1
|
Yang W, Nguyen R, Safri F, Shiddiky MJA, Warkiani ME, George J, Qiao L. Liquid Biopsy in Hepatocellular Carcinoma: ctDNA as a Potential Biomarker for Diagnosis and Prognosis. Curr Oncol Rep 2025:10.1007/s11912-025-01681-3. [PMID: 40343687 DOI: 10.1007/s11912-025-01681-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2025] [Indexed: 05/11/2025]
Abstract
PURPOSE OF REVIEW Hepatocellular carcinoma (HCC) is a leading cause of cancer-related mortality worldwide, with rising incidence and mortality. Early-stage HCC is often asymptomatic, and the lack of reliable early diagnostic markers leads to late-stage diagnosis with limited treatment options. Current treatment relies on tumour staging and patient status, but accurate staging requires invasive procedures that fail to capture tumour heterogeneity and progression. There is an urgent need for less invasive diagnostic strategies, such as liquid biopsy technologies, which allow for repeated sampling and real-time analysis of tumour dynamics. Liquid biopsies, including circulating tumour cells (CTCs) and circulating tumour DNA (ctDNA), offer the potential to monitor recurrence, metastasis, and treatment responses, potentially transforming HCC clinical management by enabling earlier intervention and personalised treatment strategies. RECENT FINDINGS Recent studies emphasise the potential of ctDNA as a non-invasive biomarker by targeting DNA methylation for early HCC detection, enabling timely intervention and personalised treatment to improve patient outcomes. Comparative analyses have shown that ctDNA mutation testing outperforms alpha-fetoprotein (AFP), with a sensitivity of 85% and a specificity of 92%, compared to 60% sensitivity and 80% specificity for AFP. Additionally, profiling the ctDNA mutation landscape of 100 HCC patients has identified recurrent mutations in genes such as TP53, CTNNB1, and AXIN1. ctDNA appears to be a promising non-invasive biomarker in the clinical management of HCC patients, with the sensitivity and specificity improving by 41.67% and 15% respectively. The ctDNA mutations, particularly those targeting DNA methylation, highlight great potential for precision medicine, critical for early diagnosis and prognosis of HCC.
Collapse
Affiliation(s)
- William Yang
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney and Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Romario Nguyen
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney and Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Fatema Safri
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney and Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Muhammad J A Shiddiky
- Rural Health Research Institute (RHRI), Charles Sturt University, Orange, NSW, 2800, Australia
| | - Majid E Warkiani
- School of Biomedical Engineering, The University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Jacob George
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney and Westmead Hospital, Westmead, NSW, 2145, Australia.
- Storr Liver Centre, Westmead Institute for Medical Research (WIMR), the University of Sydney, Westmead, NSW, 2145, Australia.
| | - Liang Qiao
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney and Westmead Hospital, Westmead, NSW, 2145, Australia.
- Storr Liver Centre, Westmead Institute for Medical Research (WIMR), the University of Sydney, Westmead, NSW, 2145, Australia.
| |
Collapse
|
2
|
Grancher A, Beaussire-Trouvay L, Vernon V, Dutherage M, Blondin V, Elie C, Bouhier-Leporrier K, Galais MP, Clabaut T, Bignon AL, Parzy A, Gangloff A, Schwarz L, Lévêque E, Sabourin JC, Michel P, Vasseur N, Sefrioui D, Gilibert A, Di Fiore F. ctDNA variations according to treatment intensity in first-line metastatic colorectal cancer. Br J Cancer 2025; 132:814-821. [PMID: 40089635 PMCID: PMC12041588 DOI: 10.1038/s41416-025-02971-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 01/14/2025] [Accepted: 02/25/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Circulating tumor DNA variations (∆ctDNA) were reported to be associated with treatment efficacy in metastatic colorectal cancer (mCRC). The present study evaluated ∆ctDNA according to first-line treatment intensity. METHODS Patients from two prospective ctDNA collections were divided into Group ≤ 2 drugs and Group ≥ 3 drugs. ∆ctDNA were analysed from baseline to cycle 3 or 4 (C3-4) according to three predefined subgroups: ∆ctDNA ≥ 80%_ undetectable, ∆ctDNA ≥ 80%_ detectable, and ∆ctDNA < 80%. Impact of ∆ctDNA on progression-free survival (PFS) and overall survival (OS) were analysed. RESULTS Pretreatment ctDNA was detected in 129/152 (84.9%) of patients. A ∆ctDNA ≥ 80%_undetectable was more frequent in Group ≥ 3 than ≤ 2 drugs (respectively 51.5% vs. 32.7%, p = 0.015). Patients with ∆ctDNA ≥ 80%_undetectable had longer survival than other ∆ctDNA subgroups, in Group ≥ 3 drugs (mPFS 11.5 vs 7.8 vs 6.3 months, p = 0.02: mOS 30.2 vs 18.1 vs 16.4 month, p = 0.04) and in Group ≤ 2 drugs (mPFS 8.4 vs 6.0 vs 5.3 months, p = 0.05; mOS 29.6 vs 14.6 vs 14.6 months, p = 0.007). DISCUSSION Early ∆ctDNA are associated to treatment intensity in first line mCRC with a significant impact on prognosis.
Collapse
Affiliation(s)
- Adrien Grancher
- Normandie Univ, UNIROUEN, Inserm U1245, IRON group, Normandy Centre for Genomic and Personalized Medicine and Department of Hepato-gastroenterology and Digestive Oncology, Rouen University Hospital, Rouen, France
| | - Ludivine Beaussire-Trouvay
- Normandie Univ, UNIROUEN, Inserm U1245, IRON group, Normandy Centre for Genomic and Personalized Medicine, Rouen University Hospital, Rouen, France
| | - Virginie Vernon
- Normandie Univ, UNIROUEN, Inserm U1245, IRON group, Normandy Centre for Genomic and Personalized Medicine and Department of Hepato-gastroenterology and Digestive Oncology, Rouen University Hospital, Rouen, France
| | - Marie Dutherage
- Normandie Univ, UNIROUEN, Inserm U1245, IRON group, Normandy Centre for Genomic and Personalized Medicine and Department of Hepato-gastroenterology and Digestive Oncology, Rouen University Hospital, Rouen, France
| | - Valérie Blondin
- CHI Elbeuf, Department of Hepatogastroenterology, Elbeuf, France
| | - Caroline Elie
- CHI Elbeuf, Department of Hepatogastroenterology, Elbeuf, France
| | | | - Marie-Pierre Galais
- Department of Hepatogastroenterology, Francois Baclesse Centre, Caen, France
| | - Tifenn Clabaut
- Department of Biostatistics, Rouen University Hospital, Rouen, France
| | - Anne-Laure Bignon
- Department of Hepatogastroenterology, Caen University Hospital, Caen, France
| | - Aurélie Parzy
- Department of Hepatogastroenterology, Francois Baclesse Centre, Caen, France
| | | | - Lilian Schwarz
- Department of Digestive Surgery, Rouen University Hospital, Rouen, France
| | - Emilie Lévêque
- Clinical Research Unit, Centre Henri Becquerel, Rouen, France
| | | | - Pierre Michel
- Normandie Univ, UNIROUEN, Inserm U1245, IRON group, Normandy Centre for Genomic and Personalized Medicine and Department of Hepato-gastroenterology and Digestive Oncology, Rouen University Hospital, Rouen, France
| | - Nasrin Vasseur
- Normandie Univ, UNIROUEN, Inserm U1245, IRON group, Normandy Centre for Genomic and Personalized Medicine, Rouen University Hospital, Rouen, France
| | - David Sefrioui
- Normandie Univ, UNIROUEN, Inserm U1245, IRON group, Normandy Centre for Genomic and Personalized Medicine and Department of Hepato-gastroenterology and Digestive Oncology, Rouen University Hospital, Rouen, France
| | - André Gilibert
- Department of Biostatistics, Rouen University Hospital, Rouen, France
| | - Frédéric Di Fiore
- Normandie Univ, UNIROUEN, Inserm U1245, IRON group, Normandy Centre for Genomic and Personalized Medicine and Department of Hepato-gastroenterology and Digestive Oncology, Rouen University Hospital, Rouen, France.
| |
Collapse
|
3
|
Li S, Geng S, Chen Y, Ren Q, Luan Y, Liang W, Chang Y, Zhang L, Zhu D, Wu W, Zhang Y, Zhang L, Wang Y, Zhong G, Wei B, Ma J, Chang Y, Wang X, Li Z, Duan C, Long G, Mao M. Clinical Validation of a Noninvasive Multi-Omics Method for Multicancer Early Detection in Retrospective and Prospective Cohorts. J Mol Diagn 2025:S1525-1578(25)00106-0. [PMID: 40311780 DOI: 10.1016/j.jmoldx.2025.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 03/04/2025] [Accepted: 04/04/2025] [Indexed: 05/03/2025] Open
Abstract
Recent studies highlight the promise of blood-based multicancer early detection (MCED) tests for identifying asymptomatic patients with cancer. However, most focus on a single cancer hallmark, thus limiting effectiveness because of cancer's heterogeneity. Here, a blood-based multi-omics test named SeekInCare for MCED is reported. SeekInCare incorporates multiple genomic and epigenetic hallmarks, including copy number aberration, fragment size, end motif, and oncogenic virus, via shallow whole-genome sequencing from cell-free DNA, alongside seven protein tumor markers in one tube of blood. Artificial intelligence algorithms were developed to distinguish patients with cancer from individuals without cancer and to predict the likely affected organ. The retrospective study included 617 patients with cancer and 580 individuals without cancer, covering 27 cancer types. SeekInCare achieved 60.0% sensitivity at 98.3% specificity, resulting in an area under the curve of 0.899. Sensitivities were 37.7%, 50.4%, 66.7%, and 78.1% in patients with stage I, II, III, and IV disease, respectively. Additionally, SeekInCare was evaluated in a prospective cohort consisting of 1203 individuals who received the test as a laboratory-developed test (median follow-up time, 753 days) in which it achieved 70.0% sensitivity at 95.2% specificity. The performances of SeekInCare in both retrospective and prospective studies demonstrate that SeekInCare is a blood-based MCED test, showing comparable performance to the other tests currently in development. These findings support its potential clinical utility as a cancer screening test in high-risk populations.
Collapse
Affiliation(s)
- Shiyong Li
- Research and Development, SeekIn Inc., Shenzhen, China
| | | | - Yan Chen
- Research and Development, SeekIn Inc., Shenzhen, China
| | - Qingqi Ren
- Department of Oncology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yi Luan
- Department of Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Weijie Liang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yinyin Chang
- Clinical Laboratories, Shenyou Bio, Zhengzhou, China
| | - Lijuan Zhang
- Clinical Laboratories, Shenyou Bio, Zhengzhou, China
| | - Dandan Zhu
- Clinical Laboratories, Shenyou Bio, Zhengzhou, China
| | - Wei Wu
- Research and Development, SeekIn Inc., Shenzhen, China
| | - Yingying Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Linfeng Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yan Wang
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guolin Zhong
- Research and Development, SeekIn Inc., Shenzhen, China
| | - Bing Wei
- Department of Molecular Pathology, Henan Cancer Hospital, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Jie Ma
- Department of Molecular Pathology, Henan Cancer Hospital, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Yu Chang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xinhua Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhiming Li
- Department of Internal Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Chaohui Duan
- Department of Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guanghui Long
- Department of Oncology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Mao Mao
- Research and Development, SeekIn Inc., San Diego, California; Yonsei Song-Dang Institute for Cancer Research, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
4
|
Juthani R, Manne A. Blood-based biomarkers in pancreatic ductal adenocarcinoma: developments over the last decade and what holds for the future- a review. Front Oncol 2025; 15:1555963. [PMID: 40330826 PMCID: PMC12052548 DOI: 10.3389/fonc.2025.1555963] [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: 01/06/2025] [Accepted: 03/24/2025] [Indexed: 05/08/2025] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) accounts for a significant burden of global cancer deaths worldwide. The dismal outcomes associated with PDAC can be overcome by detecting the disease early and developing tools that predict response to treatment, allowing the selection of the most optimal treatment. Over the last couple of years, significant progress has been made in the development of novel biomarkers that aid in diagnosis, prognosis, treatment selection, and monitoring response. Blood-based biomarkers offer an alternative to tissue-based diagnosis and offer immense potential in managing PDAC. In this review, we have discussed the advances in blood-based biomarkers in PDAC, such as DNA (mutations and methylations), RNA, protein biomarkers and circulating tumor cells (CTC) over the last decade and also elucidated all aspects of practical implementation of these biomarkers in clinical practice. We have also discussed implementing multiomics utilizing more than one biomarker and targeted therapies that have been developed using these biomarkers.
Collapse
Affiliation(s)
- Ronit Juthani
- Department of Medicine, Saint Vincent Hospital, Worcester, MA, United States
| | - Ashish Manne
- Department of Internal Medicine, Division of Medical Oncology at the Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| |
Collapse
|
5
|
Pisapia P, Iaccarino A, Troncone G, Malapelle U. Liquid Biopsy in Solid Tumours: An Overview. Cytopathology 2025. [PMID: 40219616 DOI: 10.1111/cyt.13485] [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: 09/16/2024] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/14/2025]
Abstract
The advent of personalised and precision medicine has radically modified the management and the clinical outcome of cancer patients. However, the expanding number of predictive, prognostic, and diagnostic biomarkers has raised the need for simple, noninvasive, quicker, but equally efficient tests for molecular profiling. In this complex scenario, the adoption of liquid biopsy, particularly circulating tumour DNA (ctDNA), has been a real godsend for many cancer patients who would otherwise have been denied the benefits of targeted treatments. Undeniably, ctDNA analysis has several advantages over conventional tissue-based analysis. One advantage is that it can guide treatment decision making, especially when tissue samples are scarce or totally unavailable. Indeed, a simple blood test can inform clinicians on patients' response or resistance to targeted therapies, help them monitor minimal residual disease (MRD) after surgical resections, and facilitate them with early cancer detection and interception. Finally, an equally important advantage is that ctDNA analysis can help decipher temporal and spatial tumour heterogeneity, a mechanism highly responsible for therapeutic resistance. In this review, we gathered and analysed current evidence on the clinical usefulness of ctDNA analysis in solid tumours.
Collapse
Affiliation(s)
- Pasquale Pisapia
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Antonino Iaccarino
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples Federico II, Naples, Italy
| | - Giancarlo Troncone
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Naples, Italy
| |
Collapse
|
6
|
Kubytskyi V, Khonkhodzhaev M, Tanaka A, Nguyen A, Lazarev A, Aram B, Rogers K, Mourokh L, Lazarev P. Toward In Vivo Cancer Detection: X-Ray Scattering on Thick Phantom Samples. Molecules 2025; 30:1655. [PMID: 40333542 PMCID: PMC12029292 DOI: 10.3390/molecules30081655] [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: 02/01/2025] [Revised: 03/20/2025] [Accepted: 04/04/2025] [Indexed: 05/09/2025] Open
Abstract
As the number of new breast cancer cases grows around the world, there is an unmet need for fast, accurate, and low-cost methods of early cancer detection. It was previously shown that X-ray scattering on lipid molecules can provide the necessary structural biomarker. However, these measurements were performed on small ex vivo samples, and to ensure the progress to in vivo diagnostics, the approach should be extended to larger tissues. We use the phantom fat samples to establish such a procedure. In the obtained X-ray scattering patterns, we observe the characteristic features for the inter-fatty-acid molecular distance. The large size of the samples led to the peak broadening; however, the features remain visible up to 10 cm in thickness. The experimental data are in excellent agreement with the Monte Carlo simulations based on the form factors obtained from the small samples. Our results usher the way for the in vivo monitoring of the structural biomarkers of breast cancer.
Collapse
Affiliation(s)
- Viacheslav Kubytskyi
- Matur UK Ltd., 5 New Street Square, London EC4A 3TW, UK (P.L.)
