratification of prognosis), and treatment (including more accurate prediction of treatment response and earlier detection of resistance to the treatment). Concerning the treatment-related application, miRNAs' mimics and antagonists (by using two main strategies of restoring tumor suppressor miRNAs and inhibiting oncogene miRNAs) have been shown to be effective therapeutic agents. However, these need to be further validated in clinical trials. Furthermore, novel delivery systems, such as lipid-based vectors, polymeric-based vectors, and exosome-based delivery, have been developed to enhance the performance of these agents. Moreover, this paper explores the current detection and measuring methods for these markers. These approaches are categorized into direct methods (e.g., Chem-NAT, HTG EdgeSeq, and Multiplex Circulating Fireplex) and indirect methods (e.g., Reverse transcription-quantitative polymerase chain reaction (RT-qPCR), qPCR, microarray, and NGS) for miRNA detection. For ctDNA measurement, main core technologies like NGS, digital PCR, real-time PCR, and mass spectrometry are suggested.
Collapse
Affiliation(s)
- Farhad Shaker
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Sepideh Razi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Stockholm, Sweden.
| |
Collapse
|
7
|
Díaz del Arco C, Fernández Aceñero MJ, Ortega Medina L. Liquid biopsy for gastric cancer: Techniques, applications, and future directions. World J Gastroenterol 2024; 30:1680-1705. [PMID: 38617733 PMCID: PMC11008373 DOI: 10.3748/wjg.v30.i12.1680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 02/01/2024] [Accepted: 03/08/2024] [Indexed: 03/28/2024] Open
Abstract
After the study of circulating tumor cells in blood through liquid biopsy (LB), this technique has evolved to encompass the analysis of multiple materials originating from the tumor, such as nucleic acids, extracellular vesicles, tumor-educated platelets, and other metabolites. Additionally, research has extended to include the examination of samples other than blood or plasma, such as saliva, gastric juice, urine, or stool. LB techniques are diverse, intricate, and variable. They must be highly sensitive, and pre-analytical, patient, and tumor-related factors significantly influence the detection threshold, diagnostic method selection, and potential results. Consequently, the implementation of LB in clinical practice still faces several challenges. The potential applications of LB range from early cancer detection to guiding targeted therapy or immunotherapy in both early and advanced cancer cases, monitoring treatment response, early identification of relapses, or assessing patient risk. On the other hand, gastric cancer (GC) is a disease often diagnosed at advanced stages. Despite recent advances in molecular understanding, the currently available treatment options have not substantially improved the prognosis for many of these patients. The application of LB in GC could be highly valuable as a non-invasive method for early diagnosis and for enhancing the management and outcomes of these patients. In this comprehensive review, from a pathologist's perspective, we provide an overview of the main options available in LB, delve into the fundamental principles of the most studied techniques, explore the potential utility of LB application in the context of GC, and address the obstacles that need to be overcome in the future to make this innovative technique a game-changer in cancer diagnosis and treatment within clinical practice.
