1
|
Kim TM, Subba SH, Hwang YK, Kim SG, Park J, Jin EJ, Park SY. Electrical and fluorescence in situ monitoring of tumor microenvironment-based pH-responsive polymer dot coated surface. Talanta 2025; 281:126840. [PMID: 39265419 DOI: 10.1016/j.talanta.2024.126840] [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: 06/19/2024] [Revised: 09/03/2024] [Accepted: 09/07/2024] [Indexed: 09/14/2024]
Abstract
A boronate-ester structure forming a pH-responsive polymer dot (Plu-PD) coated biosensor between carbonized-sp2 rich dopamine-alginate [PD(Alg)] and boronic acid-grafted Pluronic (BA-Pluronic) was developed for the electrochemical and fluorescence detection of cancer cells. The reduced fluorescence (FL) resulting from fluorescence resonance energy transfer (FRET) mediated by π-π interactions within Plu-PD was successfully reinvigorated through the specific cleavage of the boronate-ester bond, triggered by the acidic conditions prevailing in the cancer microenvironment. The anomalous variations in extracellular pH levels observed in cancer (pH ∼6.8), as opposed to the normal cellular pH range of approximately 7.4, serve as robust indicators for discerning cancer cells from their healthy counterparts. Moreover, the Plu-PD coated surface demonstrated remarkable adaptability in modulating its surface structure, concurrently exhibiting tunable electroconductivity under reduced pH conditions, thereby imparting selective responsiveness to cancer cells. The pH-modulated conductivity change was validated by a reduction in resistance from 211 ± 9.7 kΩ at pH 7.4 to 73.9 ± 9.4 kΩ and 61.5 ± 11.5 kΩ at pH 6.8 and 6.0, respectively. The controllable electrochemical characteristics were corroborated through in vitro treatment of cancer cells (HeLa, B16F10, and SNU-C2A) via LED experiments and wireless output analysis. In contrast, identical treatments yielded a limited response in normal cell line (CHO-K1). Notably, the Plu-PD coated surface can be seamlessly integrated with a wireless system to facilitate real-time monitoring of the sensing performance in the presence of cancer and normal cells, enabling rapid and accurate cancer diagnosis using a smartphone.
Collapse
Affiliation(s)
- Tae Min Kim
- Department of IT and Energy Convergence (BK21 FOUR), Korea National University of Transportation, Chungju, 27469, Republic of Korea
| | - Sunu Hangma Subba
- Department of IT and Energy Convergence (BK21 FOUR), Korea National University of Transportation, Chungju, 27469, Republic of Korea
| | - Yoon Kil Hwang
- Department of IT and Energy Convergence (BK21 FOUR), Korea National University of Transportation, Chungju, 27469, Republic of Korea
| | - Seul Gi Kim
- Department of Green Bio Engineering, Korea National University of Transportation, Chungju, 27469, Republic of Korea
| | - Junyoung Park
- Department of Biological Sciences, College of Health Sciences, Wonkwang University, Jeonbuk, 54538, Iksan, Republic of Korea
| | - Eun-Jung Jin
- Department of Biological Sciences, College of Health Sciences, Wonkwang University, Jeonbuk, 54538, Iksan, Republic of Korea
| | - Sung Young Park
- Department of IT and Energy Convergence (BK21 FOUR), Korea National University of Transportation, Chungju, 27469, Republic of Korea; Department of Green Bio Engineering, Korea National University of Transportation, Chungju, 27469, Republic of Korea; Department of Chemical and Biological Engineering, Korea National University of Transportation, Chungju, 27469, Republic of Korea.
| |
Collapse
|
2
|
Kocabey S, Cattin S, Gray I, Rüegg C. Ultrasensitive detection of cancer-associated nucleic acids and mutations by primer exchange reaction-based signal amplification and flow cytometry. Biosens Bioelectron 2025; 267:116839. [PMID: 39369516 DOI: 10.1016/j.bios.2024.116839] [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: 04/23/2024] [Revised: 09/27/2024] [Accepted: 10/04/2024] [Indexed: 10/08/2024]
Abstract
The detection of cancer-associated nucleic acids and mutations through liquid biopsy has emerged as a highly promising non-invasive approach for early cancer detection and monitoring. In this study, we report the development of primer exchange reaction (PER) based signal amplification strategy that enables the rapid, sensitive and specific detection of nucleic acids bearing cancer specific single nucleotide mutations using flow cytometry. Using micrometer size beads as support for immobilizing oligonucleotides and programmable PER assembly for target oligonucleotide recognition and fluorescence signal amplification, we demonstrated the versatile detection of target nucleic acids including KRAS oligonucleotide, fragmented mRNAs, and miR-21. Moreover, our detection system can discriminate single base mutations frequently occurred in cancer-associated genes including KRAS, PIK3CA and P53 from cell extracts and circulating tumor DNAs (ctDNAs). The detection is highly sensitive, with a limit of detection down to 27 fM without pre-amplification. In view of a clinical application, we demonstrate the detection of single mutations after extraction and pre-amplification of ctDNAs from the plasma of breast cancer patients. Importantly, our detection strategy enabled the detection of single KRAS mutation even in the presence of 1000-fold excess of wild type (WT) DNA using multi-color flow cytometry detection approach. Overall, our strategy holds immense potential for clinical applications, offering significant improvements for early cancer detection and monitoring.
Collapse
Affiliation(s)
- Samet Kocabey
- Laboratory of Experimental and Translational Oncology, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Chemin Du Musée 18, PER17, 1700, Fribourg, Switzerland; NCCR Bio-inspired Materials, University of Fribourg, 1700, Fribourg, Switzerland.
| | - Sarah Cattin
- Laboratory of Experimental and Translational Oncology, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Chemin Du Musée 18, PER17, 1700, Fribourg, Switzerland; NCCR Bio-inspired Materials, University of Fribourg, 1700, Fribourg, Switzerland; Cell Analytics Facility, Faculty of Science and Medicine, University of Fribourg, Chemin Du Musée 18, PER17, 1700, Fribourg, Switzerland
| | - Isabelle Gray
- Laboratory of Experimental and Translational Oncology, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Chemin Du Musée 18, PER17, 1700, Fribourg, Switzerland; NCCR Bio-inspired Materials, University of Fribourg, 1700, Fribourg, Switzerland
| | - Curzio Rüegg
- Laboratory of Experimental and Translational Oncology, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Chemin Du Musée 18, PER17, 1700, Fribourg, Switzerland; NCCR Bio-inspired Materials, University of Fribourg, 1700, Fribourg, Switzerland.
| |
Collapse
|
3
|
Rudà R, Pellerino A, Soffietti R. Blood and cerebrospinal fluid biomarkers in neuro-oncology. Curr Opin Neurol 2024; 37:693-701. [PMID: 39329301 DOI: 10.1097/wco.0000000000001317] [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: 09/28/2024]
Abstract
PURPOSE OF REVIEW The purpose of this review is to discuss the value of blood and CSF biomarkers in primary CNS tumors. RECENT FINDINGS Several analytes can be assessed with liquid biopsy techniques, including circulating tumor cells, circulating cell-free tumor DNA, circulating cell-free RNA, circulating proteins and metabolites, extracellular vesicles and tumor-educated platelets. Among diffuse gliomas of the adult, ctDNA in blood or CSF has represented the most used analyte, with the detection of molecular alterations such as MGMT promoter, PTEN, EGFRVIII, TERT promoter mutation and IDH R132H mutation. In general, CSF is enriched for ctDNA as compared with plasma. The use of MRI-guided focused ultrasounds to disrupt the blood-brain barrier could enhance the level of biomarkers in both blood and CSF. The detection of MYD88 L265P mutation with digital droplet PCR and the detection of ctDNA with next generation sequencing represent the best tools to diagnose and monitoring CNS lymphomas under treatment. In meningiomas, the low concentration of ctDNA is a limiting factor for the detection of driver mutations, such as NF2, AKTs, SMO, KLF4, TRAF7, SMARCB1, SMARCE1, PTEN, and TERT; an alternative approach could be the isolation of ctDNA through circulating extracellular vesicles. Liquid biopsies are being used extensively for diagnosis and surveillance of diffuse midline gliomas, in particular with the detection of the driver mutation H3K27M. Last, specific methylome patterns in CSF may allow the distinction of glioblastomas from CNS lymphomas or meningiomas. SUMMARY This review summarizes the current knowledge and future perspectives of liquid biopsy of blood and CSF for diagnosis and monitoring of primary CNS tumors.