- Laboratoire de Physique des 2 Infinis Irène Joliot-Curie, UMR9012, CNRS, Université Paris-Saclay, Bât. 209, 91405 Orsay, France
| | - Masroor Khonkhodzhaev
- Matur UK Ltd., 5 New Street Square, London EC4A 3TW, UK (P.L.)
- Physics Department, Queens College, City University of New York, 65-30 Kissena Blvd, Flushing, NY 11367, USA
| | - Aika Tanaka
- EosDx, Inc., 1455 Adams Drive, Menlo Park, CA 94025, USA; (A.T.); (A.N.); (B.A.); (K.R.)
| | - Audrey Nguyen
- EosDx, Inc., 1455 Adams Drive, Menlo Park, CA 94025, USA; (A.T.); (A.N.); (B.A.); (K.R.)
| | - Alexander Lazarev
- EosDx, Inc., 1455 Adams Drive, Menlo Park, CA 94025, USA; (A.T.); (A.N.); (B.A.); (K.R.)
| | - Byron Aram
- EosDx, Inc., 1455 Adams Drive, Menlo Park, CA 94025, USA; (A.T.); (A.N.); (B.A.); (K.R.)
| | - Keith Rogers
- EosDx, Inc., 1455 Adams Drive, Menlo Park, CA 94025, USA; (A.T.); (A.N.); (B.A.); (K.R.)
- Shrivenham Campus, Cranfield University, Swindon SN6 8LA, Wiltshire, UK
| | - Lev Mourokh
- Physics Department, Queens College, City University of New York, 65-30 Kissena Blvd, Flushing, NY 11367, USA
- EosDx, Inc., 1455 Adams Drive, Menlo Park, CA 94025, USA; (A.T.); (A.N.); (B.A.); (K.R.)
| | - Pavel Lazarev
- Matur UK Ltd., 5 New Street Square, London EC4A 3TW, UK (P.L.)
- EosDx, Inc., 1455 Adams Drive, Menlo Park, CA 94025, USA; (A.T.); (A.N.); (B.A.); (K.R.)
| |
Collapse
|
7
|
Kong X, Li C, Li Y, Song X, Huang L. Ultrasensitive determination of exosomes by tyramine-assisted colorimetric sensors for tumor diagnosis. Analyst 2025; 150:1670-1678. [PMID: 40111754 DOI: 10.1039/d5an00013k] [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: 03/22/2025]
Abstract
Exosomes, which are recognized as a kind of valuable liquid biopsy biomarker, exhibit significant application potential in cancer diagnosis. Therefore, it is crucial to establish a reliable detection method for their clinical application. In this study, we have presented an ultrasensitive aptasensor for the visual detection of exosomes by employing tyramine-assisted dual-signal amplification technology. First, we utilized magnetic beads modified with the nucleolin aptamer (MNPs-Aptnucleolin) to capture exosomes. This modification not only enhanced specificity, but also reduced interference of complex sample components. The captured exosomes as a rich source of proteins can bind with multiple biotinyl-tyramide (Bio-TR) molecules through a catalytic reaction involving horseradish peroxidase (HRP) and H2O2. Second, streptavidin-HRP complex-modified gold nanoparticles (GNPs-Str-HRP) as a signal amplification probe was introduced to further enhance the detection signal by binding to Bio-TR. Lastly, the addition of 3,3',5,5'-tetramethylbenzidine (TMB) solution induced a visible color change, enabling quantification of the exosome concentration. This dual-signal amplification strategy resulted in a low limit of detection (LOD) of 63 particles per μL, and it also demonstrated accurate visual diagnosis capabilities for clinical samples. The successful implementation of this approach suggests its potential as a promising tool for point-of-care testing (POCT) in cancer diagnostics.
Collapse
Affiliation(s)
- Xiaoming Kong
- Affiliated Psychological Hospital of Anhui Medical University, Anhui Mental Health Center, Hefei Fourth People's Hospital, 316 Huangshan Road, Hefei, 230022, China
| | - Chao Li
- School of Life Sciences, Anhui Medical University, Hefei, Anhui 230032, China.
| | - Yang Li
- School of Life Sciences, Anhui Medical University, Hefei, Anhui 230032, China.
| | - Xueqian Song
- School of Life Sciences, Anhui Medical University, Hefei, Anhui 230032, China.
| | - Lin Huang
- School of Life Sciences, Anhui Medical University, Hefei, Anhui 230032, China.
| |
Collapse
|
8
|
Elamin I, Rao MS, Figliozzi RW, Maahs JC, Balish M, Hsia SV, Piovezan Fugolin AP, Fan J. Protocol for Extracting Circulating Cell-Free DNA from Murine Saliva: Insights into Oral and Systemic Disease Research. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.31.645839. [PMID: 40236054 PMCID: PMC11996405 DOI: 10.1101/2025.03.31.645839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Circulating cell-free DNA (cfDNA) consists of small fragments of extracellular DNA from mammalian and bacterial cells found in bodily fluids such as blood and saliva, and it has been strongly recognized as a critical biomarker for various disease diagnoses, prognoses, and therapeutic monitoring. In this study, we present a reproducible protocol for efficiently isolating cfDNA from murine saliva using an innovative swabbing method in conjunction with the QIAamp MinElute ccfDNA Mini Kit. The quantification of isolated cfDNA is detected by a Qubit Fluorometer. Moreover, qualification assessment is conducted through BioAnalyzer analysis. This protocol facilitates research on saliva-derived cfDNA in the context of oral and systemic diseases in murine models.
Collapse
|
9
|
Heo Y, Kim WJ, Cho YJ, Jung JW, Kim NS, Choi IY. Advances in cancer genomics and precision oncology. Genes Genomics 2025; 47:399-416. [PMID: 39849190 DOI: 10.1007/s13258-024-01614-7] [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: 11/07/2024] [Accepted: 12/27/2024] [Indexed: 01/25/2025]
Abstract
BACKGROUND Next-generation sequencing has revolutionized genome science over the last two decades. Indeed, the wealth of sequence information on our genome has deepened our understanding on cancer. Cancer is a genetic disease caused by genetic or epigenetic alternations that affect the expression of genes that control cell functions, particularly cell growth and division. Utilization of next-generation sequencing in cancer gene panels has enabled the identification of actionable gene alterations in cancer patients to guide personalized precision medicine. OBJECTIVE The aim is to provide information that can identify actionable gene alterations, enabling personalized precision medicine for cancer patients. RESULTS & DISCUSSION Equipped with next-generation sequencing techniques, international collaboration programs on cancer genomics have identified numerous mutations, gene fusions, microsatellite variations, copy number variations, and epigenetics changes that promote the transformation of normal cells into tumors. Cancer classification has traditionally been based on cell type or tissue-of-origin and the morphological characteristics of the cancer. However, interactive genomic analyses have currently reclassified cancers based on systemic molecular-based taxonomy. Although all cancer-causing genes and mechanisms have yet to be completely understood or identified, personalized or precision medicine is now currently possible for some forms of cancer. Unlike the "one-size-fits-all" approach of traditional medicine, precision medicine allows for customized or personalized treatment based on genomic information. CONCLUSION Despite the availability of numerous cancer gene panels, technological innovation in genomics and expansion of knowledge on the cancer genome will allow precision oncology to manage even more types of cancers.
Collapse
Affiliation(s)
- Yonjong Heo
- Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon, 24341, Gangwon, Republic of Korea
| | - Woo-Jin Kim
- Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon, 24341, Gangwon, Republic of Korea
| | - Yong-Joon Cho
- Department of Molecular Bioscience, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Multidimensional Genomics Research Center, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Jae-Won Jung
- Genetic Sciences Group, Thermo Fisher Scientific Solutions Korea Co., Ltd., Seoul, 06349, Republic of Korea
| | - Nam-Soo Kim
- Department of Molecular Bioscience, Kangwon National University, Chuncheon, 24341, Republic of Korea.
- NBIT Co., Ltd., Chuncheon, 24341, Republic of Korea.
| | - Ik-Young Choi
- Department of Smart Farm and Agricultural Industry, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| |
Collapse
|
10
|
Aydın Ş, Özdemir S, Adıgüzel A. The Potential of cfDNA as Biomarker: Opportunities and Challenges for Neurodegenerative Diseases. J Mol Neurosci 2025; 75:34. [PMID: 40080233 PMCID: PMC11906534 DOI: 10.1007/s12031-025-02317-8] [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: 01/02/2025] [Accepted: 02/06/2025] [Indexed: 03/15/2025]
Abstract
Neurodegenerative disorders, including Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), and amyotrophic lateral sclerosis (ALS), are characterized by the progressive and gradual degeneration of neurons. The prevalence and rates of these disorders rise significantly with age. As life spans continue to increase in many countries, the number of cases is expected to grow in the foreseeable future. Early and precise diagnosis, along with appropriate surveillance, continues to pose a challenge. The high heterogeneity of neurodegenerative diseases calls for more accurate and definitive biomarkers to improve clinical therapy. Cell-free DNA (cfDNA), including fragmented DNA released into bodily fluids via apoptosis, necrosis, or active secretion, has emerged as a promising non-invasive diagnostic tool for various disorders including neurodegenerative diseases. cfDNA can serve as an indicator of ongoing cellular damage and mortality, including neuronal loss, and may provide valuable insights into disease processes, progression, and therapeutic responses. This review will first cover the key aspects of cfDNA and then examine recent advances in its potential use as a biomarker for neurodegenerative disorders.
Collapse
Affiliation(s)
- Şeyma Aydın
- Department of Genetics, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Selçuk Özdemir
- Department of Genetics, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey.
| | - Ahmet Adıgüzel
- Department of Molecular Biology and Genetics, Faculty of Science, Atatürk University, Erzurum, Turkey.
| |
Collapse
|
11
|
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 characterization of metastatic patterns in prostate cancer using circulating tumor DNA data from the SCRUM-Japan MONSTAR SCREEN project. THE JOURNAL OF LIQUID BIOPSY 2025; 7:100282. [PMID: 40027233 PMCID: PMC11863810 DOI: 10.1016/j.jlb.2024.100282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 03/05/2025]
Abstract
Purpose Genomic characterization of the predisposition of tumors to metastasize to specific sites has been performed in a few studies using mainly tissue-derived genomes. This nationwide prospective observational study investigated the association between genomic characteristics using circulating tumor DNA (ctDNA), and the synchronous and metachronous metastasis of tumors to specific target organs in advanced prostate cancer. Methods Patients with advanced prostate cancer undergoing systemic treatment were included. ctDNA was analyzed using the FoundationOne®Liquid CDx assay at enrollment. Associations between genomic characteristics and metastatic status were examined. Results Alterations in the genes MYC, APC, and BRCA2 and the DNA repair, MYC, and WNT pathways were associated with lung and liver metastasis. PTEN gene alterations and PI3K pathway alteration were associated with synchronous lung metastasis. RB1 gene alteration and RAS/RAF/MAPK pathway alteration were associated with synchronous liver metastasis. RB1 and BRCA2 gene alterations predicted metachronous lung metastasis, while TP53 and MYC gene alterations predicted metachronous liver metastasis. Conclusions This study identifies genomic alterations in ctDNA associated with synchronous and metachronous metastases. These findings may be clinically helpful for treating, managing, and monitoring cancer.
Collapse
Affiliation(s)
- Masaki Shiota
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Japan
| | - Nobuaki Matsubara
- Department of Medical Oncology, National Cancer Center Hospital East, Japan
| | - Taigo Kato
- Department of Urology, Osaka University Graduate School of Medicine, Japan
| | - Masatoshi Eto
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Japan
| | - Takahiro Osawa
- Department of Urology, Graduate School of Medicine Hokkaido University, Japan
| | - Takashige Abe
- Department of Urology, Graduate School of Medicine Hokkaido University, Japan
| | - Nobuo Shinohara
- Department of Urology, Graduate School of Medicine Hokkaido University, Japan
| | - Koshiro Nishimoto
- Department of Uro-Oncology, Saitama Medical University International Medical Center, Japan
| | - Yota Yasumizu
- Department of Urology, Keio University School of Medicine, Japan
| | - Nobuyuki Tanaka
- Department of Urology, Keio University School of Medicine, Japan
| | - Mototsugu Oya
- Department of Urology, Keio University School of Medicine, Japan
| | - Takao Fujisawa
- Department of Head and Neck Medical Oncology, National Cancer Center Hospital East, Japan
| | - Satoshi Horasawa
- Translational Research Support Office, National Cancer Center Hospital East, Japan
| | - Yoshiaki Nakamura
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Japan
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Japan
| | - Norio Nonomura
- Department of Urology, Osaka University Graduate School of Medicine, Japan
| |
Collapse
|
12
|
Ursekar R, Chaubal R, Gandhi KA, Talker E, Chitra J, Raja A, Mukhare RS, Singh A, Kadam A, Madhav M, Pandey S, Gupta S. Circulating tumour DNA as a promising biomarker for breast cancer diagnosis & treatment monitoring. Indian J Med Res 2025; 161:267-277. [PMID: 40347499 PMCID: PMC12066141 DOI: 10.25259/ijmr_1291_2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 01/31/2025] [Indexed: 05/14/2025] Open
Abstract
Breast cancer contributes a large fraction to mortality among women diagnosed with cancer. It is important to monitor residual disease and extend the lead time to detect relapse in high-risk patients. Minimally invasive techniques that utilise circulating biomarkers are being explored for their potential in diagnosis, prognosis, and disease monitoring of breast cancer. Circulating biomarkers have been investigated as tools for breast cancer diagnosis, prognosis, prediction, and monitoring of therapeutic response and resistance. Among these, circulating tumour cells and cell-free plasma DNA (cfDNA) derived from tumour cells (circulating tumour DNA i.e. ctDNA) have been integrated into clinical trial designs. Among all circulating biomarkers, ctDNA stands out as a promising biomaterial with great potential as it is thought to mirror the tumour's evolution. However, its clinical utilisation is hampered mainly by gaps in knowledge of its biological properties and specific characteristics. The development of robust and standardised methods for assessing circulating biomarkers is essential for realising the potential of personalised medicine. This review aims to summarise the characteristics of ctDNA and its role in breast cancer, drawing from both basic and translational research to provide insights into its clinical application. This review suggests that ctDNA has the potential to be a non-invasive, real-time surrogate for tumour tissue-based biomarkers. In conclusion, circulating biomarkers have the potential to revolutionise breast cancer diagnosis, prognosis, and treatment monitoring, but the development of standardised methods for their assessment is essential. ctDNA, in particular, shows great promise as a liquid biopsy tool, but further research is needed to understand its biology and ensure its clinical utility fully.