Collapse
Affiliation(s)
- Cristina Díaz del Arco
- Department of Surgical Pathology, Health Research Institute of the Hospital Clínico San Carlos, Hospital Clínico San Carlos, Madrid 28040, Spain
- Department of Legal Medicine, Psychiatry and Pathology, Universidad Complutense de Madrid, Madrid 28040, Spain
| | - M Jesús Fernández Aceñero
- Department of Surgical Pathology, Health Research Institute of the Hospital Clínico San Carlos, Hospital Clínico San Carlos, Madrid 28040, Spain
- Department of Legal Medicine, Psychiatry and Pathology, Universidad Complutense de Madrid, Madrid 28040, Spain
| | - Luis Ortega Medina
- Department of Surgical Pathology, Health Research Institute of the Hospital Clínico San Carlos, Hospital Clínico San Carlos, Madrid 28040, Spain
- Department of Legal Medicine, Psychiatry and Pathology, Universidad Complutense de Madrid, Madrid 28040, Spain
| |
Collapse
|
8
|
Zazo S, Pérez‐Buira S, Carvajal N, Plaza‐Sánchez J, Manso R, Pérez‐González N, Dominguez C, Prieto‐Potin I, Rubio J, Dómine M, Lozano V, Mohedano P, Carcedo D, Carias R, Rojo F. Actionable mutational profiling in solid tumors using hybrid-capture-based next-generation sequencing in a real-world setting in Spain. Cancer Med 2024; 13:e6827. [PMID: 38213074 PMCID: PMC10905216 DOI: 10.1002/cam4.6827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 11/09/2023] [Accepted: 12/08/2023] [Indexed: 01/13/2024] Open
Abstract
OBJECTIVE This study aimed to describe the performance of a next-generation sequencing (NGS) panel for the detection of precise genomic alterations in cancer in Spanish clinical practice. The impact of tumor characteristics was evaluated on informative NGS and actionable mutation rates. MATERIALS AND METHODS A cross-sectional study was conducted at the Fundación Jiménez Díaz University Hospital (May 2021-March 2022) where molecular diagnostic of 537 Formalin-Fixed Paraffin-Embedded (FFPE) tissue samples of diverse solid tumors (lung, colorectal, melanoma, gastrointestinal stromal, among others) was performed using AVENIO Tumor Tissue Targeted Kit. A descriptive analysis of the features of all samples was carried out. Multivariable logistic analysis was conducted to assess the impact of sample characteristics on NGS performance defined by informative results rate (for all tumors and for lung tumors), and on actionable mutations rate (for lung tumors only). RESULTS AVENIO performance rate was 75.2% in all tumor samples and 75.3% in lung cancer samples, and the multivariable analysis showed that surgical specimens are most likely to provide informative results than diagnostic biopsies. Regarding the mutational findings, 727 pathogenic, likely pathogenic, or variant of unknown significance mutations were found in all tumor samples. Single nucleotide variant was the most common genomic alteration, both for all tumor samples (85.3% and 81.9% for all solid tumors and lung samples, respectively). In lung tumors, multivariable analysis showed that it is more likely to find actionable mutations from non-smokers and patients with adenocarcinoma, large cell, or undifferentiated histologies. CONCLUSION This is the largest cohort-level study in Spain to profile the analyses of biopsy samples of different tumors using NGS in routine clinical practice. Our findings showed that the use of NGS routinely provides good rates of informative results and can improve tumor characterization and identify a greater number of actionable mutations.
Collapse
Affiliation(s)
- Sandra Zazo
- Department of PathologyFundación Jiménez Díaz University HospitalMadridSpain
- IIS‐Fundación Jimenez DiazCenter for Biomedical Network Research on Cancer (CIBERONC)MadridSpain
| | - Sandra Pérez‐Buira
- Department of PathologyFundación Jiménez Díaz University HospitalMadridSpain
| | - Nerea Carvajal
- Department of PathologyFundación Jiménez Díaz University HospitalMadridSpain
| | | | - Rebeca Manso
- Department of PathologyFundación Jiménez Díaz University HospitalMadridSpain
| | | | - Carolina Dominguez
- IIS‐Fundación Jimenez DiazCenter for Biomedical Network Research on Cancer (CIBERONC)MadridSpain
| | - Iván Prieto‐Potin
- Department of PathologyFundación Jiménez Díaz University HospitalMadridSpain
| | - Jaime Rubio
- Medical Oncology DepartmentFundación Jiménez Díaz University HospitalMadridSpain
| | - Manuel Dómine
- IIS‐Fundación Jimenez DiazCenter for Biomedical Network Research on Cancer (CIBERONC)MadridSpain
- Medical Oncology DepartmentFundación Jiménez Díaz University HospitalMadridSpain