Collapse
Affiliation(s)
- Roberta Rudà
- Division of Neuro-Oncology, Department of Neuroscience 'Rita Levi Montalcini', University and City of Health and Science Hospital
| | - Alessia Pellerino
- Division of Neuro-Oncology, Department of Neuroscience 'Rita Levi Montalcini', University and City of Health and Science Hospital
| | | |
Collapse
|
4
|
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
|
5
|
Han S, Park J, Moon S, Eom S, Jin CM, Kim S, Ryu YS, Choi Y, Lee JB, Choi I. Label-free and liquid state SERS detection of multi-scaled bioanalytes via light-induced pinpoint colloidal assembly. Biosens Bioelectron 2024; 264:116663. [PMID: 39167886 DOI: 10.1016/j.bios.2024.116663] [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: 02/11/2024] [Revised: 07/17/2024] [Accepted: 08/10/2024] [Indexed: 08/23/2024]
Abstract
Surface-enhanced Raman scattering (SERS) has been extensively applied to detect complex analytes due to its ability to enhance the fingerprint signals of molecules around nanostructured metallic surfaces. Thus, it is essential to design SERS-active nanostructures with abundant electromagnetic hotspots in a probed volume according to the dimensions of the analytes, as the analytes must be located in their hotspots for maximum signal enhancement. Herein, we demonstrate a simple method for detecting robust SERS signals from multi-scaled bioanalytes, regardless of their dimensions in the liquid state, through a photothermally driven co-assembly with colloidal plasmonic nanoparticles as signal enhancers. Under resonant light illumination, plasmonic nanoparticles and analytes in the solution quickly assemble at the focused surface area by convective movements induced by the photothermal heating of the plasmonic nanoparticles without any surface modification. Such collective assemblies of plasmonic nanoparticles and analytes were optimized by varying the optical density and surface charge of the nanoparticles, the viscosity of the solvent, and the light illumination time to maximize the SERS signals. Using these light-induced co-assemblies, the intrinsic SERS signals of small biomolecules can be detected down to nanomolar concentrations based on their fingerprint spectra. Furthermore, large-sized biomarkers, such as viruses and exosomes, were successfully detected without labels, and the complexity of the collected spectra was statistically analyzed using t-distributed stochastic neighbor embedding combined with support vector machine (t-SNE + SVM). The proposed method is expected to provide a robust and convenient method to sensitively detect biologically and environmentally relevant analytes at multiple scales in liquid samples.
Collapse
Affiliation(s)
- Seungyeon Han
- Department of Life Science, University of Seoul, Seoul, 02504, Republic of Korea
| | - Junhee Park
- Department of Life Science, University of Seoul, Seoul, 02504, Republic of Korea
| | - Sunghyun Moon
- Department of Chemical Engineering, University of Seoul, Seoul, 02504, Republic of Korea
| | - Seonghyeon Eom
- Department of Life Science, University of Seoul, Seoul, 02504, Republic of Korea
| | - Chang Min Jin
- Department of Life Science, University of Seoul, Seoul, 02504, Republic of Korea
| | - Seungmin Kim
- School of Biomedical Engineering, Korea University, Seoul, 02481, Republic of Korea; Interdisciplinary Program in Precision Public Health, Korea University, Seoul, 02481, Republic of Korea
| | - Yong-Sang Ryu
- School of Biomedical Engineering, Korea University, Seoul, 02481, Republic of Korea; Interdisciplinary Program in Precision Public Health, Korea University, Seoul, 02481, Republic of Korea
| | - Yeonho Choi
- School of Biomedical Engineering, Korea University, Seoul, 02481, Republic of Korea; Interdisciplinary Program in Precision Public Health, Korea University, Seoul, 02481, Republic of Korea; Exopert Corporation, Seoul, 02580, Republic of Korea
| | - Jong Bum Lee
- Department of Chemical Engineering, University of Seoul, Seoul, 02504, Republic of Korea
| | - Inhee Choi
- Department of Life Science, University of Seoul, Seoul, 02504, Republic of Korea; Department of Applied Chemistry, University of Seoul, Seoul, 02504, Republic of Korea.
| |
Collapse
|
6
|
Seyedi S, Harris VK, Kapsetaki SE, Narayanan S, Saha D, Compton Z, Yousefi R, May A, Fakir E, Boddy AM, Gerlinger M, Wu C, Mina L, Huijben S, Gouge DH, Cisneros L, Ellsworth PC, Maley CC. Resistance Management for Cancer: Lessons from Farmers. Cancer Res 2024; 84:3715-3727. [PMID: 39356625 DOI: 10.1158/0008-5472.can-23-3374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 06/29/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024]
Abstract
One of the main reasons we have not been able to cure cancers is that treatments select for drug-resistant cells. Pest managers face similar challenges with pesticides selecting for pesticide-resistant insects, resulting in similar mechanisms of resistance. Pest managers have developed 10 principles that could be translated to controlling cancers: (i) prevent onset, (ii) monitor continuously, (iii) identify thresholds below which there will be no intervention, (iv) change interventions in response to burden, (v) preferentially select nonchemical control methods, (vi) use target-specific drugs, (vii) use the lowest effective dose, (viii) reduce cross-resistance, (ix) evaluate success based on long-term management, and (x) forecast growth and response. These principles are general to all cancers and cancer drugs and so could be employed broadly to improve oncology. Here, we review the parallel difficulties in controlling drug resistance in pests and cancer cells. We show how the principles of resistance management in pests might be applied to cancer. Integrated pest management inspired the development of adaptive therapy in oncology to increase progression-free survival and quality of life in patients with cancers where cures are unlikely. These pest management principles have the potential to inform clinical trial design.