Collapse
Affiliation(s)
- Riddhi Ursekar
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Mumbai, Maharashtra, India
| | - Rohan Chaubal
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Mumbai, Maharashtra, India
- Homi Bhabha National Institute, Training School Complex, Mumbai, Maharashtra, India
- Department of Surgical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, Maharashtra, India
| | - Khushboo A. Gandhi
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Mumbai, Maharashtra, India
| | - Elizabeth Talker
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Mumbai, Maharashtra, India
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, Maharashtra, India
| | - Jaya Chitra
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Mumbai, Maharashtra, India
- Department of Surgical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, Maharashtra, India
| | - Aishwarya Raja
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Mumbai, Maharashtra, India
- Homi Bhabha National Institute, Training School Complex, Mumbai, Maharashtra, India
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, Maharashtra, India
| | - Rushikesh Sunil Mukhare
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Mumbai, Maharashtra, India
- Homi Bhabha National Institute, Training School Complex, Mumbai, Maharashtra, India
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, Maharashtra, India
| | - Ankita Singh
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Mumbai, Maharashtra, India
| | - Anushree Kadam
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Mumbai, Maharashtra, India
| | - Mrudula Madhav
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Mumbai, Maharashtra, India
| | - Shwetali Pandey
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Mumbai, Maharashtra, India
| | - Sudeep Gupta
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Mumbai, Maharashtra, India
- Homi Bhabha National Institute, Training School Complex, Mumbai, Maharashtra, India
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, Maharashtra, India
| |
Collapse
|
13
|
Katopodi XL, Begik O, Novoa E. Toward the use of nanopore RNA sequencing technologies in the clinic: challenges and opportunities. Nucleic Acids Res 2025; 53:gkaf128. [PMID: 40057374 PMCID: PMC11890063 DOI: 10.1093/nar/gkaf128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 01/15/2025] [Accepted: 02/10/2025] [Indexed: 05/13/2025] Open
Abstract
RNA molecules have garnered increased attention as potential clinical biomarkers in recent years. While short-read sequencing and quantitative polymerase chain reaction have been the primary methods for quantifying RNA abundance, they typically fail to capture critical post-transcriptional regulatory elements, such as RNA modifications, which are often dysregulated in disease contexts. A promising cutting-edge technique sequencing method that addresses this gap is direct RNA sequencing, offered by Oxford Nanopore Technologies, which can simultaneously capture both RNA abundance and modification information. The rapid advancements in this platform, along with growing evidence of dysregulated RNA species in biofluids, presents a compelling clinical opportunity. In this review, we discuss the challenges and the emerging opportunities for the adoption of nanopore RNA sequencing technologies in the clinic, highlighting their potential to revolutionize personalized medicine and disease monitoring.
Collapse
Affiliation(s)
- Xanthi-Lida Katopodi
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Oguzhan Begik
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain
| | - Eva Maria Novoa
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- ICREA, Pg. Lluís Companys 23, Barcelona 08010, Spain
| |
Collapse
|
14
|
Wotman MT, Xiao W, Du RR, Jiang B, Akagi K, Liu S, Gillison ML. Development and Validation of an Assay to Quantify Plasma Circulating Tumor Human Papillomavirus DNA for 13 High-Risk Types that Cause 98% of HPV-Positive Cancers. Head Neck Pathol 2025; 19:25. [PMID: 39998590 PMCID: PMC11861489 DOI: 10.1007/s12105-025-01752-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 01/18/2025] [Indexed: 02/27/2025]
Abstract
PURPOSE Plasma circulating tumor HPV DNA (ctHPVDNA) persistence after curative-intent treatment may identify patients with HPV-positive cancers at risk for recurrence. Technical validation is required for use as an integral biomarker in a prospective clinical trial. METHODS Development and analytical validation of a digital droplet PCR assay for detection and quantification of 13 high-risk HPV types (i.e., Cell-Free 13) was performed with oligonucleotides/plasmids encoding type-specific E6/E7 coding regions. Clinical performance, determinants of detection/quantification, and associations of pre-treatment ctHPVDNA with progression-free survival (PFS) were also evaluated in a prospective cohort of 272 head and neck cancer patients. RESULTS Limit of detection, limit of quantification, and linear range of quantification were 5, 16 and 16-200,000 virus copies for all 13 high-risk HPV types. No cross-reactivity was detected across all 13 HPV types. At 10,000 copies, inter-assay coefficients of variation ranged from 0.3 to 4.6%. Multiplexing, DNA purification method, input plasma volume, total input cell-free (< 1800 ng) or genomic (< 700 ng) DNA did not affect HPV detection or quantification. The assay had a sensitivity of 91.7% (95%CI 87.3-94.9%) and specificity of 97.7% (95%CI 87.7-99.9%) for ctHPVDNA detection in the setting of newly diagnosed HPV-positive oropharyngeal cancer. Tumor and nodal stage categories, tumor viral load (ρ = 0.41, p < 0.05), and HPV integration status were associated with ctHPVDNA quantitative level. Pre-treatment ctHPVDNA greater than the median (231 copies/ml) was associated with worse PFS (HR = 2.14, 95%CI 1.16-3.97, p = 0.0156) in univariate analysis. However, this was no longer significant after adjustment for clinical covariates (HRadj = 1.81, 95%CI 0.97-3.37, p = 0.0635). CONCLUSION Cell-Free 13 demonstrated excellent analytical performance and clinical sensitivity/specificity in HPV-positive oropharyngeal cancer. Pre-treatment ctHPVDNA may be associated with oncologic outcomes.
Collapse
Affiliation(s)
- Michael T Wotman
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Weihong Xiao
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Holcombe Boulevard, Unit 432, Houston, TX, 77030, USA
| | - Robyn R Du
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Holcombe Boulevard, Unit 432, Houston, TX, 77030, USA
| | - Bo Jiang
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Holcombe Boulevard, Unit 432, Houston, TX, 77030, USA
| | - Keiko Akagi
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Holcombe Boulevard, Unit 432, Houston, TX, 77030, USA
| | - Suyu Liu
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Maura L Gillison
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Holcombe Boulevard, Unit 432, Houston, TX, 77030, USA.
| |
Collapse
|
15
|
Fernández-Acosta R, Vintea I, Koeken I, Hassannia B, Vanden Berghe T. Harnessing ferroptosis for precision oncology: challenges and prospects. BMC Biol 2025; 23:57. [PMID: 39988655 PMCID: PMC11849278 DOI: 10.1186/s12915-025-02154-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 02/12/2025] [Indexed: 02/25/2025] Open
Abstract
The discovery of diverse molecular mechanisms of regulated cell death has opened new avenues for cancer therapy. Ferroptosis, a unique form of cell death driven by iron-catalyzed peroxidation of membrane phospholipids, holds particular promise for targeting resistant cancer types. This review critically examines current literature on ferroptosis, focusing on its defining features and therapeutic potential. We discuss how molecular profiling of tumors and liquid biopsies can generate extensive multi-omics datasets, which can be leveraged through machine learning-based analytical approaches for patient stratification. Addressing these challenges is essential for advancing the clinical integration of ferroptosis-driven treatments in cancer care.
Collapse
Affiliation(s)
- Roberto Fernández-Acosta
- Cell Death Signaling lab, Infla-Med Centre of Excellence, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Iuliana Vintea
- Cell Death Signaling lab, Infla-Med Centre of Excellence, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Biobix, Lab of Bioinformatics and Computational Genomics, Department of Mathematical Modelling, Statistics and Bioinformatics, Ghent University, Ghent, Belgium
| | - Ine Koeken
- Cell Death Signaling lab, Infla-Med Centre of Excellence, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Behrouz Hassannia
- Cell Death Signaling lab, Infla-Med Centre of Excellence, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Tom Vanden Berghe
- Cell Death Signaling lab, Infla-Med Centre of Excellence, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
- VIB-UGent Center for Inflammation Research, Ghent, Belgium.
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
| |
Collapse
|
16
|
Rowley SD, Albitar M, Baker MF, Ali A, Kaur S, Suh HC, Goy A, Donato ML. cfDNA Chimerism and Somatic Mutation Testing in Early Prediction of Relapse After Allogeneic Stem Cell Transplantation for Myeloid Malignancies. Cancers (Basel) 2025; 17:625. [PMID: 40002220 PMCID: PMC11853444 DOI: 10.3390/cancers17040625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/03/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Disease relapse is a primary cause of treatment failure after hematopoietic stem cell transplantation in the treatment of malignancy. Consolidation therapy early after transplantation may reduce this risk, but it is difficult to administer in the setting of various post-transplant complications. We proposed that testing donor cell chimerism and for persistent minimal residual disease (MRD) with next-generation sequencing (NGS) of plasma cell-free DNA (cfDNA) early after transplantation would identify those patients at higher risk of relapse who would possibly benefit from consolidation therapy. Methods: We enrolled 20 subjects with known tumor-associated somatic mutations into this prospective pilot study, testing plasma samples before and at 28, 56, and 84 days after transplantation. Pre- and post-transplant bone marrow samples were also analyzed. All samples were subjected to an agnostic, commercially available panel covering 302 genes. Results: Significantly more mutations (p < 0.0001) were detected in the plasma cfDNA than in the bone marrow cells in pre-transplant testing (92 versus 61 mutations, respectively), most likely reflecting sampling variation when bone marrow was used. Two subjects were negative for MRD in staging studies immediately before transplants. Most (19/20) subjects had intermittent or sustained MRD detected in post-transplant plasma cfDNA testing, albeit with much lower average variant allele frequencies (VAFs). Six out of 20 subjects suffered relapses within 12 months after transplantation, and all 6 could be identified by adverse-risk driver mutations that persisted after transplantation. No patients who cleared the adverse-risk mutations relapsed. Donor chimerism using cfDNA fell for all relapsed patients and contributed to the identification of patients at early risk for relapse. Conclusions: These data demonstrate that testing plasma cfDNA for persistent leukemia-associated somatic mutations and donor chimerism as early as 28 days after transplantation will identify a subset of patients with high-risk mutations who are at high risk of relapse. This early assessment of relapse risk may facilitate modifications to the treatment plan, reducing the risk of treatment failure.
Collapse
Affiliation(s)
- Scott D. Rowley
- John Theurer Cancer Center, 92 Second St., Hackensack, NJ 07601, USA; (M.F.B.); (S.K.); (H.C.S.); (A.G.); (M.L.D.)
- Lombardi Comprehensive Cancer Center, Georgetown University School of Medicine, 3800 Reservoir Rd. NW, Washington, DC 20057, USA;
| | - Maher Albitar
- Genomic Testing Cooperative, 25371 Commercentre Dr., Lake Forest, CA 92630, USA;
| | - Melissa F. Baker
- John Theurer Cancer Center, 92 Second St., Hackensack, NJ 07601, USA; (M.F.B.); (S.K.); (H.C.S.); (A.G.); (M.L.D.)
| | - Alaa Ali
- Lombardi Comprehensive Cancer Center, Georgetown University School of Medicine, 3800 Reservoir Rd. NW, Washington, DC 20057, USA;
| | - Sukhdeep Kaur
- John Theurer Cancer Center, 92 Second St., Hackensack, NJ 07601, USA; (M.F.B.); (S.K.); (H.C.S.); (A.G.); (M.L.D.)
| | - Hyung C. Suh
- John Theurer Cancer Center, 92 Second St., Hackensack, NJ 07601, USA; (M.F.B.); (S.K.); (H.C.S.); (A.G.); (M.L.D.)
| | - Andre Goy
- John Theurer Cancer Center, 92 Second St., Hackensack, NJ 07601, USA; (M.F.B.); (S.K.); (H.C.S.); (A.G.); (M.L.D.)
| | - Michele L. Donato
- John Theurer Cancer Center, 92 Second St., Hackensack, NJ 07601, USA; (M.F.B.); (S.K.); (H.C.S.); (A.G.); (M.L.D.)
| |
Collapse
|
17
|
Gonzalez Bosquet J, Wagner V, Polio A, Linder KE, Bender DP, Goodheart MJ, Schickling BM. Identification of Ovarian High-Grade Serous Carcinoma with Mitochondrial Gene Variation. Int J Mol Sci 2025; 26:1347. [PMID: 39941116 PMCID: PMC11818617 DOI: 10.3390/ijms26031347] [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: 12/27/2024] [Revised: 02/02/2025] [Accepted: 02/03/2025] [Indexed: 02/16/2025] Open
Abstract
Women diagnosed with advanced-stage ovarian cancer have a much worse survival rate than women diagnosed with early-stage ovarian cancer, but the early detection of this disease remains a clinical challenge. Some recent reports indicate that genetic variations could be useful for the early detection of several malignancies. In this pilot observational retrospective study, we aimed to assess whether mitochondrial DNA (mtDNA) variations could discriminate the most frequent type of ovarian cancer, high-grade serous carcinoma (HGSC), from normal tissue. We identified mtDNA variations from 20 whole-exome sequenced (WES) HGSC samples and 14 controls (normal tubes) using the best practices of genome sequencing. We built prediction models of cancer with these variants, with good performance measured by the area under the curve (AUC) of 0.88 (CI: 0.74-1.00). The variants included in the best model were correlated with gene expression to assess the potentially affected processes. These analyses were validated with the Cancer Genome Atlas (TCGA) dataset, (including over 420 samples), with a fair performance in AUC terms (0.63-0.71). In summary, we identified a set of mtDNA variations that can discriminate HGSC with good performance. Specifically, variations in the MT-CYB gene increased the risk for HGSC by over 30%, and MT-CYB expression was significantly decreased in HGSC patients. Robust models of ovarian cancer detection with mtDNA variations could be applied to liquid biopsy technology, like those which have been applied to other cancers, with a special focus on the early detection of this lethal disease.
Collapse
Affiliation(s)
- Jesus Gonzalez Bosquet
- Department of Obstetrics and Gynecology, University of Iowa, Iowa City, IA 52242, USA (A.P.); (K.E.L.); (D.P.B.); (B.M.S.)
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Vincent Wagner
- Department of Obstetrics and Gynecology, University of Iowa, Iowa City, IA 52242, USA (A.P.); (K.E.L.); (D.P.B.); (B.M.S.)
| | - Andrew Polio
- Department of Obstetrics and Gynecology, University of Iowa, Iowa City, IA 52242, USA (A.P.); (K.E.L.); (D.P.B.); (B.M.S.)
| | - Katharine E. Linder
- Department of Obstetrics and Gynecology, University of Iowa, Iowa City, IA 52242, USA (A.P.); (K.E.L.); (D.P.B.); (B.M.S.)
| | - David P. Bender
- Department of Obstetrics and Gynecology, University of Iowa, Iowa City, IA 52242, USA (A.P.); (K.E.L.); (D.P.B.); (B.M.S.)
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Michael J. Goodheart
- Department of Obstetrics and Gynecology, University of Iowa, Iowa City, IA 52242, USA (A.P.); (K.E.L.); (D.P.B.); (B.M.S.)
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Brandon M. Schickling
- Department of Obstetrics and Gynecology, University of Iowa, Iowa City, IA 52242, USA (A.P.); (K.E.L.); (D.P.B.); (B.M.S.)
| |
Collapse
|
18
|
Delrue C, De Bruyne S, Speeckaert MM. The Promise of Infrared Spectroscopy in Liquid Biopsies for Solid Cancer Detection. Diagnostics (Basel) 2025; 15:368. [PMID: 39941298 PMCID: PMC11818004 DOI: 10.3390/diagnostics15030368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/15/2025] [Accepted: 02/01/2025] [Indexed: 02/16/2025] Open
Abstract
Attenuated total reflection-Fourier transform infrared (ATR-FTIR) spectroscopy has shown significant promise in the context of liquid biopsy, offering a potential tool for cancer diagnostics. Unlike traditional tissue biopsies, which may not fully capture the clonal heterogeneity of tumors, liquid biopsy reflects the dynamic state of the disease and its progression more comprehensively. Biofluids such as serum and plasma are low-cost, minimally invasive diagnostic media with well-established clinical uses. This review assesses the use of ATR-FTIR spectroscopy to detect biochemical changes in biofluids linked to various malignancies, including breast, ovarian, endometrial, prostate, bladder, kidney, pancreatic, colorectal, hepatic, esophageal, gastric, lung, and brain cancers. While ATR-FTIR offers the advantages of rapid, minimally invasive detection and real-time disease monitoring, its integration into clinical practice faces challenges, particularly in terms of reproducibility due to variability in sample preparation, spectral acquisition, and data processing. The translation of ATR-FTIR into routine diagnostics will require validation through large-scale cohort studies and multicenter trials to ensure its clinical reliability and effectiveness.