| | | | | | | | - Rafael Carias
- Department of PathologyFundación Jiménez Díaz University HospitalMadridSpain
| | - Federico Rojo
- Department of PathologyFundación Jiménez Díaz University HospitalMadridSpain
- IIS‐Fundación Jimenez DiazCenter for Biomedical Network Research on Cancer (CIBERONC)MadridSpain
| |
Collapse
|
9
|
Wang X, Wang L, Lin H, Zhu Y, Huang D, Lai M, Xi X, Huang J, Zhang W, Zhong T. Research progress of CTC, ctDNA, and EVs in cancer liquid biopsy. Front Oncol 2024; 14:1303335. [PMID: 38333685 PMCID: PMC10850354 DOI: 10.3389/fonc.2024.1303335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 01/04/2024] [Indexed: 02/10/2024] Open
Abstract
Circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), and extracellular vehicles (EVs) have received significant attention in recent times as emerging biomarkers and subjects of transformational studies. The three main branches of liquid biopsy have evolved from the three primary tumor liquid biopsy detection targets-CTC, ctDNA, and EVs-each with distinct benefits. CTCs are derived from circulating cancer cells from the original tumor or metastases and may display global features of the tumor. ctDNA has been extensively analyzed and has been used to aid in the diagnosis, treatment, and prognosis of neoplastic diseases. EVs contain tumor-derived material such as DNA, RNA, proteins, lipids, sugar structures, and metabolites. The three provide different detection contents but have strong complementarity to a certain extent. Even though they have already been employed in several clinical trials, the clinical utility of three biomarkers is still being studied, with promising initial findings. This review thoroughly overviews established and emerging technologies for the isolation, characterization, and content detection of CTC, ctDNA, and EVs. Also discussed were the most recent developments in the study of potential liquid biopsy biomarkers for cancer diagnosis, therapeutic monitoring, and prognosis prediction. These included CTC, ctDNA, and EVs. Finally, the potential and challenges of employing liquid biopsy based on CTC, ctDNA, and EVs for precision medicine were evaluated.
Collapse
Affiliation(s)
- Xiaoling Wang
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| | - Lijuan Wang
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| | - Haihong Lin
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| | - Yifan Zhu
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| | - Defa Huang
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Mi Lai
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Xuxiang Xi
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Junyun Huang
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| | - Wenjuan Zhang
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| | - Tianyu Zhong
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| |
Collapse
|
10
|
Han HS, Lee KW. Liquid Biopsy: An Emerging Diagnostic, Prognostic, and Predictive Tool in Gastric Cancer. J Gastric Cancer 2024; 24:4-28. [PMID: 38225764 PMCID: PMC10774753 DOI: 10.5230/jgc.2024.24.e5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 01/17/2024] Open
Abstract
Liquid biopsy, a minimally invasive procedure that causes minimal pain and complication risks to patients, has been extensively studied for cancer diagnosis and treatment. Moreover, it facilitates comprehensive quantification and serial assessment of the whole-body tumor burden. Several biosources obtained through liquid biopsy have been studied as important biomarkers for establishing early diagnosis, monitoring minimal residual disease, and predicting the prognosis and response to treatment in patients with cancer. Although the clinical application of liquid biopsy in gastric cancer is not as robust as that in other cancers, biomarker studies using liquid biopsy are being actively conducted in patients with gastric cancer. Herein, we aimed to review the role of various biosources that can be obtained from patients with gastric cancer through liquid biopsies, such as blood, saliva, gastric juice, urine, stool, peritoneal lavage fluid, and ascites, by dividing them into cellular and acellular components. In addition, we reviewed previous studies on the diagnostic, prognostic, and predictive biomarkers for gastric cancer using liquid biopsy and discussed the limitations of liquid biopsy and the challenges to overcome these limitations in patients with gastric cancer.