Collapse
Affiliation(s)
- Sareh Seyedi
- Arizona Cancer Evolution Center, Arizona State University, Tempe, Arizona
- Center for Biocomputing, Security and Society, Biodesign Institute, Arizona State University, Tempe, Arizona
- School of Life Sciences, Arizona State University, Tempe, Arizona
| | - Valerie K Harris
- Arizona Cancer Evolution Center, Arizona State University, Tempe, Arizona
- Center for Biocomputing, Security and Society, Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Stefania E Kapsetaki
- Arizona Cancer Evolution Center, Arizona State University, Tempe, Arizona
- Center for Biocomputing, Security and Society, Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Shrinath Narayanan
- Center for Biocomputing, Security and Society, Biodesign Institute, Arizona State University, Tempe, Arizona
- Department of Ecology and Evolution, University of Lausanne, Lausanne, Switzerland
| | - Daniel Saha
- Arizona Cancer Evolution Center, Arizona State University, Tempe, Arizona
- Center for Biocomputing, Security and Society, Biodesign Institute, Arizona State University, Tempe, Arizona
- School of Life Sciences, Arizona State University, Tempe, Arizona
| | - Zachary Compton
- Arizona Cancer Evolution Center, Arizona State University, Tempe, Arizona
- Center for Biocomputing, Security and Society, Biodesign Institute, Arizona State University, Tempe, Arizona
- School of Life Sciences, Arizona State University, Tempe, Arizona
- University of Arizona Cancer Center, University of Arizona College of Medicine, Tucson, Arizona
| | - Rezvan Yousefi
- Arizona Cancer Evolution Center, Arizona State University, Tempe, Arizona
- Center for Biocomputing, Security and Society, Biodesign Institute, Arizona State University, Tempe, Arizona
- The Polytechnic School, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, Arizona
| | - Alexander May
- Research Casting International, Quinte West, Ontario, Canada
| | - Efe Fakir
- Istanbul University Cerrahpasa School of Medicine, Istanbul, Turkey
| | - Amy M Boddy
- Arizona Cancer Evolution Center, Arizona State University, Tempe, Arizona
- Exotic Species Cancer Research Alliance, North Carolina State University, Raleigh, North Carolina
- Department of Anthropology, University of California Santa Barbara, Santa Barbara, California
| | - Marco Gerlinger
- Translational Oncogenomics Laboratory, Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
- Gastrointestinal Cancer Unit, The Royal Marsden Hospital, London, United Kingdom
| | - Christina Wu
- Division of Hematology and Medical Oncology, Department of Medicine, Mayo Clinic, Phoenix, Arizona
| | | | - Silvie Huijben
- School of Life Sciences, Arizona State University, Tempe, Arizona
- Center for Evolution and Medicine, Arizona State University, Tempe, Arizona
| | - Dawn H Gouge
- Department of Entomology, University of Arizona, Tucson, Arizona
| | - Luis Cisneros
- Arizona Cancer Evolution Center, Arizona State University, Tempe, Arizona
- Center for Biocomputing, Security and Society, Biodesign Institute, Arizona State University, Tempe, Arizona
- School of Life Sciences, Arizona State University, Tempe, Arizona
| | | | - Carlo C Maley
- Arizona Cancer Evolution Center, Arizona State University, Tempe, Arizona
- Center for Biocomputing, Security and Society, Biodesign Institute, Arizona State University, Tempe, Arizona
- School of Life Sciences, Arizona State University, Tempe, Arizona
- Center for Evolution and Medicine, Arizona State University, Tempe, Arizona
| |
Collapse
|
7
|
Wang Y, Cao Y, Wang Y, Sun J, Wang L, Song X, Zhao X. Construction and analysis of protein-protein interaction network for esophageal squamous cell carcinoma. Comput Biol Med 2024; 182:109156. [PMID: 39276610 DOI: 10.1016/j.compbiomed.2024.109156] [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: 04/16/2024] [Revised: 09/08/2024] [Accepted: 09/11/2024] [Indexed: 09/17/2024]
Abstract
Esophageal squamous cell carcinoma (ESCC) is a prevalent malignant tumor of the digestive tract. Clinical findings reveal that the five-year survival rate for mid-to late-stage ESCC patients is merely around 20 %, whereas those diagnosed at an early stage can achieve up to a 95 % survival rate. Consequently, early detection is paramount to improving ESCC patient survival. Protein markers are essential for diagnosing diseases, and the identification of new candidate proteins associated with ESCC through the protein-protein interaction (PPI) network is aimed for in this paper. The PPI network related to ESCC was constructed using protein data, comprising 2094 nodes and 19,660 edges. To assess the nodes' importance in the network, three metrics-degree centrality, betweenness centrality, and closeness centrality-were employed, leading to the identification of 81 key proteins. Subsequently, the biological significance of these proteins in the network was explored, combining biomedical knowledge from three perspectives: network, node, and cluster. The results demonstrated that 52 out of 81 key proteins were confirmed to be linked to ESCC. Among the remaining 29 unreported proteins, 18 displayed significant biological significance, indicating their potential as protein markers related to ESCC.
Collapse
Affiliation(s)
- Yanfeng Wang
- School of Electrical and Information Engineering, Zhengzhou University of Light Industry, Zhengzhou, 450002, China
| | - Yuhan Cao
- School of Electrical and Information Engineering, Zhengzhou University of Light Industry, Zhengzhou, 450002, China
| | - Yingcong Wang
- School of Electrical and Information Engineering, Zhengzhou University of Light Industry, Zhengzhou, 450002, China.
| | - Junwei Sun
- School of Electrical and Information Engineering, Zhengzhou University of Light Industry, Zhengzhou, 450002, China
| | - Lidong Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment and Henan Key Laboratory for Esophageal Cancer Research of the First Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, China
| | - Xin Song
- State Key Laboratory of Esophageal Cancer Prevention & Treatment and Henan Key Laboratory for Esophageal Cancer Research of the First Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, China
| | - Xueke Zhao
- State Key Laboratory of Esophageal Cancer Prevention & Treatment and Henan Key Laboratory for Esophageal Cancer Research of the First Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, China
| |
Collapse
|
8
|
Sirajee AS, Kabiraj D, De S. Cell-free nucleic acid fragmentomics: A non-invasive window into cellular epigenomes. Transl Oncol 2024; 49:102085. [PMID: 39178576 PMCID: PMC11388671 DOI: 10.1016/j.tranon.2024.102085] [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: 05/07/2024] [Revised: 08/01/2024] [Accepted: 08/11/2024] [Indexed: 08/26/2024] Open
Abstract
Clinical genomic profiling of cell-free nucleic acids (e.g. cell-free DNA or cfDNA) from blood and other body fluids has ushered in a new era in non-invasive diagnostics and treatment monitoring strategies for health conditions and diseases such as cancer. Genomic analysis of cfDNAs not only identifies disease-associated mutations, but emerging findings suggest that structural, topological, and fragmentation characteristics of cfDNAs reveal crucial information about the location of source tissues, their epigenomes, and other clinically relevant characteristics, leading to the burgeoning field of fragmentomics. The field has seen rapid developments in computational and genomics methodologies for conducting large-scale studies on health conditions and diseases - that have led to fundamental, mechanistic discoveries as well as translational applications. Several recent studies have shown the clinical utilities of the cfDNA fragmentomics technique which has the potential to be effective for early disease diagnosis, determining treatment outcomes, and risk-free continuous patient monitoring in a non-invasive manner. In this article, we outline recent developments in computational genomic methodologies and analysis strategies, as well as the emerging insights from cfNA fragmentomics. We conclude by highlighting the current challenges and opportunities.
Collapse
Affiliation(s)
- Ahmad Salman Sirajee
- Department of Pathology and Laboratory Medicine, Rutgers Cancer Institute, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA.
| | - Debajyoti Kabiraj
- Department of Pathology and Laboratory Medicine, Rutgers Cancer Institute, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Subhajyoti De
- Department of Pathology and Laboratory Medicine, Rutgers Cancer Institute, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA.