Collapse
Affiliation(s)
- Charlotte Delrue
- Department of Nephrology, Ghent University Hospital, 9000 Ghent, Belgium;
| | - Sander De Bruyne
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, Belgium;
- Department of Laboratory Medicine, AZ Sint-Blasius, 9200 Dendermonde, Belgium
| | - Marijn M. Speeckaert
- Department of Nephrology, Ghent University Hospital, 9000 Ghent, Belgium;
- Research Foundation-Flanders (FWO), 1000 Brussels, Belgium
| |
Collapse
|
19
|
Veronese N, Luchini C, Ciriminna S, Spinelli K, Fruscione S, Mattiolo P, Belluzzo M, Messina V, Smith L, Barbagallo M, Mazzucco W. Potentialities and critical issues of liquid biopsy in clinical practice: An umbrella review. Transl Oncol 2025; 52:102172. [PMID: 39817953 PMCID: PMC11786759 DOI: 10.1016/j.tranon.2024.102172] [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: 08/20/2024] [Revised: 10/09/2024] [Accepted: 10/29/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Liquid biopsy (LB) is a laboratory test performed on a fluid sample aiming at analyzing molecular data derived from circulating cells and related entities, or from nucleic acids. This umbrella review aims to map and evaluate the evidence supporting the use of LB in medicine across different medical specialities and conditions. METHODS We searched three repositories from database inception up to October 1, 2023 and we included meta-analyses of observational studies reporting data on the use of LB, compared to gold standard, and its accuracy (area under the curve, AUC). RESULTS Among 726 articles initially screened, 42 systematic reviews were included. Most of the outcomes explored (202/211) were related to cancer. We found that 75/211 had an excellent accuracy (AUC >0.90), with one comparison with an AUC equal to 1, i.e., Cell-Free Human Papillomavirus DNA (cfHPV-DNA) for HPV-positive oropharyngeal squamous cell carcinoma. However, considering published meta-analyses, all the outcomes were graded as very low on the GRADE criteria, and the heterogeneity was never reported. DISCUSSION The literature about LB is rapidly increasing and some promising data about precision oncology are now available. However, this umbrella review on existing meta-analyses highlighted some critical issues for providing quantitative estimations on the different roles of LB.
Collapse
Affiliation(s)
- Nicola Veronese
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy.
| | - Claudio Luchini
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Stefano Ciriminna
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Katia Spinelli
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Santo Fruscione
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Paola Mattiolo
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Miriam Belluzzo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Veronica Messina
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Lee Smith
- Centre for Health Performance and Wellbeing, Anglia Ruskin University, Cambridge, UK
| | - Mario Barbagallo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Walter Mazzucco
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy; Division of Biostatistics & Epidemiology Research, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, United States
| |
Collapse
|
20
|
Jacobsen CM, Matos do Canto L, Kahns S, Hansen TF, Andersen RF. What the Clinician Needs to Know About Laboratory Analyses of Circulating Tumor DNA. Clin Colorectal Cancer 2025:S1533-0028(25)00003-9. [PMID: 39956753 DOI: 10.1016/j.clcc.2025.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 12/02/2024] [Accepted: 01/20/2025] [Indexed: 02/18/2025]
Abstract
Liquid biopsies offer the possibility to evaluate cancer patients using noninvasive approaches. Circulating cell-free DNA (ccfDNA) is 1 of the most used and promising sources. Detecting tumor DNA among ccfDNA (ctDNA) can be used for early cancer detection, treatment response assessment, prognosis, and predictive evaluations. Providing analyses that can increase the quality of patient treatment is very much a joint effort between laboratory scientists and clinicians. With its use approaching clinical practice, it is important for clinicians to be familiar with the basic concepts and analyses behind ctDNA results in a similar way as laboratory scientists should have knowledge of the clinical needs to provide relevant analyses. In this Perspective, we describe the whole process of ctDNA analyses, from the preanalytical standards to reporting/analyzing results, and highlight some important factors that need to be addressed in the process of implementing them to clinical practice.
Collapse
Affiliation(s)
- Cecilie Mondrup Jacobsen
- Department of Biochemistry and Immunology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark; Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark
| | - Luisa Matos do Canto
- Department of Biochemistry and Immunology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark; Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark
| | - Søren Kahns
- Department of Biochemistry and Immunology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark
| | - Torben Frøstrup Hansen
- Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark; Department of Regional Health Research, University of Southern Denmark, Odense C, Denmark
| | - Rikke Fredslund Andersen
- Department of Biochemistry and Immunology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark; Department of Oncology, Vejle Hospital, University Hospital of Southern Denmark, Vejle, Denmark.
| |
Collapse
|
21
|
Quesada S, Penault-Llorca F, Matias-Guiu X, Banerjee S, Barberis M, Coleman RL, Colombo N, DeFazio A, McNeish IA, Nogueira-Rodrigues A, Oaknin A, Pignata S, Pujade-Lauraine É, Rouleau É, Ryška A, Van Der Merwe N, Van Gorp T, Vergote I, Weichert W, Wu X, Ray-Coquard I, Pujol P. Homologous recombination deficiency in ovarian cancer: Global expert consensus on testing and a comparison of companion diagnostics. Eur J Cancer 2025; 215:115169. [PMID: 39693891 DOI: 10.1016/j.ejca.2024.115169] [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: 11/07/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND Poly (ADP ribose) polymerase inhibitors (PARPis) are a treatment option for patients with advanced high-grade serous or endometrioid ovarian carcinoma (OC). Recent guidelines have clarified how homologous recombination deficiency (HRD) may influence treatment decision-making in this setting. As a result, numerous companion diagnostic assays (CDx) have been developed to identify HRD. However, the optimal HRD testing strategy is an area of debate. Moreover, recently published clinical and translational data may impact how HRD status may be used to identify patients likely to benefit from PARPi use. We aimed to extensively compare available HRD CDx and establish a worldwide expert consensus on HRD testing in primary and recurrent OC. METHODS A group of 99 global experts from 31 different countries was formed. Using a modified Delphi process, the experts aimed to establish consensus statements based on a systematic literature search and CDx information sought from investigators, companies and/or publications. RESULTS Technical information, including analytical and clinical validation, were obtained from 14 of 15 available HRD CDx (7 academic; 7 commercial). Consensus was reached on 36 statements encompassing the following topics: 1) the predictive impact of HRD status on PARPi use in primary and recurrent OC; 2) analytical and clinical validation requirements of HRD CDx; 3) resource-stratified HRD testing; and 4) how future CDx may include additional approaches to help address unmet testing needs. CONCLUSION This manuscript provides detailed information on currently available HRD CDx and up-to-date guidance from global experts on HRD testing in patients with primary and recurrent OC.
Collapse
Affiliation(s)
- Stanislas Quesada
- Department of Medical Oncology, Institut régional du Cancer de Montpellier (ICM), Montpellier, France; Department of Cancer Genetics, University Hospital of Montpellier, Montpellier, France; Groupe d'Investigateurs Nationaux pour l'Etude des cancers de l'ovaire et du sein (GINECO), Paris, France; Société Française de Médecine Prédictive et Personnalisée (SFMPP), Montpellier, France
| | - Frédérique Penault-Llorca
- Société Française de Médecine Prédictive et Personnalisée (SFMPP), Montpellier, France; Department of Biology and Pathology, Centre de Lutte Contre le Cancer Jean Perrin, Imagerie Moléculaire et Stratégies Théranostiques, Université Clermont Auvergne, UMR 1240 INSERM-UCA, Clermont-Ferrand, France; Cours St Paul, Saint Paul, Réunion, France
| | - Xavier Matias-Guiu
- Department of Pathology, Hospital Universitari Arnau de Vilanova, IRBLLEIDA, University of Lleida, Lleida, Spain; Department of Pathology, Hospital Universitari de Bellvitge, IDIBELL, University of Barcelona, Barcelona, Spain; European Society of Pathology (ESP), Belgium
| | - Susana Banerjee
- The Royal Marsden NHS Foundation Trust and Institute of Cancer Research, London, UK
| | - Massimo Barberis
- Division of Experimental Oncology, European Institute of Oncology, IRCCS, Milan, Italy
| | | | - Nicoletta Colombo
- Gynecologic Oncology Program, European Institute of Oncology IRCCS, Milan, Italy; Department of Medicine and Surgery, University of Milan-Bicocca, Milan, Italy
| | - Anna DeFazio
- Centre for Cancer Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; Department of Gynaecological Oncology, Westmead Hospital, Sydney, NSW, Australia; The Daffodil Centre, The University of Sydney, a joint venture with Cancer Council NSW, Sydney, NSW, Australia
| | - Iain A McNeish
- Division of Cancer and Ovarian Cancer Action Research Centre, Department of Surgery & Cancer, Imperial College London, London, UK
| | - Angélica Nogueira-Rodrigues
- Federal University MG, Brazilian Group of Gynecologic Oncology (EVA), Latin American Cooperative Oncology Group (LACOG), Oncoclínicas, DOM Oncologia, Brazil
| | - Ana Oaknin
- Medical Oncology Service, Vall d'Hebron Institute of Oncology, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Sandro Pignata
- Department of Urology and Gynecology, Istituto Nazionale Tumori di Napoli, IRCCS Fondazione Pascale, Napoli, Italy
| | - Éric Pujade-Lauraine
- Association de Recherche Cancers Gynécologiques - Groupe d'Investigateurs Nationaux pour l'Etude des Cancers de l'ovaire et du Sein (ARCAGY-GINECO), Paris, France
| | - Étienne Rouleau
- Coordinator of Gen&Tiss GFCO, Université Paris-Saclay, Gustave-Roussy Cancer Campus, Inserm U981, Villejuif, France; Cancer Genetics Laboratory, Medical Biology and Pathology Department, Gustave-Roussy Cancer Campus, Villejuif, France
| | - Aleš Ryška
- European Society of Pathology (ESP), Belgium; The Fingerland Department of Pathology, Faculty of Medicine, Charles University and University Hospital, Hradec Kralove, Czech Republic
| | - Nerina Van Der Merwe
- Division of Human Genetics, National Health Laboratory Service, Universitas Hospital, Bloemfontein, South Africa; Division of Human Genetics, Faculty of Health Sciences, University of the Free State, Bloemfontein, South Africa
| | - Toon Van Gorp
- Division of Gynaecological Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium; Belgium and Luxembourg Gynaecological Oncology Group (BGOG), Leuven, Belgium
| | - Ignace Vergote
- Division of Gynaecological Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium; Belgium and Luxembourg Gynaecological Oncology Group (BGOG), Leuven, Belgium
| | - Wilko Weichert
- Institute of Pathology, School of Medicine and Health, Technical University Munich, Munich, Germany
| | - Xiaohua Wu
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Isabelle Ray-Coquard
- Groupe d'Investigateurs Nationaux pour l'Etude des cancers de l'ovaire et du sein (GINECO), Paris, France; Medical Oncology, Centre Léon Bérard and Université Claude Bernard Lyon, Lyon, France
| | - Pascal Pujol
- Department of Medical Oncology, Institut régional du Cancer de Montpellier (ICM), Montpellier, France; Société Française de Médecine Prédictive et Personnalisée (SFMPP), Montpellier, France; Center for Ecological and Evolutionary Cancer Research (CREEC), Montpellier University, Montpellier, France.
| |
Collapse
|
22
|
Schettini F, Sirico M, Loddo M, Williams GH, Hardisty KM, Scorer P, Thatcher R, Rivera P, Milani M, Strina C, Ferrero G, Ungari M, Bottin C, Zanconati F, de Manzini N, Aguggini S, Tancredi R, Fiorio E, Fioravanti A, Scaltriti M, Generali D. Next-generation sequencing-based evaluation of the actionable landscape of genomic alterations in solid tumors: the "MOZART" prospective observational study. Oncologist 2025; 30:oyae206. [PMID: 39177668 PMCID: PMC11783315 DOI: 10.1093/oncolo/oyae206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 07/10/2024] [Indexed: 08/24/2024] Open
Abstract
BACKGROUND The identification of the most appropriate targeted therapies for advanced cancers is challenging. We performed a molecular profiling of metastatic solid tumors utilizing a comprehensive next-generation sequencing (NGS) assay to determine genomic alterations' type, frequency, actionability, and potential correlations with PD-L1 expression. METHODS A total of 304 adult patients with heavily pretreated metastatic cancers treated between January 2019 and March 2021 were recruited. The CLIA-/UKAS-accredit Oncofocus assay targeting 505 genes was used on newly obtained or archived biopsies. Chi-square, Kruskal-Wallis, and Wilcoxon rank-sum tests were used where appropriate. Results were significant for P < .05. RESULTS A total of 237 tumors (78%) harbored potentially actionable genomic alterations. Tumors were positive for PD-L1 in 68.9% of cases. The median number of mutant genes/tumor was 2.0 (IQR: 1.0-3.0). Only 34.5% were actionable ESCAT Tier I-II with different prevalence according to cancer type. The DNA damage repair (14%), the PI3K/AKT/mTOR (14%), and the RAS/RAF/MAPK (12%) pathways were the most frequently altered. No association was found among PD-L1, ESCAT, age, sex, and tumor mutational status. Overall, 62 patients underwent targeted treatment, with 37.1% obtaining objective responses. The same molecular-driven treatment for different cancer types could be associated with opposite clinical outcomes. CONCLUSIONS We highlight the clinical value of molecular profiling in metastatic solid tumors using comprehensive NGS-based panels to improve treatment algorithms in situations of uncertainty and facilitate clinical trial recruitment. However, interpreting genomic alterations in a tumor type-specific manner is critical.
Collapse
Affiliation(s)
- Francesco Schettini
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Medical Oncology Department, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
- Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
| | - Marianna Sirico
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori,”47014, Meldola, Italy
| | - Marco Loddo
- Oncologica UK Ltd, Cambridge CB10 1XL, United Kingdom
| | | | | | - Paul Scorer
- Oncologica UK Ltd, Cambridge CB10 1XL, United Kingdom
| | | | - Pablo Rivera
- Medical Oncology Department, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
| | - Manuela Milani
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34147, Trieste, Italy
| | - Carla Strina
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34147, Trieste, Italy
| | - Giuseppina Ferrero
- Multidisciplinary Unit of Breast Pathology and Translational Research, Cremona Hospital, 26100, Cremona, Italy
| | - Marco Ungari
- Multidisciplinary Unit of Breast Pathology and Translational Research, Cremona Hospital, 26100, Cremona, Italy
| | - Cristina Bottin
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34147, Trieste, Italy
| | - Fabrizio Zanconati
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34147, Trieste, Italy
| | - Nicolò de Manzini
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34147, Trieste, Italy
| | - Sergio Aguggini
- Multidisciplinary Unit of Breast Pathology and Translational Research, Cremona Hospital, 26100, Cremona, Italy
| | - Richard Tancredi
- Multidisciplinary Unit of Breast Pathology and Translational Research, Cremona Hospital, 26100, Cremona, Italy
| | - Elena Fiorio
- Section of Oncology, Department of Medicine, University of Verona School of Medicine and Verona University Hospital Trust, 37134, Verona, Italy
| | | | - Maurizio Scaltriti
- Neurosurgery Unit, ASST Cremona, 26100, Cremona, Italy
- AstraZeneca, Gaithersburg, MD 20876, United States
| | - Daniele Generali
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34147, Trieste, Italy
- Multidisciplinary Unit of Breast Pathology and Translational Research, Cremona Hospital, 26100, Cremona, Italy
| |
Collapse
|
23
|
Luo C, Lin Z, Huang F, Ning L, Yuan Y. Tumor-Educated Platelets lncRNA-STARD4-AS1 and ELOA-AS1 as Potential Novel Biomarkers for the Early Diagnosis of Non-Small Cell Lung Cancer. Cancer Manag Res 2025; 17:1-9. [PMID: 39781562 PMCID: PMC11705986 DOI: 10.2147/cmar.s498516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 12/14/2024] [Indexed: 01/12/2025] Open
Abstract
Purpose (Tumor-educated platelets) TEPs have emerged as active players in all steps of tumorigenesis, confrontation of platelets with tumor cells via transfer of tumor-associated biomolecules and results in the sequestration of such biomolecules. The current study was aimed to examine whether TEPs lncRNA-STARD4-AS1 and ELOA-AS1 might be potential biomarkers for NSCLC. Materials and Methods TEPs were obtained by low-speed centrifugation. Quantitative real-time PCR was used to determine the expression level of TEPs-STARD4-AS1, ELOA-AS1 in the training cohort and the validation cohort. ROC curve was generated to evaluate their diagnostic value. Correlations between TEPs-STARD4-AS1, ELOA-AS1 and clinical parameters were further analyzed. Results Our results showed that the level of TEPs-STARD4-AS1 and ELOA-AS1 significantly upregulated in patients with NSCLC compared with healthy controls in the two cohorts. By ROC analysis, we found that TEPs-STARD4-AS1, ELOA-AS1 could offer valuable diagnostic performance for NSCLC patients (AUCSTARD4-AS1 = 0.800/0.774, and AUCELOA-AS1 = 0.754/0.718 for diagnosing adenocarcinoma and squamous cell carcinoma cases from controls, respectively). The combination of TEP-STARD4-AS1 and ELOA-AS1 improved the diagnostic efficiency of NSCLC. Clinicopathological analysis further revealed that TEPs-STARD4-AS1 level significantly correlated with tumor-node-metastasis (TNM) stage (p = 0.011), while TEPs-ELOA-AS1 expression significantly correlated with tumor-node-metastasis (TNM) stage and (p = 0.019) distant metastasis (p = 0.004). Conclusion Our data suggested that TEPs-STARD4-AS1 and ELOA-AS1 are promising non-invasive circulating diagnostic markers for NSCLC.