Collapse
Affiliation(s)
- Hye Sook Han
- Department of Internal Medicine, Chungbuk National University Hospital, Chungbuk National University College of Medicine, Cheongju, Korea
| | - Keun-Wook Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea.
| |
Collapse
|
11
|
Seo SH, Park YS, Nam SK, Lee HS, Park DJ, Park KU. Concordance of circulating tumor DNA and matched formalin-fixed paraffin-embedded tumor tissue in gastric cancer as a predictor of recurrence. KOREAN JOURNAL OF CLINICAL ONCOLOGY 2023; 19:45-51. [PMID: 38229488 DOI: 10.14216/kjco.23009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 10/24/2023] [Indexed: 01/18/2024]
Abstract
PURPOSE Combined analysis of the variant composition of circulating tumor DNA (ctDNA) from cell-free plasma and DNA from tumor tissue could provide insight into the implications of the genetic alterations responsible for the intratumoral and intertumoral heterogeneity of gastric cancer. We aimed to evaluate the usefulness of this approach in these patients. METHODS Cell-free plasma and formalin-fixed paraffin-embedded tumor tissue samples from 46 patients with gastric cancer were examined. Targeted deep sequencing was performed using a commercially available kit. RESULTS The cell-free DNA (cfDNA) concentration was higher in stage II-IV versus stage I patients and in larger versus smaller tumors. Only 12 of the 36 (33.3%) alterations in the tumor tissue samples were in concordance with those in the ctDNA samples. Two variants were in concordance in stage I samples and 10 in stage II-IV samples. Actionable variants that were detected in concordance were in the stage II-IV samples. Preoperative ctDNA positivity of actionable variants was significantly associated with cfDNA concentration, lymphatic invasion, N stage, and TNM stage. Cancer recurrence was significantly associated with tumor size, lymphatic/vascular invasion, TNM stage, and ctDNA-tumor tissue variant concordance. CONCLUSION Preoperative ctDNA genetic analysis using a multigene panel offers substantial clinical benefits when performed in conjunction with targeted deep sequencing of tumor tissue. Concordance between preoperative ctDNA and tumor tissue mutations may serve as a prognostic indicator in patients with gastric cancer.
Collapse
Affiliation(s)
- Soo Hyun Seo
- Department of Laboratory Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Young Suk Park
- Department of Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Soo Kyung Nam
- Department of Interdisciplinary Program in Cancer Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Hye Seung Lee
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Do Joong Park
- Department of Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Kyoung Un Park
- Department of Laboratory Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| |
Collapse
|
12
|
Yun JH. Integrating ctDNA and tumor tissue analysis in gastric cancer: a synergistic approach unveils prognostic insights. KOREAN JOURNAL OF CLINICAL ONCOLOGY 2023; 19:43-44. [PMID: 38229487 DOI: 10.14216/kjco.23008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 12/26/2023] [Indexed: 01/18/2024]
Affiliation(s)
- Jong Hyuk Yun
- Department of Surgery, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| |
Collapse
|
13
|
Zhao J, Reuther J, Scozzaro K, Hawley M, Metzger E, Emery M, Chen I, Barbosa M, Johnson L, O'Connor A, Washburn M, Hartje L, Reckase E, Johnson V, Zhang Y, Westheimer E, O'Callaghan W, Malani N, Chesh A, Moreau M, Daber R. Personalized Cancer Monitoring Assay for the Detection of ctDNA in Patients with Solid Tumors. Mol Diagn Ther 2023; 27:753-768. [PMID: 37632661 PMCID: PMC10590345 DOI: 10.1007/s40291-023-00670-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/27/2023] [Indexed: 08/28/2023]
Abstract