| |
Collapse
|
9
|
Zhang Y, Tian L. Advances and challenges in the use of liquid biopsy in gynaecological oncology. Heliyon 2024; 10:e39148. [PMID: 39492906 PMCID: PMC11530831 DOI: 10.1016/j.heliyon.2024.e39148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 10/08/2024] [Accepted: 10/08/2024] [Indexed: 11/05/2024] Open
Abstract
Ovarian cancer, endometrial cancer, and cervical cancer are the three primary gynaecological cancers that pose a significant threat to women's health on a global scale. Enhancing global cancer survival rates necessitates advancements in illness detection and monitoring, with the goal of improving early diagnosis and prognostication of disease recurrence. Conventional methods for identifying and tracking malignancies rely primarily on imaging techniques and, when possible, protein biomarkers found in blood, many of which lack specificity. The process of collecting tumour samples necessitates intrusive treatments that are not suitable for specific purposes, such as screening, predicting, or evaluating the effectiveness of treatment, monitoring the presence of remaining illness, and promptly detecting relapse. Advancements in treatment are being made by the detection of genetic abnormalities in tumours, both inherited and acquired. Newly designed therapeutic approaches can specifically address some of these abnormalities. Liquid biopsy is an innovative technique for collecting samples that examine specific cancer components that are discharged into the bloodstream, such as circulating tumour DNA (ctDNA), circulating tumour cells (CTCs), cell-free RNA (cfRNA), tumour-educated platelets (TEPs), and exosomes. Mounting data indicates that liquid biopsy has the potential to improve the clinical management of gynaecological cancers through enhanced early diagnosis, prognosis prediction, recurrence detection, and therapy response monitoring. Understanding the distinct genetic composition of tumours can also inform therapy choices and the identification of suitable targeted treatments. The main benefits of liquid biopsy are its non-invasive characteristics and practicality, enabling the collection of several samples and the continuous monitoring of tumour changes over time. This review aims to provide an overview of the data supporting the therapeutic usefulness of each component of liquid biopsy. Additionally, it will assess the benefits and existing constraints associated with the use of liquid biopsy in the management of gynaecological malignancies. In addition, we emphasise future prospects in light of the existing difficulties and investigate areas where further research is necessary to clarify its rising clinical capabilities.
Collapse
Affiliation(s)
- Yingfeng Zhang
- University-Town Hospital of Chongqing Medical University, Chongqing, 401331, China
| | - Libi Tian
- University-Town Hospital of Chongqing Medical University, Chongqing, 401331, China
| |
Collapse
|
10
|
Gagliardi I, Campolo F, Borges de Souza P, Rossi L, Albertelli M, Grillo F, Caputi L, Mazza M, Faggiano A, Zatelli MC. Comparative Targeted Genome Profiling between Solid and Liquid Biopsies in Gastroenteropancreatic Neuroendocrine Neoplasms: A Proof-of-Concept Pilot Study. Neuroendocrinology 2024:1-12. [PMID: 39447548 DOI: 10.1159/000541346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/19/2024] [Indexed: 10/26/2024]
Abstract
INTRODUCTION Clinical presentation and genetic profile of gastroenteropancreatic neuroendocrine tumors (GEP-NETs) are highly variable, hampering their management. Sequencing of circulating tumor DNA from liquid biopsy (LB) has been proposed as a less invasive alternative to solid biopsy (SB). Our aim was to compare the mutational profile (MP) provided by LB with that deriving from SB in GEP-NETs. METHODS SB and LB were derived simultaneously from 6 GEP-NET patients. A comparative targeted next-generation sequencing (NGS) analysis was performed on DNA from SB and LB to evaluate the mutational status of 11 genes (MEN1, DAXX, ATRX, MUTYH, SETD2, DEPDC5, TSC2, ARID1A, CHECK2, MTOR, and PTEN). RESULTS Patients (M:F = 2:1; median age 64 years) included 3 with pancreatic and 3 with ileal NETs. NGS detected a median number of 55 variants/sample in SB and 66.5 variants/sample in LB specimens (mutational burden: 0.2-1.9 and 0.3-1.8 mut/Mb, respectively). Missense and nonsense mutations were prevalent in both, mainly represented by C>T transitions. ARID1A, MTOR, and ATRX were consistently mutated in SB, and ARID1A, TSC2, MEN1, PTEN, SETD2, and MUTYH were consistently mutated in LB. DAXX mutations were absent in LB. Seventeen recurrent mutations were shared between SB and LB; in particular, MTOR single-nucleotide variants c.G4731A and c.C2997T were shared by 5 out of 6 patients. Hierarchical clustering supported genetic similarity between SB and LB. CONCLUSIONS This pilot study explores the applicability of LB in GEP-NET MP evaluation. Further studies with larger cohorts are needed to validate LB and to define the clinical impact.
Collapse
Affiliation(s)
- Irene Gagliardi
- Section of Endocrinology and Internal Medicine, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Federica Campolo
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Lucrezia Rossi
- Section of Endocrinology and Internal Medicine, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Manuela Albertelli
- Endocrinology, Department of Internal Medicine and Medical Specialties (DiMI), University of Genova, Genova, Italy
- Endocrinology Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Federica Grillo
- Anatomic Pathology, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genova, Genova, Italy
- Anatomic Pathology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Luigi Caputi
- Freelancer - Independent Researcher, Naples, Italy
| | - Massimiliano Mazza
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Antongiulio Faggiano
- Endocrinology Unit, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Maria Chiara Zatelli
- Section of Endocrinology and Internal Medicine, Department of Medical Sciences, University of Ferrara, Ferrara, Italy,
| |
Collapse
|
11
|
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
|
12
|
Fagery M, Khorshidi HA, Wong SQ, Karanfil Ö, Emery J, IJzerman MJ. Integrating Multi-Cancer Early Detection (MCED) Tests with Standard Cancer Screening: System Dynamics Model Development and Feasibility Testing. PHARMACOECONOMICS - OPEN 2024:10.1007/s41669-024-00533-3. [PMID: 39424759 DOI: 10.1007/s41669-024-00533-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/25/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Cancer screening plays a critical role in early disease detection and improving outcomes. In Australia, established screening protocols for colorectal, breast and cervical cancer have significantly contributed to timely cancer detection. However, the recent introduction of multi-cancer early detection (MCED) tests arguably can disrupt current screening, yet the extent to which these tests provide additional benefits remains uncertain. We present the development and initial validation of a system dynamics (SD) model that estimates the additional cancer detections and costs associated with MCED tests. AIM This article describes the development of a simulation model built to evaluate the additional patient diagnoses and the economic impact of incorporating MCED testing alongside Australia's well-established standard of care (SOC) screening programs for colorectal, breast, cervical and lung cancers. The model was designed to estimate the additional number of patients diagnosed at each cancer stage (stage I, II, III, IV, or unknown) and the associated costs. This simulation model allows for the analysis of multiple scenarios under a plausible set of assumptions regarding population-level participation rates. METHODS An SD model was developed to represent the existing SOC national cancer screening pathways and to integrate potential clinical pathways that could be introduced by MCED tests. The SD model was built to investigate three scenarios for the use of MCED testing: firstly, to explore the viability of MCED testing as a substitute among individuals who are not opting for SOC screening for any reason; secondly, to implement MCED testing exclusively for individuals ineligible for SOC screening, yet have high-risk characteristics; and thirdly, to employ MCED testing after SOC screening to serve as a triaging/confirmatory tool for individuals receiving inconclusive test results. The three primary scenarios were constructed by varying diagnostic accuracy and uptake rates of MCED tests. DISCUSSION The clinical utility and outcomes of MCED testing for screening and early detection still lack comprehensive evidence. Nonetheless, this simulation model facilitates a thorough analysis of MCED tests within the Australian healthcare context, providing insights into potential additional detections and costs to the healthcare system, which may help prioritise future evidence development. The adaptable yet novel SD model presented herein is anticipated to be of considerable interest to industry, policymakers, consumers and clinicians involved in informing clinical and economic decisions regarding integrating MCED tests as cancer screening and early detection tools. The expected results of applying this SD model will determine whether using MCED testing in conjunction with SOC screening offers any potential benefits, possibly guiding policy decisions and clinical practices towards the adoption of MCED tests.
Collapse
Affiliation(s)
- Mussab Fagery
- Cancer Health Services Research, Centre for Cancer Research, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia.