Collapse
Affiliation(s)
- ChangLiang Luo
- Department of Clinical Laboratory, The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, People’s Republic of China
| | - Zhongyuan Lin
- Department of Clinical Laboratory, The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, People’s Republic of China
| | - Fangfang Huang
- Department of Clinical Laboratory, The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, People’s Republic of China
| | - Leping Ning
- Department of Clinical Laboratory, The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, People’s Republic of China
| | - Yulin Yuan
- Department of Clinical Laboratory, The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, People’s Republic of China
| |
Collapse
|
24
|
Hu Y, Zhang Y, Wang S, Wang R, Yuan Q, Zhu L, Xia F, Xue M, Wang Y, Li Y, Yuan C. LINC00667: A Novel Vital Oncogenic LincRNA. Curr Med Chem 2025; 32:678-687. [PMID: 37855347 DOI: 10.2174/0109298673248494231010044348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 06/14/2023] [Accepted: 09/11/2023] [Indexed: 10/20/2023]
Abstract
Long intergenic noncoding RNAs (lincRNAs) have a variety of properties that differ from those of messenger RNAs (mRNAs) encoding proteins. Long intergenic nonprotein coding RNA 667 (LINC00667) is a non-coding transcript located on chromosome 18p11.31. Recently, many studies have found that LINC00667 can enhance the progression of various cancers and play a key part in a lot of diseases, such as tumorigenesis. Therefore, LINC00667 can be recognized as a potential biomarker and therapeutic target. So, we reviewed the biological functions, relevant mechanisms, as well as clinical significance of LINC00667 in several human cancers in detail.
Collapse
Affiliation(s)
- Yaqi Hu
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Yifan Zhang
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Shuwen Wang
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Rui Wang
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Qi Yuan
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- College of Medicine and Health Science, China Three Gorges University, Yichang, 443002, China
| | - Leiqi Zhu
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Fangqi Xia
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Mengzhen Xue
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Yaqi Wang
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Yuanyang Li
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- College of Medicine and Health Science, China Three Gorges University, Yichang, 443002, China
| | - Chengfu Yuan
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| |
Collapse
|
25
|
Yuan S, Jia N, Lu G, Lai J, Liang W, Li L, Zhang C, Diao J. Development and validation of an ultrasensitive qPCR method to identify and quantify EGFR T790M in cell-free DNA. Bioanalysis 2025; 17:49-62. [PMID: 39812332 PMCID: PMC11749345 DOI: 10.1080/17576180.2025.2451527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 01/07/2025] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Circulating tumor DNA (ctDNA) is a promising biomarker for cancer prognosis and drug development. A major challenge in the ctDNA determination method is discriminating ctDNA from highly similar but significantly more abundant wild-type DNA sensitively and accurately. METHOD An ultrasensitive qPCR method termed Triple Enrichment Amplification of Mutation PCR (TEAM-PCR) was developed to detect EGFR T790M mutation. RESULTS EGFR T790M was quantified over the assay range of 25-106 copies/reaction in the presence of 106 wild-type copies. This method was fully validated following the essential bioanalysis guidance, with the limit of detection (LOD) being five copies/reaction. CONCLUSION This study established and validated a qPCR-based strategy to detect EGFR T790M mutation with ultra-high sensitivity and reliability.
Collapse
Affiliation(s)
- Shenglei Yuan
- Bioanalytical Services Department, WuXi AppTec (Shanghai) Co. Ltd, Shanghai, China
| | - Nan Jia
- Bioanalytical Services Department, WuXi AppTec (Shanghai) Co. Ltd, Shanghai, China
| | - Guofu Lu
- Bioanalytical Services Department, WuXi AppTec (Shanghai) Co. Ltd, Shanghai, China
| | - Jinping Lai
- Bioanalytical Services Department, WuXi AppTec, Plainsboro, NJ, USA
| | - Wenzhong Liang
- Bioanalytical Services Department, WuXi AppTec (Shanghai) Co. Ltd, Shanghai, China
| | - Lan Li
- Bioanalytical Services Department, WuXi AppTec (Shanghai) Co. Ltd, Shanghai, China
| | - Chenpu Zhang
- Bioanalytical Services Department, WuXi AppTec (Shanghai) Co. Ltd, Shanghai, China
| | - Jianbo Diao
- Bioanalytical Services Department, WuXi AppTec (Shanghai) Co. Ltd, Shanghai, China
| |
Collapse
|
26
|
Yazdi ZF, Roshannezhad S, Sharif S, Abbaszadegan MR. Recent progress in prompt molecular detection of liquid biopsy using Cas enzymes: innovative approaches for cancer diagnosis and analysis. J Transl Med 2024; 22:1173. [PMID: 39741289 DOI: 10.1186/s12967-024-05908-y] [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: 08/10/2024] [Accepted: 11/20/2024] [Indexed: 01/02/2025] Open
Abstract
Creating fast, non-invasive, precise, and specific diagnostic tests is crucial for enhancing cancer treatment outcomes. Among diagnostic methods, those relying on nucleic acid detection are highly sensitive and specific. Recent developments in diagnostic technologies, particularly those leveraging Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR), are revolutionizing cancer detection, providing accurate and timely results. In clinical oncology, liquid biopsy has become a noninvasive and early-detectable alternative to traditional biopsies over the last two decades. Analyzing the nucleic acid content of liquid biopsy samples, which include Circulating Tumor Cells (CTCs), Circulating Tumor DNA (ctDNA), Circulating Cell-Free RNA (cfRNA), and tumor extracellular vesicles, provides a noninvasive method for cancer detection and monitoring. In this review, we explore how the characteristics of various Cas (CRISPR-associated) enzymes have been utilized in diagnostic assays for cancer liquid biopsy and highlight their main applications of innovative approaches in monitoring, as well as early and rapid detection of cancers.
Collapse
Affiliation(s)
- Zahra Farshchian Yazdi
- Department of Medical Genetics, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | - Samaneh Sharif
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Mashhad University of Medical Sciences, Azadi Square, Mashhad, Iran.
| | - Mohammad Reza Abbaszadegan
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Mashhad University of Medical Sciences, Azadi Square, Mashhad, Iran.
| |
Collapse
|
27
|
Ebrahimi F, Kumari A, Ghadami S, Al Abdullah S, Dellinger K. The Potential for Extracellular Vesicles in Nanomedicine: A Review of Recent Advancements and Challenges Ahead. Adv Biol (Weinh) 2024:e2400623. [PMID: 39739455 DOI: 10.1002/adbi.202400623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/02/2024] [Indexed: 01/02/2025]
Abstract
Extracellular vesicles (EVs) have emerged as promising tools in diagnostics and therapy for chronic diseases, including cancer and Alzheimer's. Small EVs, also called exosomes, are lipid-bound particles (≈30-150 nm) that play a role in healthy and pathophysiological interactions, including intercellular communication, by transporting bioactive molecules, including proteins, lipids, and nucleic acids. Their ability to cross biological barriers, such as the blood-brain barrier, makes them ideal candidates for targeted therapeutic interventions. In the context of chronic diseases, exosomes can be engineered to deliver active agents, including small molecules and siRNAs to specific target cells, providing a novel approach to precision medicine. Moreover, exosomes show great promise as repositories for diagnostic biomarkers. Their cargo can reflect the physiological and pathological status of the parent cells, making them valuable indicators of disease progression and response to treatment. This paper presents a comprehensive review of the application of exosomes in four chronic diseases: cancer, cardiovascular disease, neurodegenerative disease, and orthopedic disease, which significantly impact global public health due to their high prevalence and associated morbidity and mortality rates. Furthermore, the potential of exosomes as valuable tools for theranostics and disease management is highlighted. Finally, the challenges associated with exosomes and their demonstrated potential for advancing future nanomedicine applications are discussed.
Collapse
Affiliation(s)
- Farbod Ebrahimi
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering, North Carolina A&T State University, 2907 E Gate City Blvd, Greensboro, NC, 27401, USA
| | - Anjali Kumari
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering, North Carolina A&T State University, 2907 E Gate City Blvd, Greensboro, NC, 27401, USA
| | - Samaneh Ghadami
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering, North Carolina A&T State University, 2907 E Gate City Blvd, Greensboro, NC, 27401, USA
| | - Saqer Al Abdullah
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering, North Carolina A&T State University, 2907 E Gate City Blvd, Greensboro, NC, 27401, USA
| | - Kristen Dellinger
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering, North Carolina A&T State University, 2907 E Gate City Blvd, Greensboro, NC, 27401, USA
| |
Collapse
|
28
|
Hussung S, Hess ME, Haghighi EB, Wittel UA, Boerries M, Fritsch RM. Integrated Analysis of Cell-Free DNA and Novel Protein Biomarkers for Stratification and Therapy Monitoring in Stage IV Pancreatic Cancer: A Preliminary Study. Diagnostics (Basel) 2024; 15:49. [PMID: 39795577 PMCID: PMC11720586 DOI: 10.3390/diagnostics15010049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 12/25/2024] [Accepted: 12/26/2024] [Indexed: 01/13/2025] Open
Abstract
Background: Given the poor prognosis of metastatic pancreatic adenocarcinoma (mPDAC), closer disease monitoring through liquid biopsy, most frequently based on serial measurements of cell-free mutated KRAS (KRASmut cfDNA), has become a highly active research focus, aimed at improving patients' long-term outcomes. However, most of the available data show only a limited predictive and prognostic value of single-parameter-based methods. We hypothesized that a combined longitudinal analysis of KRASmut cfDNA and novel protein biomarkers could improve risk stratification and molecular monitoring of patients with mPDAC. Methods: We prospectively collected 160 plasma samples from 47 patients with mPDAC at our institution. Highly sensitive single-target ddPCR assays were employed to detect and quantify KRASmut cfDNA. Additionally, analysis of ten protein biomarkers was performed through Enzyme-linked Immunosorbent Assay (ELISA), and Carbohydrate-Antigen 19-9 (CA 19-9) dynamics were registered. Results: KRASmut cfDNA was detectable in 37/47 (78.7%) patients throughout the course of study, and CA 19-9 levels were elevated in 40 out of 47 (85.1%) patients. KRASmut cfDNA increase at the time of the first follow-up could predict inferior progression-free survival (PFS) (Hazard ratio (HR) = 3.40, p = 0.0003) and overall survival (OS) (HR = 4.91, p < 0.0001). In contrast to CA 19-9 kinetics, which were not predictive of outcome, integrated analysis of KRASmut cfDNA combined with six evaluated circulating protein biomarkers allowed basal risk stratification at the time of the first follow-up (HR = 10.2, p = 0.0014). Conclusions: A combined longitudinal analysis of KRASmut cfDNA with selected protein biomarkers offers significantly improved prognostic value for patients with mPDAC compared to single-parameter methods. This innovative approach is a step forward in the molecular monitoring of mPDAC and should be validated in further prospective studies.
Collapse
Affiliation(s)
- Saskia Hussung
- Department of Medicine I (Hematology, Oncology and Stem Cell Transplantation), Freiburg University Medical Center, 79106 Freiburg, Germany;
- Department of Medical Oncology and Hematology, Zurich University Hospital, 8091 Zurich, Switzerland
| | - Maria E. Hess
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (M.E.H.); (E.B.H.); (M.B.)
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Elham Bavafaye Haghighi
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (M.E.H.); (E.B.H.); (M.B.)
| | - Uwe A. Wittel
- Department of Surgery, Freiburg University Medical Center, 79106 Freiburg, Germany;
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (M.E.H.); (E.B.H.); (M.B.)
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Comprehensive Cancer Center Freiburg (CCCF), Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Ralph M. Fritsch
- Department of Medicine I (Hematology, Oncology and Stem Cell Transplantation), Freiburg University Medical Center, 79106 Freiburg, Germany;
- Department of Medical Oncology and Hematology, Zurich University Hospital, 8091 Zurich, Switzerland
- Department of Surgery, Freiburg University Medical Center, 79106 Freiburg, Germany;
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
29
|
Büdeyri I, Guckelberger O, Oppermann E, Roy D, Sliwinski S, Becker F, Struecker B, Vogl TJ, Pascher A, Bechstein WO, Lorentzen A, Heikenwalder M, Juratli MA. Ezrin Polarization as a Diagnostic Marker for Circulating Tumor Cells in Hepatocellular Carcinoma. Cells 2024; 14:6. [PMID: 39791707 PMCID: PMC11720075 DOI: 10.3390/cells14010006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 12/08/2024] [Accepted: 12/24/2024] [Indexed: 01/12/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is the sixth most common cancer and the third leading cause of cancer-related death worldwide, with no precise method for early detection. Circulating tumor cells (CTCs) expressing the dynamic polarity of the cytoskeletal membrane protein, ezrin, have been proposed to play a crucial role in tumor progression and metastasis. This study investigated the diagnostic and prognostic potential of polarized circulating tumor cells (p-CTCs) in HCC patients. CTCs were isolated from the peripheral blood of 20 HCC patients and 18 patients with nonmalignant liver disease (NMLD) via an OncoQuick® kit and immunostained with Ezrin-Alexa Fluor 488®, CD146-PE, and CD45-APC. A fluorescence microscopy was then performed for analysis. The HCC group exhibited significantly higher levels of p-CTCs, with median values of 0.56 p-CTCs/mL, compared to 0.02 p-CTCs/mL (p = 0.03) in the NMLD group. CTCs were detected in 95% of the HCC patients, with a sensitivity of 95% and specificity of 89%. p-CTCs were present in 75% of the HCC patients, with a sensitivity of 75% and a specificity of 94%. Higher p-CTC counts were associated with the significantly longer overall survival in HCC patients (p = 0.05). These findings suggest that p-CTCs could serve as valuable diagnostic and prognostic markers for HCC. The incorporation of p-CTCs into diagnostic strategies could enhance therapeutic decision-making and improve patient outcomes.