BACKGROUND Highly sensitive molecular assays have been developed to detect plasma-based circulating tumor DNA (ctDNA), and emerging evidence suggests their clinical utility for monitoring minimal residual disease and recurrent disease, providing prognostic information, and monitoring therapy responses in patients with solid tumors. The Invitae Personalized Cancer Monitoring™ assay uses a patient-specific, tumor-informed variant signature identified through whole exome sequencing to detect ctDNA in peripheral blood of patients with solid tumors. METHODS The assay's tumor whole exome sequencing and ctDNA detection components were analytically validated using 250 unique human specimens and nine commercial reference samples that generated 1349 whole exome sequencing and cell-free DNA (cfDNA)-derived libraries. A comparison of tumor and germline whole exome sequencing was used to identify patient-specific tumor variant signatures and generate patient-specific panels, followed by targeted next-generation sequencing of plasma-derived cfDNA using the patient-specific panels with anchored multiplex polymerase chain reaction chemistry leveraging unique molecular identifiers. RESULTS Whole exome sequencing resulted in overall sensitivity of 99.8% and specificity of > 99.9%. Patient-specific panels were successfully designed for all 63 samples (100%) with ≥ 20% tumor content and 24 (80%) of 30 samples with ≥ 10% tumor content. Limit of blank studies using 30 histologically normal, formalin-fixed paraffin-embedded specimens resulted in 100% expected panel design failure. The ctDNA detection component demonstrated specificity of > 99.9% and sensitivity of 96.3% for a combination of 10 ng of cfDNA input, 0.008% allele frequency, 50 variants on the patient-specific panels, and a baseline threshold. Limit of detection ranged from 0.008% allele frequency when utilizing 60 ng of cfDNA input with 18-50 variants in the patient-specific panels (> 99.9% sensitivity) with a baseline threshold, to 0.05% allele frequency when using 10 ng of cfDNA input with an 18-variant panel with a monitoring threshold (> 99.9% sensitivity). CONCLUSIONS The Invitae Personalized Cancer Monitoring assay, featuring a flexible patient-specific panel design with 18-50 variants, demonstrated high sensitivity and specificity for detecting ctDNA at variant allele frequencies as low as 0.008%. This assay may support patient prognostic stratification, provide real-time data on therapy responses, and enable early detection of residual/recurrent disease.
Collapse
Affiliation(s)
- Jianhua Zhao
- Invitae Corp., 1400 16th Street, San Francisco, CA, 94103, USA.
| | | | - Kaylee Scozzaro
- Invitae Corp., 1400 16th Street, San Francisco, CA, 94103, USA
| | - Megan Hawley
- Invitae Corp., 1400 16th Street, San Francisco, CA, 94103, USA
| | - Emily Metzger
- Invitae Corp., 1400 16th Street, San Francisco, CA, 94103, USA
| | - Matthew Emery
- Invitae Corp., 1400 16th Street, San Francisco, CA, 94103, USA
| | - Ingrid Chen
- Invitae Corp., 1400 16th Street, San Francisco, CA, 94103, USA
| | | | - Laura Johnson
- Invitae Corp., 1400 16th Street, San Francisco, CA, 94103, USA
- Affiliated with Invitae Corp. at the time of the study, currently employees at Integrated DNA Technologies, 1710 Commercial Park, Coralville, IA, 52241, USA
| | - Alijah O'Connor
- Invitae Corp., 1400 16th Street, San Francisco, CA, 94103, USA
| | - Mike Washburn
- Invitae Corp., 1400 16th Street, San Francisco, CA, 94103, USA
- Affiliated with Invitae Corp. at the time of the study, currently employees at Integrated DNA Technologies, 1710 Commercial Park, Coralville, IA, 52241, USA
| | - Luke Hartje
- Invitae Corp., 1400 16th Street, San Francisco, CA, 94103, USA
- Affiliated with Invitae Corp. at the time of the study, currently employees at Integrated DNA Technologies, 1710 Commercial Park, Coralville, IA, 52241, USA
| | - Erik Reckase
- Invitae Corp., 1400 16th Street, San Francisco, CA, 94103, USA
- Affiliated with Invitae Corp. at the time of the study, currently employees at Integrated DNA Technologies, 1710 Commercial Park, Coralville, IA, 52241, USA
| | - Verity Johnson
- Invitae Corp., 1400 16th Street, San Francisco, CA, 94103, USA
- Affiliated with Invitae Corp. at the time of the study, currently employees at Integrated DNA Technologies, 1710 Commercial Park, Coralville, IA, 52241, USA
| | - Yuhua Zhang
- Invitae Corp., 1400 16th Street, San Francisco, CA, 94103, USA
| | | | | | - Nirav Malani
- Invitae Corp., 1400 16th Street, San Francisco, CA, 94103, USA
| | - Adrian Chesh
- Invitae Corp., 1400 16th Street, San Francisco, CA, 94103, USA
| | - Michael Moreau
- Invitae Corp., 1400 16th Street, San Francisco, CA, 94103, USA
| | - Robert Daber
- Invitae Corp., 1400 16th Street, San Francisco, CA, 94103, USA
| |
Collapse
|
14
|