- Cancer Health Services Research, Centre for Health Policy, Melbourne School of Population and Global Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia.
| | - Hadi A Khorshidi
- Cancer Health Services Research, Centre for Cancer Research, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
- Cancer Health Services Research, Centre for Health Policy, Melbourne School of Population and Global Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
| | - Stephen Q Wong
- Peter MacCallum Cancer Centre, Melbourne, Australia
- Department of Oncology, Sir Peter MacCallum, University of Melbourne, Melbourne, Australia
| | - Özge Karanfil
- College of Administrative Sciences and Economics and School of Medicine, Koç University, Istanbul, Turkey
- MIT Sloan School of Management, Boston, MA, USA
| | - Jon Emery
- Department of General Practice and Primary Care, Centre for Cancer Research, University of Melbourne, Melbourne, Australia
| | - Maarten J IJzerman
- Cancer Health Services Research, Centre for Cancer Research, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
- Cancer Health Services Research, Centre for Health Policy, Melbourne School of Population and Global Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
- Erasmus School of Health Policy and Management, Erasmus University Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|
13
|
Boscolo Bragadin A, Del Bianco P, Zulato E, Attili I, Pavan A, Carlet J, Marra L, Guarneri V, Indraccolo S, Bonanno L. Longitudinal liquid biopsy predicts clinical benefit from immunotherapy in advanced non-small cell lung cancer. NPJ Precis Oncol 2024; 8:234. [PMID: 39420036 PMCID: PMC11486993 DOI: 10.1038/s41698-024-00704-9] [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: 01/09/2024] [Accepted: 09/11/2024] [Indexed: 10/19/2024] Open
Abstract
High heterogeneity in clinical benefit characterizes the use of immune checkpoint inhibitors (ICIs) in non-small cell lung cancer (NSCLC). We prospectively enrolled 113 advanced NSCLC patients treated with ICIs and performed liquid biopsy at the time of ICI start (T1), after 3 weeks (T2) and at the time of radiological evaluation (T3). Molecular variables were associated with outcome endpoints: cfDNA quantification, its dynamic change (∆T2-T1), variant allele frequency (VAF) of the gene with the highest frequency detected at baseline with NGS (maxVAF) and its dynamic change (∆T2-T1). At multivariate analysis, PD-L1 negativity, T1 cfDNA, cfDNA increase (∆T2-T1), and T2 VAF were significantly associated with shorter progression-free survival (PFS); PD-L1 negativity, squamous histology, T1 cfDNA, cfDNA (∆T2-T1) increase, and T2 maxVAF affected overall survival (OS). Among high PD-L1 expressing patients treated in first-line, elevated T2 maxVAF and cfDNA increase (∆T2-T1) correlated with worse PFS; higher T2 maxVAF and cfDNA increase (∆T2-T1) with worse OS. Derived integrated models were used to build nomograms and categorize different risk groups.
Collapse
Affiliation(s)
- Andrea Boscolo Bragadin
- Basic and Translational Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Paola Del Bianco
- Clinical Research Unit, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Elisabetta Zulato
- Basic and Translational Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Ilaria Attili
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
- Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Alberto Pavan
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
- Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Jessica Carlet
- Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Ludovica Marra
- Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Valentina Guarneri
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
- Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Stefano Indraccolo
- Basic and Translational Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Laura Bonanno
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy.
- Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy.
| |
Collapse
|
14
|
Gu Y, Wang J, Luo Z, Luo X, Lin LL, Ni S, Wang C, Chen H, Su Z, Lu Y, Gan LY, Chen Z, Ye J. Multiwavelength Surface-Enhanced Raman Scattering Fingerprints of Human Urine for Cancer Diagnosis. ACS Sens 2024. [PMID: 39420643 DOI: 10.1021/acssensors.4c01873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Label-free surface-enhanced Raman spectroscopy (SERS) is capable of capturing rich compositional information from complex biosamples by providing vibrational spectra that are crucial for biosample identification. However, increasing complexity and subtle variations in biological media can diminish the discrimination accuracy of traditional SERS excited by a single laser wavelength. Herein, we introduce a multiwavelength SERS approach combined with machine learning (ML)-based classification to improve the discrimination accuracy of human urine specimens for bladder cancer (BCa) diagnosis. This strategy leverages the excitation-wavelength-dependent SERS spectral profiles of complex matrices, which are mainly attributed to wavelength-related vibrational changes in individual analytes and differences in the variation ratios of SERS intensity across different wavelengths among various analytes. By capturing SERS fingerprints under multiple excitation wavelengths, we can acquire more comprehensive and unique chemical information on complex samples. Further experimental examinations with clinical urine specimens, supported by ML algorithms, demonstrate the effectiveness of this multiwavelength strategy and improve the diagnostic accuracy of BCa and staging of its invasion with SERS spectra from increasing numbers of wavelengths. The multiwavelength SERS holds promise as a convenient, cost-effective, and broadly applicable technique for the precise identification of complex matrices and diagnosis of diseases based on body fluids.
Collapse
Affiliation(s)
- Yuqing Gu
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
| | - Jiayi Wang
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P. R. China
| | - Zhewen Luo
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
| | - Xingyi Luo
- College of Physics and Center for Quantum Materials and Devices, Chongqing University, Chongqing 401331, P. R. China
| | - Linley Li Lin
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
| | - Shuang Ni
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
| | - Cong Wang
- Beijing Key Laboratory of Microstructure and Properties of Solids, Institute of Microstructure and Property of Advanced Materials, College of Materials Science and Engineering, Beijing University of Technology, Beijing 100124, P. R. China
| | - Haoran Chen
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
| | - Zehou Su
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
| | - Yao Lu
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
| | - Li-Yong Gan
- College of Physics and Center for Quantum Materials and Devices, Chongqing University, Chongqing 401331, P. R. China
| | - Zhou Chen
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
- Institute of Medical Robotics, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| | - Jian Ye
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
- Institute of Medical Robotics, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
- Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P. R. China
| |
Collapse
|
15
|
Tian H, Shi H, Chen J, Zhu T, Huang Z, Zong C, Jia S, Ruan J, Ge S, Yuan H, Zhang Y, Jiang B, Liu R, Jia R, Fan X, Xu S. Circulating tumor DNA in conjunctival melanoma: landscape and surveillance value. Am J Ophthalmol 2024:S0002-9394(24)00479-3. [PMID: 39424026 DOI: 10.1016/j.ajo.2024.10.012] [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: 05/29/2024] [Revised: 10/03/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024]
Abstract
PURPOSE To evaluate the surveillance value of circulating tumor DNA (ctDNA) for detecting distant metastasis and indicating systemic therapeutic efficacy in conjunctival melanoma (CoM). DESIGN Retrospective, observational case series. METHODS From July 2021 to June 2023, 30 CoM patients in our center underwent plasma ctDNA assessment, out of which 12 individuals presented with distant metastases. We employed a 437-gene panel containing common mutations in CoM and common drug-sensitive mutations using next-generation sequencing (NGS) technology to analyze ctDNA mutations in plasma. Clinical and radiological records were used to assess tumor status. The relationship between ctDNA characteristics, tissue gene mutations, and clinical manifestations were explored. RESULTS CoM-related driver mutations were detected in ctDNA of 11 patients with distant metastasis. The ctDNA were highly consistent with tissue sequencing, mutual driver mutation including BRAF, NRAS, KRAS, NF1, CTNNB1, and TP53 mutation. those with a higher VAF had shorter progression-free survival (PFS, p=0.0475) and overall survival (OS, p=0.0043). The ctDNA variant allele fraction (VAF) was not correlated with the sum of the longest diameters (SLD, p=0.8192) in distant metastasis patients. CONCLUSIONS Positive plasma ctDNA reflected the presence of metastases. The ctDNA could be used as a complement or alternative to tissue sequencing. High VAF ctDNA might indicate rapid disease progression in distant metastasis patients.