Collapse
Affiliation(s)
- Ibrahim Büdeyri
- Department of General, Visceral and Transplant Surgery, University Hospital Muenster, University of Muenster, Albert-Schweitzer-Campus 1, 48149 Muenster, Germany; (I.B.)
| | - Olaf Guckelberger
- Department of General, Visceral and Transplant Surgery, University Hospital Muenster, University of Muenster, Albert-Schweitzer-Campus 1, 48149 Muenster, Germany; (I.B.)
| | - Elsie Oppermann
- Department of General, Visceral and Transplant Surgery, Frankfurt University Hospital, 60596 Frankfurt, Germany
| | - Dhruvajyoti Roy
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Svenja Sliwinski
- Department of General, Visceral and Transplant Surgery, Frankfurt University Hospital, 60596 Frankfurt, Germany
| | - Felix Becker
- Department of General, Visceral and Transplant Surgery, University Hospital Muenster, University of Muenster, Albert-Schweitzer-Campus 1, 48149 Muenster, Germany; (I.B.)
| | - Benjamin Struecker
- Department of General, Visceral and Transplant Surgery, University Hospital Muenster, University of Muenster, Albert-Schweitzer-Campus 1, 48149 Muenster, Germany; (I.B.)
| | - Thomas J. Vogl
- Department of Diagnostic and Interventional Radiology, Frankfurt University Hospital, Goethe University, 60596 Frankfurt, Germany
| | - Andreas Pascher
- Department of General, Visceral and Transplant Surgery, University Hospital Muenster, University of Muenster, Albert-Schweitzer-Campus 1, 48149 Muenster, Germany; (I.B.)
| | - Wolf O. Bechstein
- Department of General, Visceral and Transplant Surgery, Frankfurt University Hospital, 60596 Frankfurt, Germany
| | - Anna Lorentzen
- Department of Biomedicine, Aarhus University, 8200 Aarhus, Denmark
| | - Mathias Heikenwalder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Mazen A. Juratli
- Department of General, Visceral and Transplant Surgery, University Hospital Muenster, University of Muenster, Albert-Schweitzer-Campus 1, 48149 Muenster, Germany; (I.B.)
| |
Collapse
|
30
|
Thusgaard CF, Sadegh S, Jochumsen KM, Kruse TA, Thomassen M. A Sensitive and Transparent Method for Tumor-Informed Detection of Circulating Tumor DNA in Ovarian Cancer Using Whole-Genome Sequencing. Int J Mol Sci 2024; 25:13349. [PMID: 39769113 PMCID: PMC11678811 DOI: 10.3390/ijms252413349] [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: 10/14/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Circulating tumor DNA (ctDNA) is a biomarker that could potentially improve the survival rate of ovarian cancer (OC), e.g., by monitoring treatment response and early relapse detection. However, an optimal method for ctDNA analysis in OC remains to be established. We developed a method for tumor-informed single-nucleotide variant detection of ctDNA in OC using whole-genome sequencing. Tumor and plasma samples obtained at the time of diagnosis from 10 patients with OC were included. The tested method involved applying basic filters with different cut-offs of read depth, allelic depth, and variant allele frequency of tumor and normal DNA. In addition, we applied a new filtering approach using plasma samples from the other included OC patients (the plasma pool) for specific removal of artefacts. The basic filters with varying cut-offs showed minor improvement in signal-to-noise ratio (S2N). However, the addition of the plasma pool filter resulted in a considerable ctDNA signal improvement, indicated by both S2N and z-score. This study demonstrates a promising method for ctDNA detection in OC patients using a tumor-informed approach for whole-genome sequencing. Despite the limited number of patients involved, the results suggest a significant potential of the method for ctDNA signal detection in patients with OC.
Collapse
Affiliation(s)
- Christine Fribert Thusgaard
- Department of Gynecology and Obstetrics, Odense University Hospital, 5000 Odense, Denmark;
- Research Unit of Gynecology and Obstetrics, Department of Clinical Research, University of Southern Denmark, Odense University Hospital, 5000 Odense, Denmark
| | - Sepideh Sadegh
- Department of Clinical Genetics, Odense University Hospital, 5000 Odense, Denmark; (S.S.); (T.A.K.); (M.T.)
- Clinical Genome Center, Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
| | - Kirsten Marie Jochumsen
- Department of Gynecology and Obstetrics, Odense University Hospital, 5000 Odense, Denmark;
- Research Unit of Gynecology and Obstetrics, Department of Clinical Research, University of Southern Denmark, Odense University Hospital, 5000 Odense, Denmark
| | - Torben Arvid Kruse
- Department of Clinical Genetics, Odense University Hospital, 5000 Odense, Denmark; (S.S.); (T.A.K.); (M.T.)
- Clinical Genome Center, Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
| | - Mads Thomassen
- Department of Clinical Genetics, Odense University Hospital, 5000 Odense, Denmark; (S.S.); (T.A.K.); (M.T.)
- Clinical Genome Center, Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
| |
Collapse
|
31
|
Kepsha MA, Timofeeva AV, Chernyshev VS, Silachev DN, Mezhevitinova EA, Sukhikh GT. MicroRNA-Based Liquid Biopsy for Cervical Cancer Diagnostics and Treatment Monitoring. Int J Mol Sci 2024; 25:13271. [PMID: 39769036 PMCID: PMC11678179 DOI: 10.3390/ijms252413271] [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: 10/31/2024] [Revised: 12/03/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Despite prevention strategies, cervical cancer remains a significant public health issue. Human papillomavirus plays a critical role in its development, and early detection is vital to improve patient outcomes. The incidence of cervical cancer is projected to rise, necessitating better diagnostic tools. Traditional screening methods like the cytological examination and human papillomavirus testing have limitations in sensitivity and reproducibility. Liquid-based cytology offers some improvements, but the need for more reliable and sensitive techniques persists, particularly for detecting precancerous lesions. Liquid biopsy is a non-invasive method that analyzes cancer-derived products in biofluids like blood, offering potential for real-time monitoring of tumor progression, metastasis, and treatment response. It can be based on detection of circulating tumor cells (CTCs), circulating free DNA (cfDNA), and microRNAs (miRNAs). This review particularly underlines the potential of microRNAs, which are transported by extracellular vesicles. Overall, this article underscores the importance of continued research into non-invasive diagnostic methods like liquid biopsy to enhance cervical cancer screening and treatment monitoring.
Collapse
Affiliation(s)
| | | | - Vasiliy S. Chernyshev
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov, Ministry of Healthcare of the Russian Federation, Moscow 117997, Russia (D.N.S.)
| | | | | | | |
Collapse
|
32
|
Li R, Qian J, Zhu X, Tao T, Zhou X. Nanomolecular machines: Pioneering precision medicine for neoplastic diseases through advanced diagnosis and treatment. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167486. [PMID: 39218275 DOI: 10.1016/j.bbadis.2024.167486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/16/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Tumors pose a major threat to human health, accounting for nearly one-sixth of global deaths annually. The primary treatments include surgery, radiotherapy, chemotherapy, and immunotherapy, each associated with significant side effects. This has driven the search for new therapies with fewer side effects and greater specificity. Nanotechnology has emerged as a promising field in this regard, particularly nanomolecular machines at the nanoscale. Nanomolecular machines are typically constructed from biological macromolecules like proteins, DNA, and RNA. These machines can be programmed to perform specialized tasks with precise instructions. Recent research highlights their potential in tumor diagnostics-identifying susceptibility genes, detecting viruses, and pinpointing tumor markers. Nanomolecular machines also offer advancements in tumor therapy. They can reduce traditional treatment side effects by delivering chemotherapy drugs and enhancing immunotherapy, and they support innovative treatments like sonodynamic and phototherapy. Additionally, they can starve tumors by blocking blood vessels, and eliminate tumors by disrupting cell membranes or lysosomes. This review categorizes and explains the latest achievements in molecular machine research, explores their models, and practical clinical uses in tumor diagnosis and treatment. It aims to broaden the research perspective and accelerate the clinical adoption of these technologies.
Collapse
Affiliation(s)
- Ruming Li
- Department of Immunology, School of Medicine, Nantong University, Nantong, China; The Second Affiliated Hospital, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, China
| | - Jialu Qian
- Department of Immunology, School of Medicine, Nantong University, Nantong, China
| | - Xiao Zhu
- The Second Affiliated Hospital, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, China
| | - Tao Tao
- Department of Gastroenterology, Zibo Central Hospital, Zibo, China.
| | - Xiaorong Zhou
- Department of Immunology, School of Medicine, Nantong University, Nantong, China.
| |
Collapse
|
33
|
Kurma K, Eslami-S Z, Alix-Panabières C, Cayrefourcq L. Liquid biopsy: paving a new avenue for cancer research. Cell Adh Migr 2024; 18:1-26. [PMID: 39219215 PMCID: PMC11370957 DOI: 10.1080/19336918.2024.2395807] [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: 04/04/2024] [Revised: 05/21/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024] Open
Abstract
The current constraints associated with cancer diagnosis and molecular profiling, which rely on invasive tissue biopsies or clinical imaging, have spurred the emergence of the liquid biopsy field. Liquid biopsy involves the extraction of circulating tumor cells (CTCs), circulating free or circulating tumor DNA (cfDNA or ctDNA), circulating cell-free RNA (cfRNA), extracellular vesicles (EVs), and tumor-educated platelets (TEPs) from bodily fluid samples. Subsequently, these components undergo molecular characterization to identify biomarkers that are critical for early cancer detection, prognosis, therapeutic assessment, and post-treatment monitoring. These innovative biosources exhibit characteristics analogous to those of the primary tumor from which they originate or interact. This review comprehensively explores the diverse technologies and methodologies employed for processing these biosources, along with their principal clinical applications.
Collapse
Affiliation(s)
- Keerthi Kurma
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, Montpellier, France
- CREEC/CANECEV, MIVEGEC (CREES),
University of Montpellier, CNRS, IRD, Montpellier, France
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| | - Zahra Eslami-S
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, Montpellier, France
- CREEC/CANECEV, MIVEGEC (CREES),
University of Montpellier, CNRS, IRD, Montpellier, France
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, Montpellier, France
- CREEC/CANECEV, MIVEGEC (CREES),
University of Montpellier, CNRS, IRD, Montpellier, France
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| | - Laure Cayrefourcq
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, Montpellier, France
- CREEC/CANECEV, MIVEGEC (CREES),
University of Montpellier, CNRS, IRD, Montpellier, France
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| |
Collapse
|
34
|
Li M. Atomic force microscopy as a nanomechanical tool for cancer liquid biopsy. Biochem Biophys Res Commun 2024; 734:150637. [PMID: 39226737 DOI: 10.1016/j.bbrc.2024.150637] [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: 07/25/2024] [Revised: 08/24/2024] [Accepted: 08/30/2024] [Indexed: 09/05/2024]
Abstract
Liquid biopsies have been receiving tremendous attention for their potential to reshape cancer management. Though current studies of cancer liquid biopsy primarily focus on applying biochemical assays to characterize the genetic/molecular profiles of circulating tumor cells (CTCs) and their secondary products shed from tumor sites in bodily fluids, delineating the nanomechanical properties of tumor-associated materials in liquid biopsy specimens yields complementary insights into the biology of tumor dissemination and evolution. Particularly, atomic force microscopy (AFM) has become a standard and versatile toolbox for characterizing the mechanical properties of living biological systems at the micro/nanoscale, and AFM has been increasingly utilized to probe the nanomechanical properties of various tumor-derived analytes in liquid biopsies, including CTCs, tumor-associated cells, circulating tumor DNA (ctDNA) molecules, and extracellular vesicles (EVs), offering additional possibilities for understanding cancer pathogenesis from the perspective of mechanobiology. Herein, the applications of AFM in cancer liquid biopsy are summarized, and the challenges and future directions of AFM as a nanomechanical analysis tool in cancer liquid biopsy towards clinical utility are discussed and envisioned.
Collapse
Affiliation(s)
- Mi Li
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, 110016, China.
| |
Collapse
|
35
|
Pollard CA, Saito ER, Burns JM, Hill JT, Jenkins TG. Considering Biomarkers of Neurodegeneration in Alzheimer's Disease: The Potential of Circulating Cell-Free DNA in Precision Neurology. J Pers Med 2024; 14:1104. [PMID: 39590596 PMCID: PMC11595805 DOI: 10.3390/jpm14111104] [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: 10/10/2024] [Revised: 10/30/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
Neurodegenerative diseases, such as Alzheimer's disease (AD), are a growing public health crisis, exacerbated by an aging global population and the lack of effective early disease-modifying therapies. Early detection of neurodegenerative disorders is critical to delaying symptom onset and mitigating disease progression, but current diagnostic tools often rely on detecting pathology once clinical symptoms have emerged and significant neuronal damage has already occurred. While disease-specific biomarkers, such as amyloid-beta and tau in AD, offer precise insights, they are too limited in scope for broader neurodegeneration screening for these conditions. Conversely, general biomarkers like neurofilament light chain (NfL) provide valuable staging information but lack targeted insights. Circulating cell-free DNA (cfDNA), released during cell death, is emerging as a promising biomarker for early detection. Derived from dying cells, cfDNA can capture both general neurodegenerative signals and disease-specific insights, offering multi-layered genomic and epigenomic information. Though its clinical potential remains under investigation, advances in cfDNA detection sensitivity, standardized protocols, and reference ranges could establish cfDNA as a valuable tool for early screening. cfDNA methylation signatures, in particular, show great promise for identifying tissue-of-origin and disease-specific changes, offering a minimally invasive biomarker that could transform precision neurology. However, further research is required to address technological challenges and validate cfDNA's utility in clinical settings. Here, we review recent work assessing cfDNA as a potential early biomarker in AD. With continued advances, cfDNA could play a pivotal role in shifting care from reactive to proactive, improving diagnostic timelines and patient outcomes.
Collapse
Affiliation(s)
- Chad A. Pollard
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA
- Resonant, Heber, UT 84032, USA
| | | | - Jeffrey M. Burns
- University of Kansas Alzheimer’s Disease Research Center, Fairway, KS 66205, USA
| | - Jonathon T. Hill
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA
| | - Timothy G. Jenkins
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA
- Resonant, Heber, UT 84032, USA
| |
Collapse
|
36
|
Luo J, Zhang C, Wu M, Yao X, Duan Y, Li Y. Excitation/emission-enhanced heterostructure photonic crystal array synergizing with "DD-A" FRET entropy-driven circuit for high-resolution and ultrasensitive analysis of ctDNA. Biosens Bioelectron 2024; 263:116615. [PMID: 39106690 DOI: 10.1016/j.bios.2024.116615] [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: 07/21/2024] [Accepted: 07/29/2024] [Indexed: 08/09/2024]
Abstract
Circulating tumor DNA (ctDNA) is an emerging biomarker of liquid biopsy for cancer. But it remains a challenge to achieve simple, sensitive and specific detection of ctDNA because of low abundance and single-base mutation. In this work, an excitation/emission-enhanced heterostructure photonic crystal (PC) array synergizing with entropy-driven circuit (EDC) was developed for high-resolution and ultrasensitive analysis of ctDNA. The donor donor-acceptor FÖrster resonance energy transfer ("DD-A" FRET) was integrated in EDC based on the introduction of simple auxiliary strand, which exhibited higher sensitivity than that of traditional EDC. The heterostructure PC array was constructed with the bilayer periodic nanostructures of nanospheres. Because the heterostructure PC has the adjustable dual photonic band gaps (PBGs) by changing nanosphere sizes, and the "DD-A" FRET can offer the excitation and emission peak with enough distance, it helps the successful matches between the dual PBGs of heterostructure PC and the excitation/emission peaks of "DD-A" FRET; thus, the fluorescence from EDC can be enhanced effectively from both of excitation and emission processes on heterostructure PC array. Besides, high-resolution of single-base mutation was obtained through the strict recognition of EDC. Benefiting from the specific spectrum-matched and synergetic amplification of heterostructure PC and EDC with "DD-A" FRET, the proposed array obtained ultrasensitive detection of ctDNA with LOD of 12.9 fM, and achieved the analysis of mutation frequency as low as 0.01%. Therefore, the proposed strategy has the advantages of simple operation, mild conditions (enzyme-free and isothermal), high-sensitivity, high-resolution and high-throughput analysis, showing potential in bioassay and clinical application.