Takayama T, Tsuji Y. Updated Adjuvant Chemotherapy for Gastric Cancer. J Clin Med 2023; 12:6727. [PMID: 37959193 PMCID: PMC10648766 DOI: 10.3390/jcm12216727] [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: 08/25/2023] [Revised: 10/03/2023] [Accepted: 10/18/2023] [Indexed: 11/15/2023] Open
Abstract
Surgical resection is currently the best curative approach for gastric cancer (GC); however, the prognosis of patients with advanced GC remains poor even with curative resection. For this reason, perioperative chemotherapy has been combined with surgery to reduce the risk of postoperative recurrence. Standard perioperative chemotherapy for resectable advanced GC varies from region to region. Postoperative S-1 therapy was standardized via the ACTS-GC study in East Asia, perioperative ECF (Epirubicin + Cisplatin + Fluorouracil) was standardized via the MAGIC study in Europe, and postoperative chemoradiotherapy was standardized via the US intergroup study in North America. Since then, more intensive regimens have been developed. In recent years, perioperative therapy using novel agents, such as molecular-targeted drugs and immune checkpoint inhibitors (ICIs), has also been tested and evaluated in the three major regions (East Asia, Europe, and North America) with promising results. Perioperative chemotherapy has become an integral part of many treatment strategies and requires continued research and evaluation.
Collapse
Affiliation(s)
- Toshizo Takayama
- Department of Medical Oncology, Tonan Hospital, Sapporo 060-0004, Japan
- Department of Medical Oncology, Daido Hospital, Nagoya 457-8511, Japan
| | - Yasushi Tsuji
- Department of Medical Oncology, Tonan Hospital, Sapporo 060-0004, Japan
| |
Collapse
|
15
|
Assumpção PPD, Silva JMCD, Calcagno DQ, Barra WF, Ishak G, Kassab P. OLIGOMETASTASIS IN GASTRIC CANCER TREATMENT: IS THERE A PLACE FOR THE SURGEON? ARQUIVOS BRASILEIROS DE CIRURGIA DIGESTIVA : ABCD = BRAZILIAN ARCHIVES OF DIGESTIVE SURGERY 2023; 36:e1752. [PMID: 37729281 PMCID: PMC10510098 DOI: 10.1590/0102-672020230034e1752] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 06/01/2023] [Indexed: 09/22/2023]
Abstract
Metastatic gastric cancer traditionally hinders surgical treatment options, confining them to palliative procedures. The presence of metastases in these tumors is classified as M1, irrespective of their characteristics, quantity, or location. However, oligometastatic disease emerged as an intermediate state between localized and widely disseminated cancer. It exhibits diverse patterns based on metastatic disease extent, type, and location. Adequately addressing this distinctive metastatic state necessitates tailored strategies that surpass the realm of palliative care. Differentprimary tumor types present discernible scenarios of oligometastatic disease, including preferred sites of occurrence and chronological progression. Due to the novelty of this theme and the heterogeneity of the disease, uncertainties still exist, and the ability to provide confident guidelines is challenging. Currently, there are no effective predictors to determine the response and provide clear indications for surgical interventions and systemic treatments in oligometastatic disease. Treatment decisions are commonly based on apparent disease control by systemic therapies, with a short observation period and imaging assessments. Nonetheless, the inherent risk of misinterpretation remains a constant concern. The emergence of novel technologies and therapeutic modalities, such as immunotherapy, cellular therapy, and adoptive therapies, holds the potential to reshape the landscape of surgical treatment for the oligometastatic disease in gastric cancer, expanding the surgeon's role in this multidisciplinary approach. Prospective tools for patient selection in oligometastatic gastric cancer are being explored. Using non-invasive, cost-effective, widely available imaging techniques that provide real-time information may revolutionize medical practice, ensuring precision medicine accessibility, even in resource-constrained small healthcare facilities. Incorporating molecular classifications, liquid biopsies, and radiomic analysis in a complementary protocol will augment patient selection precision for surgical intervention in oligometastasis. Hopefully, these advancements will render surgeries unnecessary in many cases by providing highly effective alternative treatments.