Collapse
Affiliation(s)
- Hao Tian
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 20025, People's Republic of China
| | - Hanhan Shi
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 20025, People's Republic of China
| | - Jie Chen
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 20025, People's Republic of China
| | - Tianyu Zhu
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 20025, People's Republic of China
| | - Ziyue Huang
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 20025, People's Republic of China
| | - Chunyan Zong
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 20025, People's Republic of China
| | - Shichong Jia
- Tianjin Eye Hospital, Tianjin Key Lab of Ophthalmology and Visual Science, Nankai University Affiliated Eye Hospital, Tianjin Eye Institute, Tianjin, China
| | - Jing Ruan
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 20025, People's Republic of China
| | - Shengfang Ge
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 20025, People's Republic of China
| | - Haihua Yuan
- Department of Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| | - Yanjie Zhang
- Department of Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| | - Bin Jiang
- Department of Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| | - Rong Liu
- Opthalmology Department of Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Renbing Jia
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 20025, People's Republic of China
| | - Xianqun Fan
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 20025, People's Republic of China.
| | - Shiqiong Xu
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 20025, People's Republic of China.
| |
Collapse
|
16
|
Palacín-Aliana I, García-Romero N, Carrión-Navarro J, Puig-Serra P, Torres-Ruiz R, Rodríguez-Perales S, Viñal D, González-Rumayor V, Ayuso-Sacido Á. ddPCR Overcomes the CRISPR-Cas13a-Based Technique for the Detection of the BRAF p.V600E Mutation in Liquid Biopsies. Int J Mol Sci 2024; 25:10902. [PMID: 39456686 PMCID: PMC11507125 DOI: 10.3390/ijms252010902] [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: 09/06/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
The isolation of circulating tumoral DNA (ctDNA) present in the bloodstream brings about the opportunity to detect genomic aberrations from the tumor of origin. However, the low amounts of ctDNA present in liquid biopsy samples makes the development of highly sensitive techniques necessary to detect targetable mutations for the diagnosis, prognosis, and monitoring of cancer patients. Here, we employ standard genomic DNA (gDNA) and eight liquid biopsy samples from different cancer patients to examine the newly described CRISPR-Cas13a-based technology in the detection of the BRAF p.V600E actionable point mutation and appraise its diagnostic capacity with two PCR-based techniques: quantitative Real-Time PCR (qPCR) and droplet digital PCR (ddPCR). Regardless of its lower specificity compared to the qPCR and ddPCR techniques, the CRISPR-Cas13a-guided complex was able to detect inputs as low as 10 pM. Even though the PCR-based techniques have similar target limits of detection (LoDs), only the ddPCR achieved a 0.1% variant allele frequency (VAF) detection with elevated reproducibility, thus standing out as the most powerful and suitable tool for clinical diagnosis purposes. Our results also demonstrate how the CRISPR-Cas13a can detect low amounts of the target of interest, but its base-pair specificity failed in the detection of actionable point mutations at a low VAF; therefore, the ddPCR is still the most powerful and suitable technique for these purposes.
Collapse
Affiliation(s)
- Irina Palacín-Aliana
- Atrys Health, 08025 Barcelona, Spain; (I.P.-A.); (V.G.-R.)
- Fundación de Investigación HM Hospitales, HM Hospitales, 28015 Madrid, Spain
- Faculty of Science, Universidad de Alcalá, 28801 Madrid, Spain
| | - Noemí García-Romero
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Madrid, Spain; (N.G.-R.); (J.C.-N.)
- Brain Tumor Laboratory, Fundación Vithas, Grupo Hospitales Vithas, 28043 Madrid, Spain
| | - Josefa Carrión-Navarro
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Madrid, Spain; (N.G.-R.); (J.C.-N.)
- Brain Tumor Laboratory, Fundación Vithas, Grupo Hospitales Vithas, 28043 Madrid, Spain
| | - Pilar Puig-Serra
- Human Cancer Genetics Program, Centro Nacional de Investigaciones Oncológicas (CNIO), Molecular Cytogenetics & Genome Editing Unit, Melchor Fernández Almagro, 3, 28029 Madrid, Spain; (P.P.-S.); (R.T.-R.); (S.R.-P.)
| | - Raul Torres-Ruiz
- Human Cancer Genetics Program, Centro Nacional de Investigaciones Oncológicas (CNIO), Molecular Cytogenetics & Genome Editing Unit, Melchor Fernández Almagro, 3, 28029 Madrid, Spain; (P.P.-S.); (R.T.-R.); (S.R.-P.)
- Centro de Investigación Energéticas Medioambientales y Tecnológicas (CIEMAT), Advanced Therapies Unit, Hematopoietic Innovative Therapies Division, Instituto de Investigación Sanitaria Fundación Jimenez Diaz (IIS-FJD, UAM), 28040 Madrid, Spain
| | - Sandra Rodríguez-Perales
- Human Cancer Genetics Program, Centro Nacional de Investigaciones Oncológicas (CNIO), Molecular Cytogenetics & Genome Editing Unit, Melchor Fernández Almagro, 3, 28029 Madrid, Spain; (P.P.-S.); (R.T.-R.); (S.R.-P.)
| | - David Viñal
- Department of Medical Oncology, Hospital Universitario La Paz, 28046 Madrid, Spain;
| | | | - Ángel Ayuso-Sacido
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Madrid, Spain; (N.G.-R.); (J.C.-N.)
- Brain Tumor Laboratory, Fundación Vithas, Grupo Hospitales Vithas, 28043 Madrid, Spain
- Faculty of Medicine, Universidad Francisco de Vitoria, 28223 Madrid, Spain
| |
Collapse
|
17
|
Song J, Cho MH, Cho H, Song Y, Lee SW, Nam HC, Yoon TH, Shin JC, Hong JS, Kim Y, Ekanayake E, Jeon J, You DG, Im SG, Choi GS, Park JS, Carter BC, Balaj L, Seo AN, Miller MA, Park SY, Kang T, Castro CM, Lee H. Amplifying mutational profiling of extracellular vesicle mRNA with SCOPE. Nat Biotechnol 2024:10.1038/s41587-024-02426-6. [PMID: 39375445 DOI: 10.1038/s41587-024-02426-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 09/09/2024] [Indexed: 10/09/2024]
Abstract
Sequencing of messenger RNA (mRNA) found in extracellular vesicles (EVs) in liquid biopsies can provide clinical information such as somatic mutations, resistance profiles and tumor recurrence. Despite this, EV mRNA remains underused due to its low abundance in liquid biopsies, and large sample volumes or specialized techniques for analysis are required. Here we introduce Self-amplified and CRISPR-aided Operation to Profile EVs (SCOPE), a platform for EV mRNA detection. SCOPE leverages CRISPR-mediated recognition of target RNA using Cas13 to initiate replication and signal amplification, achieving a sub-attomolar detection limit while maintaining single-nucleotide resolution. As a proof of concept, we designed probes for key mutations in KRAS, BRAF, EGFR and IDH1 genes, optimized protocols for single-pot assays and implemented an automated device for multi-sample detection. We validated SCOPE's ability to detect early-stage lung cancer in animal models, monitored tumor mutational burden in patients with colorectal cancer and stratified patients with glioblastoma. SCOPE can expedite readouts, augmenting the clinical use of EVs in precision oncology.
Collapse
Affiliation(s)
- Jayeon Song
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Forensic Sciences, Sungkyunkwan University, Suwon, Republic of Korea
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Mi Hyeon Cho
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hayoung Cho
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Younseong Song
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | | | - Ho Chul Nam
- RevoSketch, Inc., Daejeon, Republic of Korea
| | - Tae Ho Yoon
- RevoSketch, Inc., Daejeon, Republic of Korea
| | | | - Jae-Sang Hong
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yejin Kim
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea
| | - Emil Ekanayake
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
| | - Jueun Jeon
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Dong Gil You
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
| | - Sung Gap Im
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Gyu-Seog Choi
- Colorectal Cancer Center, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Jun Seok Park
- Colorectal Cancer Center, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Bob C Carter
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
| | - Leonora Balaj
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
| | - An Na Seo
- Department of Pathology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Miles A Miller
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Soo Yeun Park
- Colorectal Cancer Center, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Taejoon Kang
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea.