Collapse
Affiliation(s)
- Jie Luo
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Chuyan Zhang
- Precision Medicine Translational Research Center, Medical Equipment Innovation Research Center, Med-X Center for Manufacturing, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Mengfan Wu
- Research Center of Analytical Instrumentation, School of Mechanical Engineering, Sichuan University, Chengdu, Sichuan, 610065, PR China
| | - Xiuyuan Yao
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Yixiang Duan
- Research Center of Analytical Instrumentation, School of Mechanical Engineering, Sichuan University, Chengdu, Sichuan, 610065, PR China
| | - Yongxin Li
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China.
| |
Collapse
|
37
|
Li JY, Zuo LP, Xu J, Sun CY. Clinical applications of circulating tumor DNA in hematological malignancies: From past to the future. Blood Rev 2024; 68:101237. [PMID: 39261219 DOI: 10.1016/j.blre.2024.101237] [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: 05/27/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/13/2024]
Abstract
Liquid biopsy, particularly circulating tumor DNA (ctDNA), has drawn a lot of attention as a non- or minimal-invasive detection approach for clinical applications in patients with cancer. Many hematological malignancies are well suited for serial and repeated ctDNA surveillance due to relatively high ctDNA concentrations and high loads of tumor-specific genetic and epigenetic abnormalities. Progress of detecting technology in recent years has improved sensitivity and specificity significantly, thus broadening and strengthening the potential utilities of ctDNA including early diagnosis, prognosis estimation, treatment response evaluation, minimal residual disease monitoring, targeted therapy selection, and immunotherapy surveillance. This manuscript reviews the detection methodologies, clinical application and future challenges of ctDNA in hematological malignancies, especially for lymphomas, myeloma and leukemias.
Collapse
Affiliation(s)
- Jun-Ying Li
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of science and Technology, Wuhan, Hubei, China
| | - Li-Ping Zuo
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of science and Technology, Wuhan, Hubei, China
| | - Jian Xu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of science and Technology, Wuhan, Hubei, China
| | - Chun-Yan Sun
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
38
|
Kramer A, Rubio‐Alarcón C, van den Broek D, Vessies DCL, van't Erve I, Meijer GA, Vink GR, Schuuring E, Fijneman RJA, Coupé VMH, Retèl VP. A scenario-drafting study to explore potential future implementation pathways of circulating tumor DNA testing in oncology. Mol Oncol 2024; 18:2730-2742. [PMID: 38060377 PMCID: PMC11547223 DOI: 10.1002/1878-0261.13562] [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: 07/07/2023] [Revised: 11/17/2023] [Accepted: 12/05/2023] [Indexed: 01/13/2024] Open
Abstract
Circulating tumor DNA (ctDNA) detection has multiple promising applications in oncology, but the road toward implementation in clinical practice is unclear. We aimed to support the implementation process by exploring potential future pathways of ctDNA testing. To do so, we studied four ctDNA-testing applications in two cancer types and elicited opinions from 30 ctDNA experts in the Netherlands. Our results showed that the current available evidence differed per application and cancer type. Tumor profiling and monitoring treatment response were found most likely to be implemented in non-small cell lung cancer (NSCLC) within 5 years. For colorectal cancer, applications of ctDNA testing were found to be at an early stage in the implementation process. Demonstrating clinical utility was found a key aspect for successful implementation, but there was no consensus regarding the evidence requirements. The next step toward implementation is to define how clinical utility of biomarkers should be evaluated. Finally, these data indicate that specific challenges for each clinical application and tumor type should be appropriately addressed in a deliberative process involving all stakeholders to ensure implementation of ctDNA testing and timely access for patients.
Collapse
Affiliation(s)
- Astrid Kramer
- Department of Epidemiology and Data ScienceAmsterdam UMCThe Netherlands
| | | | - Daan van den Broek
- Department of Laboratory MedicineNetherlands Cancer InstituteAmsterdamThe Netherlands
| | - Daan C. L. Vessies
- Department of Laboratory MedicineNetherlands Cancer InstituteAmsterdamThe Netherlands
| | - Iris van't Erve
- Department of PathologyNetherlands Cancer InstituteAmsterdamThe Netherlands
| | - Gerrit A. Meijer
- Department of PathologyNetherlands Cancer InstituteAmsterdamThe Netherlands
| | - Geraldine R. Vink
- Department of Medical Oncology, University Medical Center UtrechtUniversity of UtrechtThe Netherlands
- Department of Research and DevelopmentIKNLUtrechtThe Netherlands
| | - Ed Schuuring
- Department of Pathology and Medical BiologyUniversity Medical Center GroningenThe Netherlands
| | | | | | - Valesca P. Retèl
- Department of Psychosocial Research and EpidemiologyNetherlands Cancer InstituteAmsterdamThe Netherlands
- Erasmus School of Health Policy and ManagementErasmus University RotterdamThe Netherlands
| |
Collapse
|
39
|
Uotani K, Fujiwara T, Ueda K, Yoshida A, Iwata S, Morita T, Kiyono M, Kunisada T, Takeda K, Hasei J, Yoshioka Y, Ochiya T, Ozaki T. Identification of ENO-1 positive extracellular vesicles as a circulating biomarker for monitoring of Ewing sarcoma. Cancer Sci 2024; 115:3660-3671. [PMID: 39307979 PMCID: PMC11531948 DOI: 10.1111/cas.16343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 08/20/2024] [Accepted: 08/31/2024] [Indexed: 11/05/2024] Open
Abstract
The lack of circulating biomarkers for tumor monitoring is a major problem in Ewing sarcoma management. The development of methods for accurate tumor monitoring is required, considering the high recurrence rate of drug-resistant Ewing sarcoma. Here, we describe a sensitive analytical technique for tumor monitoring of Ewing sarcoma by detecting circulating extracellular vesicles secreted from Ewing sarcoma cells. Proteomic analysis of Ewing sarcoma cell-derived extracellular vesicles identified 564 proteins prominently observed in extracellular vesicles from three Ewing sarcoma cell lines. Among these, CD99, SLC1A5, and ENO-1 were identified on extracellular vesicles purified from sera of patients with Ewing sarcoma before treatment but not on extracellular vesicles from those after treatment and healthy individuals. Notably, not only Ewing sarcoma-derived extracellular vesicles but also Ewing sarcoma cells demonstrated proteomic expression of CD99 and ENO-1 on their surface membranes. ENO-1+CD63+ extracellular vesicle detection was reduced after tumor resection while both CD99+CD63+ and ENO-1+CD63+ extracellular vesicles were detected in serum from Ewing sarcoma-bearing mice. Finally, the accuracy of liquid biopsy targeting these candidates was assessed using extracellular vesicles from the sera of patients with Ewing sarcoma. Elevated ENO-1+CD81+ extracellular vesicles in the serum of patients before treatments distinguished patients with Ewing sarcoma from healthy individuals with an area under the curve value of 0.92 (P < 0.001) and reflected the tumor burden in patients with Ewing sarcoma during multidisciplinary treatments. Collectively, circulating ENO-1+CD81+ extracellular vesicle detection could represent a novel tool for tumor monitoring of Ewing sarcoma.
Collapse
Affiliation(s)
- Koji Uotani
- Department of Orthopedic SurgeryOkayama University Graduate School of Medicine, Dentistry, and Pharmaceutical SciencesOkayamaJapan
| | - Tomohiro Fujiwara
- Department of Orthopedic SurgeryOkayama University Graduate School of Medicine, Dentistry, and Pharmaceutical SciencesOkayamaJapan
- Center of Innovative MedicineOkayama University HospitalOkayamaJapan
| | - Koji Ueda
- Cancer Precision Medicine CenterJapanese Foundation for Cancer ResearchTokyoJapan
| | - Aki Yoshida
- Department of Orthopedic SurgeryOkayama University Graduate School of Medicine, Dentistry, and Pharmaceutical SciencesOkayamaJapan
| | - Shintaro Iwata
- Department of Musculoskeletal OncologyNational Cancer Center HospitalTokyoJapan
| | - Takuya Morita
- Department of Orthopedic SurgeryOkayama University Graduate School of Medicine, Dentistry, and Pharmaceutical SciencesOkayamaJapan
| | - Masahiro Kiyono
- Department of Orthopedic SurgeryOkayama University Graduate School of Medicine, Dentistry, and Pharmaceutical SciencesOkayamaJapan
| | - Toshiyuki Kunisada
- Department of Medical Materials for Musculoskeletal ReconstructionOkayama University Graduate School of Medicine, Dentistry, and Pharmaceutical SciencesOkayamaJapan
| | - Ken Takeda
- Department of Intelligent Orthopedic SystemOkayama University Graduate School of Medicine, Dentistry, and Pharmaceutical SciencesOkayamaJapan
| | - Joe Hasei
- Department of Orthopedic SurgeryOkayama University Graduate School of Medicine, Dentistry, and Pharmaceutical SciencesOkayamaJapan
| | - Yusuke Yoshioka
- Department of Molecular and Cellular MedicineInstitute of Medical Science, Tokyo Medical UniversityTokyoJapan
| | - Takahiro Ochiya
- Department of Molecular and Cellular MedicineInstitute of Medical Science, Tokyo Medical UniversityTokyoJapan
| | - Toshifumi Ozaki
- Department of Orthopedic SurgeryOkayama University Graduate School of Medicine, Dentistry, and Pharmaceutical SciencesOkayamaJapan
| |
Collapse
|
40
|
Alexander EM, Miller HA, Egger ME, Smith ML, Yaddanapudi K, Linder MW. The Correlation between Plasma Circulating Tumor DNA and Radiographic Tumor Burden. J Mol Diagn 2024; 26:952-961. [PMID: 39181324 PMCID: PMC11524323 DOI: 10.1016/j.jmoldx.2024.07.001] [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: 01/23/2024] [Revised: 06/05/2024] [Accepted: 07/15/2024] [Indexed: 08/27/2024] Open
Abstract
Conventional blood-based biomarkers and radiographic imaging are excellent for use in monitoring different aspects of malignant disease, but given their specific shortcomings, their integration with other, complementary markers such as plasma circulating tumor DNA (ctDNA) will be beneficial toward a precision medicine-driven future. Plasma ctDNA analysis utilizes the measurement of cancer-specific molecular alterations in a variety of bodily fluids released by dying tumor cells to monitor and profile response to therapy, and is being employed in several clinical scenarios. Plasma concentrations of ctDNA have been reported to correlate with tumor burden. However, the strength of this association is generally poor and highly variable, confounding the interpretation of longitudinal plasma ctDNA measurements in conjunction with routine radiographic assessments. Herein is discussed what is currently understood with respect to the fundamental characteristics of tumor growth that dictate plasma ctDNA concentrations, with a perspective on its interpretation in conjunction with radiographically determined tumor burden assessments.
Collapse
Affiliation(s)
- Evan M Alexander
- Department of Pathology and Laboratory Medicine, University of Louisville, Louisville, Kentucky
| | - Hunter A Miller
- Department of Pathology and Laboratory Medicine, University of Louisville, Louisville, Kentucky
| | - Michael E Egger
- Hiram C. Polk, Jr, MD, Department of Surgery, University of Louisville, Louisville, Kentucky; UofL Health-Brown Cancer Center, University of Louisville, Louisville, Kentucky
| | - Melissa L Smith
- UofL Health-Brown Cancer Center, University of Louisville, Louisville, Kentucky; Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, Kentucky; Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky
| | - Kavitha Yaddanapudi
- UofL Health-Brown Cancer Center, University of Louisville, Louisville, Kentucky; Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky; Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Mark W Linder
- Department of Pathology and Laboratory Medicine, University of Louisville, Louisville, Kentucky; UofL Health-Brown Cancer Center, University of Louisville, Louisville, Kentucky.
| |
Collapse
|
41
|
Wang W, Hu Y, Fu F, Ren W, Wang T, Wang S, Li Y. Advancement in Multi-omics approaches for Uterine Sarcoma. Biomark Res 2024; 12:129. [PMID: 39472980 PMCID: PMC11523907 DOI: 10.1186/s40364-024-00673-y] [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: 06/19/2024] [Accepted: 10/14/2024] [Indexed: 11/02/2024] Open
Abstract
Uterine sarcoma (US) is a rare malignant tumor that has various pathological types and high heterogeneity in the female reproductive system. Its subtle early symptoms, frequent recurrence, and resistance to radiation and chemotherapy make the prognosis for US patients very poor. Therefore, understanding the molecular mechanisms underlying tumorigenesis and progression is essential for an accurate diagnosis and targeted therapy to improve patient outcomes. Recent advancements in high-throughput molecular sequencing have allowed for a deeper understanding of diseases through multi-omics technologies. In this review, the latest progress and future potential of multi-omics technologies in US research is examined, and their roles in biomarker discovery and their application in the precise diagnosis and treatment of US are highlighted.
Collapse
Affiliation(s)
- Wuyang Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Anv. Wuhan, Wuhan, Hubei, 430030, P.R. China
| | - Yu Hu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Anv. Wuhan, Wuhan, Hubei, 430030, P.R. China
| | - Fangfang Fu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Anv. Wuhan, Wuhan, Hubei, 430030, P.R. China
| | - Wu Ren
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Anv. Wuhan, Wuhan, Hubei, 430030, P.R. China
| | - Tian Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Anv. Wuhan, Wuhan, Hubei, 430030, P.R. China.
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Anv. Wuhan, Wuhan, Hubei, 430030, P.R. China.
| | - Yan Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Anv. Wuhan, Wuhan, Hubei, 430030, P.R. China.
| |
Collapse
|
42
|
Duo Y, Han L, Yang Y, Wang Z, Wang L, Chen J, Xiang Z, Yoon J, Luo G, Tang BZ. Aggregation-Induced Emission Luminogen: Role in Biopsy for Precision Medicine. Chem Rev 2024; 124:11242-11347. [PMID: 39380213 PMCID: PMC11503637 DOI: 10.1021/acs.chemrev.4c00244] [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: 04/03/2024] [Revised: 09/11/2024] [Accepted: 09/17/2024] [Indexed: 10/10/2024]
Abstract
Biopsy, including tissue and liquid biopsy, offers comprehensive and real-time physiological and pathological information for disease detection, diagnosis, and monitoring. Fluorescent probes are frequently selected to obtain adequate information on pathological processes in a rapid and minimally invasive manner based on their advantages for biopsy. However, conventional fluorescent probes have been found to show aggregation-caused quenching (ACQ) properties, impeding greater progresses in this area. Since the discovery of aggregation-induced emission luminogen (AIEgen) have promoted rapid advancements in molecular bionanomaterials owing to their unique properties, including high quantum yield (QY) and signal-to-noise ratio (SNR), etc. This review seeks to present the latest advances in AIEgen-based biofluorescent probes for biopsy in real or artificial samples, and also the key properties of these AIE probes. This review is divided into: (i) tissue biopsy based on smart AIEgens, (ii) blood sample biopsy based on smart AIEgens, (iii) urine sample biopsy based on smart AIEgens, (iv) saliva sample biopsy based on smart AIEgens, (v) biopsy of other liquid samples based on smart AIEgens, and (vi) perspectives and conclusion. This review could provide additional guidance to motivate interest and bolster more innovative ideas for further exploring the applications of various smart AIEgens in precision medicine.