Collapse
Affiliation(s)
- Paulo Pimentel de Assumpção
- Universidade Federal do Pará, Oncology Research Center - Belém (PA), Brazil
- Universidade Federal do Pará, João de Barros Barreto University Hospital, General Surgery and Digestive Tract Service - Belém (PA), Brazil
| | | | | | | | - Geraldo Ishak
- Universidade Federal do Pará, Oncology Research Center - Belém (PA), Brazil
- Universidade Federal do Pará, João de Barros Barreto University Hospital, General Surgery and Digestive Tract Service - Belém (PA), Brazil
| | - Paulo Kassab
- Faculdade de Ciências Médicas da Santa Casa de São Paulo, Department of Surgery - São Paulo (SP), Brazil
| |
Collapse
|
16
|
Januszewicz W, Turkot MH, Malfertheiner P, Regula J. A Global Perspective on Gastric Cancer Screening: Which Concepts Are Feasible, and When? Cancers (Basel) 2023; 15:664. [PMID: 36765621 PMCID: PMC9913879 DOI: 10.3390/cancers15030664] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/12/2023] [Accepted: 01/16/2023] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Gastric cancer (GC) remains the fifth most common cancer and the third most common cause of cancer-related death globally. In 2022, GC fell into the scope of the updated EU recommendations for targeted cancer screening. Given the growing awareness of the GC burden, we aimed to review the existing screening strategies for GC in high-risk regions and discuss potentially applicable modalities in countries with low-to-intermediate incidence. METHODS The references for this Review article were identified through searches of PubMed with the search terms "gastric cancer", "stomach cancer", "Helicobacter pylori", and "screening" over the period from 1995 until August 2022. RESULTS As Helicobacter pylori (H. pylori)-induced gastritis is the primary step in the development of GC, the focus on GC prevention may be directed toward testing for and treating this infection. Such a strategy may be appealing in countries with low- and intermediate- GC incidence. Other biomarker-based approaches to identify at-risk individuals in such regions are being evaluated. Within high-incidence areas, both primary endoscopic screening and population-based H. pylori "test-and-treat" strategies represent cost-effective models. CONCLUSIONS Given the significant variations in GC incidence and healthcare resources around the globe, screening strategies for GC should be adjusted to the actual conditions in each region. While several proven tools exist for accurate GC diagnosis, a universal modality for the screening of GC populations remains elusive.
Collapse
Affiliation(s)
- Wladyslaw Januszewicz
- Department of Oncological Gastroenterology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, 02-781 Warsaw, Poland
| | - Maryla Helena Turkot
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, 02-781 Warsaw, Poland
| | - Peter Malfertheiner
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University, 39120 Magdeburg, Germany
| | - Jaroslaw Regula
- Department of Oncological Gastroenterology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, 02-781 Warsaw, Poland
| |
Collapse
|