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.
| | - Cesar M Castro
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA.
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Hakho Lee
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA.
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea.
| |
Collapse
|
18
|
Fu SW, Tang C, Tan X, Srivastava S. Liquid biopsy for early cancer detection: technological revolutions and clinical dilemma. Expert Rev Mol Diagn 2024:1-19. [PMID: 39360748 DOI: 10.1080/14737159.2024.2408744] [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: 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
|
19
|
Reina C, Šabanović B, Lazzari C, Gregorc V, Heeschen C. Unlocking the future of cancer diagnosis - promises and challenges of ctDNA-based liquid biopsies in non-small cell lung cancer. Transl Res 2024; 272:41-53. [PMID: 38838851 DOI: 10.1016/j.trsl.2024.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/29/2024] [Accepted: 05/30/2024] [Indexed: 06/07/2024]
Abstract
The advent of liquid biopsies has brought significant changes to the diagnosis and monitoring of non-small cell lung cancer (NSCLC), presenting both promise and challenges. Molecularly targeted drugs, capable of enhancing survival rates, are now available to around a quarter of NSCLC patients. However, to ensure their effectiveness, precision diagnosis is essential. Circulating tumor DNA (ctDNA) analysis as the most advanced liquid biopsy modality to date offers a non-invasive method for tracking genomic changes in NSCLC. The potential of ctDNA is particularly rooted in its ability to furnish comprehensive (epi-)genetic insights into the tumor, thereby aiding personalized treatment strategies. One of the key advantages of ctDNA-based liquid biopsies in NSCLC is their ability to capture tumor heterogeneity. This capability ensures a more precise depiction of the tumor's (epi-)genomic landscape compared to conventional tissue biopsies. Consequently, it facilitates the identification of (epi-)genetic alterations, enabling informed treatment decisions, disease progression monitoring, and early detection of resistance-causing mutations for timely therapeutic interventions. Here we review the current state-of-the-art in ctDNA-based liquid biopsy technologies for NSCLC, exploring their potential to revolutionize clinical practice. Key advancements in ctDNA detection methods, including PCR-based assays, next-generation sequencing (NGS), and digital PCR (dPCR), are discussed, along with their respective strengths and limitations. Additionally, the clinical utility of ctDNA analysis in guiding treatment decisions, monitoring treatment response, detecting minimal residual disease, and identifying emerging resistance mechanisms is examined. Liquid biopsy analysis bears the potential of transforming NSCLC management by enabling non-invasive monitoring of Minimal Residual Disease and providing early indicators for response to targeted treatments including immunotherapy. Furthermore, considerations regarding sample collection, processing, and data interpretation are highlighted as crucial factors influencing the reliability and reproducibility of ctDNA-based assays. Addressing these challenges will be essential for the widespread adoption of ctDNA-based liquid biopsies in routine clinical practice, ultimately paving the way toward personalized medicine and improved outcomes for patients with NSCLC.
Collapse
Affiliation(s)
- Chiara Reina
- Pancreatic Cancer Heterogeneity, Candiolo Cancer Institute FPO-IRCCS, Candiolo, Turin, Italy
| | - Berina Šabanović
- Pancreatic Cancer Heterogeneity, Candiolo Cancer Institute FPO-IRCCS, Candiolo, Turin, Italy
| | - Chiara Lazzari
- Department of Medical Oncology, Cancer Institute FPO-IRCCS, Candiolo, Turin, Italy
| | - Vanesa Gregorc
- Department of Medical Oncology, Cancer Institute FPO-IRCCS, Candiolo, Turin, Italy
| | - Christopher Heeschen
- Pancreatic Cancer Heterogeneity, Candiolo Cancer Institute FPO-IRCCS, Candiolo, Turin, Italy;.
| |
Collapse
|
20
|
Romani C, Assoni C, Mattavelli D, Rampinelli V, Piazza C. The prognostic role of salivary miRNAs in oral squamous cell carcinoma: technical challenges and clinical perspectives. ACTA OTORHINOLARYNGOLOGICA ITALICA : ORGANO UFFICIALE DELLA SOCIETA ITALIANA DI OTORINOLARINGOLOGIA E CHIRURGIA CERVICO-FACCIALE 2024; 44:279-284. [PMID: 39283255 PMCID: PMC11556778 DOI: 10.14639/0392-100x-n2981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 04/06/2024] [Indexed: 11/12/2024]
Abstract
In management of oral squamous cell carcinoma (OSCC), only a few biomarkers, ranging from clinical and histopathological features to molecular alterations, have been demonstrated to have clinical and prognostic utility. The intent of this narrative review is to present current findings on the use of salivary microRNAs (miRNAs) as prognostic oncologic biomarkers in patients with OSCC. The ability to predict survival or recurrence during follow-up by quantification of specific miRNAs in saliva has been shown in a number of studies, and serves as a possible feature to address in future well-designed clinical studies to confirm their prognostic value. The non-invasiveness of liquid biopsy techniques, the ease of saliva collection, and the abundance and stability of miRNAs in such a biologic fluid make it an attractive combination to improve management of OSCC. For salivary miRNAs to be used in routine practice, however, methodological and sampling standardisation are still needed to increase the power and accuracy of the results obtained.
Collapse
Affiliation(s)
- Chiara Romani
- Angelo Nocivelli Institute of Molecular Medicine, ASST Spedali Civili of Brescia, Brescia, Italy
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, School of Medicine, Brescia, Italy
| | - Claudia Assoni
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, School of Medicine, Brescia, Italy
- Medical Oncology Unit, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Davide Mattavelli
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, School of Medicine, Brescia, Italy
- Unit of Otorhinolaryngology – Head and Neck Surgery, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Vittorio Rampinelli
- Unit of Otorhinolaryngology – Head and Neck Surgery, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Cesare Piazza
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, School of Medicine, Brescia, Italy
- Unit of Otorhinolaryngology – Head and Neck Surgery, ASST Spedali Civili of Brescia, Brescia, Italy
| |
Collapse
|
21
|
Peng T, Zhang H, Li L, Cao C, Xu M, Liu X, Lin S, Wu P, Chu T, Liu B, Xu Y, Zhang Y, Wang Y, Yu J, Ding W, Jin X, Wu P. Plasma Cell-Free DNA Concentration and Fragmentomes Predict Neoadjuvant Chemotherapy Response in Cervical Cancer Patients. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2309422. [PMID: 39319610 DOI: 10.1002/advs.202309422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 05/18/2024] [Indexed: 09/26/2024]
Abstract
Cervical cancer remains one of the most lethal gynecological malignancies. However, biomarkers for more precise patient care are an unmet need. Herein, the concentration of 285 plasma cell-free DNA (cfDNA) samples are analyzed from 84 cervical patients and the clinical significance of cfDNA fragmentomic characteristics across the neoadjuvant chemotherapy (NACT) treatment. Patients with poor NACT response exhibit a significantly greater escalation in cfDNA levels following the initial cycle of treatment, in comparison to patients with a favorable response. Distinctive end motif profiles and promoter coverages of cfDNA in initial plasma are observed between patients with differing NACT responses. Notably, the DNASE1L3 analysis further demonstrates the intrinsic association between cfDNA characteristics and chemotherapy resistance. The cfDNA and motif ratios show a good discriminative capacity for predicting non-responders from responders (area under the curve (AUC) > 0.8). In addition, transcriptional start sites (TSS) coverages around promoters discern the alteration of biological processes associated with chemotherapy resistance and reflect the potential value in predicting chemotherapy response. These findings in predictive biomarkers may optimize treatment selection, minimize unnecessary treatment, and assist in establishing personalized treatment strategies for cervical cancer patients.