Collapse
Affiliation(s)
- Yanhong Duo
- Department
of Radiation Oncology, Shenzhen People’s Hospital, The Second
Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong China
- Wyss
Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02138, United States
| | - Lei Han
- College of
Chemistry and Pharmaceutical Sciences, Qingdao
Agricultural University, 700 Changcheng Road, Qingdao 266109, Shandong China
| | - Yaoqiang Yang
- Department
of Radiation Oncology, Shenzhen People’s Hospital, The Second
Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong China
| | - Zhifeng Wang
- Department
of Urology, Henan Provincial People’s Hospital, Zhengzhou University
People’s Hospital, Henan University
People’s Hospital, Zhengzhou, 450003, China
| | - Lirong Wang
- State
Key Laboratory of Luminescent Materials and Devices, South China University of Technology, Guangzhou 510640, China
| | - Jingyi Chen
- Wyss
Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02138, United States
| | - Zhongyuan Xiang
- Department
of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha 410000, Hunan, China
| | - Juyoung Yoon
- Department
of Chemistry and Nanoscience, Ewha Womans
University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Korea
| | - Guanghong Luo
- Department
of Radiation Oncology, Shenzhen People’s Hospital, The Second
Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong China
| | - Ben Zhong Tang
- School
of Science and Engineering, Shenzhen Institute of Aggregate Science
and Technology, The Chinese University of
Hong Kong, Shenzhen 518172, Guangdong China
| |
Collapse
|
43
|
Ferreira M, Morais M, Medeiros R, Teixeira AL. MicroRNAs as Promising Therapeutic Agents Against Prostate Cancer Resistant to Castration-Where Are We Now? Pharmaceutics 2024; 16:1347. [PMID: 39598472 PMCID: PMC11597238 DOI: 10.3390/pharmaceutics16111347] [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: 09/02/2024] [Revised: 10/17/2024] [Accepted: 10/19/2024] [Indexed: 11/29/2024] Open
Abstract
MicroRNAs are a conserved class of small, tissue-specific, non-coding RNAs that regulate gene expression to preserve cellular homeostasis. Proper miRNA expression is crucial for physiological balance because it affects numerous genetic pathways, including cell cycle control, proliferation, and apoptosis, through gene expression targeting. Deregulated miRNA expression has been implicated in several cancer types, including prostate cancer (PC), acting as tumor suppressors or oncogenes. Despite the availability of promising therapies to control tumor growth and progression, effective diagnostic and therapeutic strategies for different types of cancer are still lacking. PC continues to be a significant health challenge, particularly its castration-resistant (CRPC) form, which presents major therapeutic obstacles because of its resistance to conventional androgen deprivation treatments. This review explores miRNAs' critical roles in gene regulation and cancer biology, as well as various miRNA delivery systems, highlighting their potential and the challenges in effectively targeting cancer cells. It aims to provide a comprehensive overview of the status of miRNA research in the fight against CRPC, summarizing miRNA-based therapies' successes and limitations. It also highlights the promise of miRNAs as therapeutic agents for CRPC, underlining the need for further research to overcome existing challenges and move these therapies toward clinical applications.
Collapse
Affiliation(s)
- Mariana Ferreira
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal; (M.F.); (M.M.); (R.M.)
- ICBAS, Abel Salazar Institute for the Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal
| | - Mariana Morais
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal; (M.F.); (M.M.); (R.M.)
- ICBAS, Abel Salazar Institute for the Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal; (M.F.); (M.M.); (R.M.)
- ICBAS, Abel Salazar Institute for the Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal
- Biomedical Research Center (CEBIMED), Faculty of Health Sciences, Fernando Pessoa University (UFP), 4249-004 Porto, Portugal
- Research Department, LPCC-Portuguese League Against Cancer (NRNorte), 4200-172 Porto, Portugal
- Faculty of Medicine (FMUP), University of Porto, 4200-319 Porto, Portugal
| | - Ana Luísa Teixeira
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal; (M.F.); (M.M.); (R.M.)
| |
Collapse
|
44
|
Cossu G, Ramsay DSC, Daniel RT, El Cadhi A, Kerherve L, Morlaix E, Houidi SA, Millot-Piccoli C, Chapon R, Le Van T, Cao C, Farah W, Lleu M, Baland O, Beaurain J, Petit JM, Lemogne B, Messerer M, Berhouma M. Update on Neoadjuvant and Adjuvant BRAF Inhibitors in Papillary Craniopharyngioma: A Systematic Review. Cancers (Basel) 2024; 16:3479. [PMID: 39456573 PMCID: PMC11506763 DOI: 10.3390/cancers16203479] [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: 08/31/2024] [Revised: 09/26/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: The recent discovery of BRAF mutation in papillary craniopharyngiomas opened new avenues for targeted therapies to control tumour growth, decreasing the need for invasive treatments and relative complications. The aim of this systematic review was to summarize the recent scientific data dealing with the use of targeted therapies in papillary craniopharyngiomas, as adjuvant and neoadjuvant treatments. Methods: The PRISMA guidelines were followed with searches performed in Scopus, MEDLINE, and Embase, following a dedicated PICO approach. Results: We included 21 pertinent studies encompassing 53 patients: 26 patients received BRAF inhibitors (BRAFi) as adjuvant treatment, while 25 received them as neoadjuvant treatment. In the adjuvant setting, BRAFi were used to treat recurrent tumours after surgery or adjuvant radiation therapy. The most common regimen combined dabrafenib (BRAFi) with trametinib (MEK1 and 2 inhibitor) in 81% of cases. The mean treatment length was 8.8 months (range 1.6 to 28 months) and 32% were continuing BRAFi. A reduction of tumour volume variable from 24% to 100% was observed at cerebral MRI during treatment and volumetric reduction ≥80% was described in 64% of cases. Once the treatment was stopped, adjuvant treatments were performed to stabilize patients in remission in 11 cases (65%) or when a progression was detected in three cases (12%). In four cases no further therapies were administered (16%). Mean follow-up after the end of targeted therapy was 17.1 months. As neoadjuvant regimen, 36% of patients were treated with dabrafenib and trametinib with a near complete radiological response in all the cases with a mean treatment of 5.7 months. The neoadjuvant use of verumafenib (BRAFi) and cometinib (MEK1 inhibitor) induced a near complete response in 15 patients (94%), with a median volumetric reduction between 85% and 91%. Ten patients did not receive further treatments. Side effects varied among studies. The optimal timing, sequencing, and duration of treatment of these new therapies should be established. Moreover, questions remain about the choice of specific BRAF/MEK inhibitors, the optimal protocol of treatment, and the strategies for managing adverse events. Conclusions: Treatment is shifting to a wider multidisciplinary management, where a key role is played by targeted therapies, to improve outcomes and quality of life for patients with BRAF-mutated craniopharyngiomas. Future, larger comparative trials will optimize their protocol of use and integration into multimodal strategies of treatment.
Collapse
Affiliation(s)
- Giulia Cossu
- Department of Neurosurgery, University Hospital of Lausanne and University of Lausanne, 1011 Lausanne, Switzerland; (R.T.D.)
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, 21000 Dijon, France (C.C.); (W.F.); (M.L.)
| | - Daniele S. C. Ramsay
- Imperial Brain and Spine Initiative, London W2 1NY, UK
- Imperial College School of Medicine, London W2 1PG, UK
| | - Roy T. Daniel
- Department of Neurosurgery, University Hospital of Lausanne and University of Lausanne, 1011 Lausanne, Switzerland; (R.T.D.)
| | - Ahmed El Cadhi
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, 21000 Dijon, France (C.C.); (W.F.); (M.L.)
| | - Luc Kerherve
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, 21000 Dijon, France (C.C.); (W.F.); (M.L.)
| | - Edouard Morlaix
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, 21000 Dijon, France (C.C.); (W.F.); (M.L.)
| | - Sayda A. Houidi
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, 21000 Dijon, France (C.C.); (W.F.); (M.L.)
| | - Clément Millot-Piccoli
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, 21000 Dijon, France (C.C.); (W.F.); (M.L.)
| | - Renan Chapon
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, 21000 Dijon, France (C.C.); (W.F.); (M.L.)
| | - Tuan Le Van
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, 21000 Dijon, France (C.C.); (W.F.); (M.L.)
| | - Catherine Cao
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, 21000 Dijon, France (C.C.); (W.F.); (M.L.)
| | - Walid Farah
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, 21000 Dijon, France (C.C.); (W.F.); (M.L.)
| | - Maxime Lleu
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, 21000 Dijon, France (C.C.); (W.F.); (M.L.)
| | - Olivier Baland
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, 21000 Dijon, France (C.C.); (W.F.); (M.L.)
| | - Jacques Beaurain
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, 21000 Dijon, France (C.C.); (W.F.); (M.L.)
| | - Jean Michel Petit
- Department of Endocrinology, University Hospital of Dijon Bourgogne, 21000 Dijon, France
| | - Brivaël Lemogne
- Department of Neuroradiology, University Hospital of Dijon Bourgogne, 21000 Dijon, France
| | - Mahmoud Messerer
- Department of Neurosurgery, University Hospital of Lausanne and University of Lausanne, 1011 Lausanne, Switzerland; (R.T.D.)
| | - Moncef Berhouma
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, 21000 Dijon, France (C.C.); (W.F.); (M.L.)
- Functional and Molecular Imaging Team (CNRS 6302), Molecular Chemistry Institute (ICMUB), University of Burgundy, 21078 Dijon, France
| |
Collapse
|
45
|
Soliman SS, Abd El-Samie FE, Abd El-Atty SM, Badawy W, Eshra A. DNA nanotechnology for cell-free DNA marker for tumor detection: a comprehensive overview. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2024; 44:276-290. [PMID: 39357047 DOI: 10.1080/15257770.2024.2337853] [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: 10/16/2023] [Revised: 02/13/2024] [Accepted: 03/17/2024] [Indexed: 10/04/2024]
Abstract
Advancements in DNA nanotechnology have led to new exciting ways to detect cell-free tumor biomarkers, revolutionizing cancer diagnostics. This article comprehensively reviews recent developments in this field, discussing the significance of liquid biopsies and DNA nanomachines in early cancer detection. The accuracy of cancer diagnosis at its early stages is expected to be significantly improved by identifying biomarkers. Liquid biopsies, offering minimally-invasive testing, hold the potential for capturing tumor-specific components like circulating tumor cells, cell-free DNA, and exosomes. DNA nanomachines are advanced molecular devices that exploit the programmability of DNA sequences for the ultrasensitive and specific detection of these markers. DNA nanomachines, nanostructures made of DNA that can be designable and switchable nanostructures, have a wide range of advantages for detecting tumor biomarkers, including non-invasiveness, affordability, high sensitivity, and specificity. Scientists also work on dealing with challenges like low marker concentrations and interference, which are addressed through microfluidic integration, nanomaterial amplification, and indirect signal detection. Despite advances, multiplex detection remains a challenge. In conclusion, DNA nanomachines bear immense promise for cancer diagnostics, advocating personalized treatment and improving patient outcomes. Continued research could redefine how we find and treat tumors, leading to better patient outcomes.
Collapse
Affiliation(s)
- Sara Sami Soliman
- Department of Electronics and Electrical Communications Engineering, Faculty of Electronic Engineering, Menoufia University, Menouf, Egypt
| | - Fathi E Abd El-Samie
- Department of Electronics and Electrical Communications Engineering, Faculty of Electronic Engineering, Menoufia University, Menouf, Egypt
| | - Saied M Abd El-Atty
- Department of Electronics and Electrical Communications Engineering, Faculty of Electronic Engineering, Menoufia University, Menouf, Egypt
| | - Wael Badawy
- School of physics, Engineering, and Computer Science, University of Hertfordshire Hosted by GAF, Cairo, Egypt
| | - Abeer Eshra
- Department of Computer Science and Engineering, Faculty of Electronic Engineering, Menoufia University, Menouf, Egypt
- Hamilton Institute, Maynooth University, Co. Kildare, Ireland
| |
Collapse
|
46
|
Kinane DF, Gabert J, Xynopoulos G, Guzeldemir‐Akcakanat E. Strategic approaches in oral squamous cell carcinoma diagnostics using liquid biopsy. Periodontol 2000 2024; 96:316-328. [PMID: 38676371 PMCID: PMC11579816 DOI: 10.1111/prd.12567] [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: 11/01/2023] [Revised: 02/06/2024] [Accepted: 03/23/2024] [Indexed: 04/28/2024]
Abstract
Liquid biopsy is a noninvasive diagnostic technique used for monitoring cancer utilizing specific genetic biomarkers present in bodily fluids, such as blood, saliva, or urine. These analyses employ multiple biomolecular sources including circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), and exosomes (that contain DNA fragments) to detect genetic biomarkers that can predict, disclose, and/or monitor cancers. Levels of these biomarkers can inform on the presence of cancer, its genetic characteristics, and its potential treatment response and also provide predictive genetic predisposition information for specific cancers including oral squamous cell carcinomas (OSCC). Liquid biopsies can aid cancer management as they offer real-time dynamic information on the response to say chemotherapy or radiotherapy and recurrence following surgical excision. Unlike traditional tissue biopsies, which are invasive with a degree of morbidity and require specific tumor location sampling, liquid biopsies are noninvasive and can be repeated frequently. For oral squamous cell carcinoma, on which this review focuses, liquid biopsy of blood or saliva can be valuable in predicting susceptibility, providing early detection, and monitoring the disease's progression and response to therapy. This review gives a general narrative overview of the technology, its current medical usage, and advantages and disadvantages compared with current techniques and discusses a range of current potential biomarkers for disclosing OSCC and predicting its risk. Oral squamous cell carcinoma is all too often detected in the late stages. In future, liquid biopsy may provide an effective screening process such that cancers including OSCC will be detected in the early stages rather than later when prognosis is poor and morbidity and debilitation are greater. In this screening process, periodontists and hygienists have a critical role in that they are adept in examining mucosa, they see patients with shared risk factors for periodontitis and OSCC, namely smoking and poor oral hygiene, and they see patients frequently such that OSCC examinations should be a routine part of the recall visit. With this additional screening manpower, oral medicine and oral surgery colleagues will detect OSCC earlier and this coupled with new techniques such as liquid biopsy may greatly decrease global morbidity in OSCC.
Collapse
Affiliation(s)
- Denis F. Kinane
- Department of Periodontology, Dental SchoolUniversity BernBernSwitzerland
- ExpressTestCignpost Diagnostics Ltd.FarnboroughUnited Kingdom
| | | | | | - Esra Guzeldemir‐Akcakanat
- Department of Periodontology, Faculty of DentistryKocaeli UniversityİzmitTurkey
- College of Dental MedicineQU Health, Qatar UniversityQatarQatar
| |
Collapse
|
47
|
Fu SW, Tang C, Tan X, Srivastava S. Liquid biopsy for early cancer detection: technological revolutions and clinical dilemma. Expert Rev Mol Diagn 2024; 24:937-955. [PMID: 39360748 DOI: 10.1080/14737159.2024.2408744] [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: 05/08/2024] [Accepted: 09/22/2024] [Indexed: 10/05/2024]
Abstract
INTRODUCTION Liquid biopsy is an innovative advancement in oncology, offering a noninvasive method for early cancer detection and monitoring by analyzing circulating tumor cells, DNA, RNA, and other biomarkers in bodily fluids. This technique has the potential to revolutionize precision oncology by providing real-time analysis of tumor dynamics, enabling early detection, monitoring treatment responses, and tailoring personalized therapies based on the molecular profiles of individual patients. AREAS COVERED In this review, the authors discuss current methodologies, technological challenges, and clinical applications of liquid biopsy. This includes advancements in detecting minimal residual disease, tracking tumor evolution, and combining liquid biopsy with other diagnostic modalities for precision oncology. Key areas explored are the sensitivity, specificity, and integration of multi-omics, AI, ML, and LLM technologies. EXPERT OPINION Liquid biopsy holds great potential to revolutionize cancer care through early detection and personalized treatment strategies. However, its success depends on overcoming technological and clinical hurdles, such as ensuring high sensitivity and specificity, interpreting results amidst tumor heterogeneity, and making tests accessible and affordable. Continued innovation and collaboration are crucial to fully realize the potential of liquid biopsy in improving early cancer detection, treatment, and monitoring.
Collapse
Affiliation(s)
- Sidney W Fu
- Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Cong Tang
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Xiaohui Tan
- Division of LS Research, LSBioscience, LLC, Frederick, USA
| | - Sudhir Srivastava
- Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
48
|
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
|
49
|
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
|
50
|
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
|