Collapse
Affiliation(s)
- Ting Peng
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
- National Clinical Research Center for Gynecology and Obstetrics, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | | | - Lingguo Li
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Canhui Cao
- National Clinical Research Center for Gynecology and Obstetrics, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Miaochun Xu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
- National Clinical Research Center for Gynecology and Obstetrics, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Xiaojie Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
- National Clinical Research Center for Gynecology and Obstetrics, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Shitong Lin
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
- National Clinical Research Center for Gynecology and Obstetrics, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Ping Wu
- National Clinical Research Center for Gynecology and Obstetrics, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Tian Chu
- National Clinical Research Center for Gynecology and Obstetrics, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Binghan Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
- National Clinical Research Center for Gynecology and Obstetrics, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Yashi Xu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
- National Clinical Research Center for Gynecology and Obstetrics, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Yan Zhang
- BGI Research, Shenzhen, 518083, China
| | | | - Jinjin Yu
- Department of Obstetrics and Gynecology, Affiliated Hospital of Jiangnan University, Wuxi Medical College, Jiangnan University, Wuxi, 214000, China
| | - Wencheng Ding
- National Clinical Research Center for Gynecology and Obstetrics, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Xin Jin
- BGI Research, Shenzhen, 518083, China
| | - Peng Wu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
- National Clinical Research Center for Gynecology and Obstetrics, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| |
Collapse
|
22
|
Sebutsoe XM, Tsotetsi NJN, Jantjies ZE, Raphela-Choma PP, Choene MS, Motadi LR. Therapeutic Strategies in Advanced Cervical Cancer Detection, Prevention and Treatment. Onco Targets Ther 2024; 17:785-801. [PMID: 39345275 PMCID: PMC11439348 DOI: 10.2147/ott.s475132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 08/08/2024] [Indexed: 10/01/2024] Open
Abstract
Cervical cancer is ranked the fourth most common cause of cancer related deaths amongst women. The situation is particularly dire in low to lower middle-income countries. It continues to affect these countries due to poor vaccine coverage and screening. Cervical cancer is mostly detected in the advanced stages leading to poor outcomes. This review focuses on the progress made to date to improve early detection and targeted therapy using both circulating RNA. Vaccine has played a major role in cervical cancer control in vaccinated young woman in mainly developed countries yet in low-income countries with challenges of 3 dose vaccination affordability, cervical cancer continues to be the second most deadly amongst women. In this review, we show the progress made in reducing cervical cancer using vaccination that in combination with other treatments that might improve survival in cervical cancer. We further show with both miRNA and siRNA that targeted therapy and specific markers might be ideal for early detection of cervical cancer in low-income countries. These markers are either upregulated or down regulated in cancer providing clue to the stage of the cancer.
Collapse
Affiliation(s)
- Xolisiwe M Sebutsoe
- Department of Biochemistry C2 Lab, University of Johannesburg, Auckland Park Kingsway Campus, Johannesburg, South Africa
| | | | - Zodwa Edith Jantjies
- Department of Biochemistry C2 Lab, University of Johannesburg, Auckland Park Kingsway Campus, Johannesburg, South Africa
| | - Portia Pheladi Raphela-Choma
- Department of Biochemistry C2 Lab, University of Johannesburg, Auckland Park Kingsway Campus, Johannesburg, South Africa
| | - Mpho S Choene
- Department of Biochemistry C2 Lab, University of Johannesburg, Auckland Park Kingsway Campus, Johannesburg, South Africa
| | - Lesetja R Motadi
- Department of Biochemistry C2 Lab, University of Johannesburg, Auckland Park Kingsway Campus, Johannesburg, South Africa
| |
Collapse
|
23
|
Casas-Arozamena C, Vilar A, Cueva J, Arias E, Sampayo V, Diaz E, Oltra SS, Moiola CP, Cabrera S, Cortegoso A, Curiel T, Abalo A, Pamies Serrano M, Domingo S, Padilla-Iserte P, Arnaez de la Cruz M, Hernández A, García-Pineda V, Ruiz-Bañobre J, López R, Matias-Guiu X, Colás E, Gil-Moreno A, Abal M, Moreno-Bueno G, Muinelo-Romay L. Role of cfDNA and ctDNA to improve the risk stratification and the disease follow-up in patients with endometrial cancer: towards the clinical application. J Exp Clin Cancer Res 2024; 43:264. [PMID: 39304963 DOI: 10.1186/s13046-024-03158-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 08/08/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND There has been a rise in endometrial cancer (EC) incidence leading to increased mortality. To counter this trend, improving the stratification of post-surgery recurrence risk and anticipating disease relapse and treatment resistance is essential. Liquid biopsy analyses offer a promising tool for these clinical challenges, though the best strategy for applying them in EC must be defined. This study was designed to determine the value of cfDNA/ctDNA monitoring in improving the clinical management of patients with localized and recurrent disease. METHODS Plasma samples and uterine aspirates (UA) from 198 EC patients were collected at surgery and over time. The genetic landscape of UAs was characterized using targeted sequencing. Total cfDNA was analyzed for ctDNA presence based on the UA mutational profile. RESULTS High cfDNA levels and detectable ctDNA at baseline correlated with poor prognosis for DFS (p-value < 0.0001; HR = 9.25) and DSS (p-value < 0.0001; HR = 11.20). This remained clinically significant when stratifying tumors by histopathological risk factors. Of note, cfDNA/ctDNA analyses discriminated patients with early post-surgery relapse and the ctDNA kinetics served to identify patients undergoing relapse before any clinical evidence emerged. CONCLUSIONS This is the most comprehensive study on cfDNA/ctDNA characterization in EC, demonstrating its value in improving risk stratification and anticipating disease relapse in patients with localized disease. CtDNA kinetics assessment complements current strategies to monitor the disease evolution and the treatment response. Therefore, implementing cfDNA/ctDNA monitoring in clinical routines offers a unique opportunity to improve EC management. TRANSLATIONAL RELEVANCE The study demonstrates that high levels of cfDNA and detectable ctDNA at baseline are strong indicators of poor prognosis. This enables more accurate risk stratification beyond traditional histopathological factors, allowing clinicians to identify high-risk patients who may benefit from more aggressive treatment and closer monitoring. Moreover, longitudinal analysis of cfDNA/ctDNA can detect disease recurrence months before clinical symptoms or imaging evidence appear. This early warning system offers a significant advantage in clinical practice, providing a window of opportunity for early intervention and potentially improving patient outcomes.
Collapse
Affiliation(s)
- Carlos Casas-Arozamena
- Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago de Compostela (IDIS), University Hospital of Santiago de Compostela (SERGAS), Trav. Choupana s/n, Santiago de Compostela, 15706, Spain
- University of Santiago de Compostela (USC), Praza do Obradoiro, 0, Santiago de Compostela, 15705, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Ana Vilar
- Department of Gynecology, University Hospital of Santiago de Compostela (SERGAS), Trav. Choupana s/n, Santiago de Compostela, 15706, Spain
| | - Juan Cueva
- Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago de Compostela (IDIS), University Hospital of Santiago de Compostela (SERGAS), Trav. Choupana s/n, Santiago de Compostela, 15706, Spain
- Department of Medical Oncology, University Clinical Hospital of Santiago de Compostela, University of Santiago de Compostela (USC), Santiago de Compostela, 15706, Spain
| | - Efigenia Arias
- Department of Gynecology, University Hospital of Santiago de Compostela (SERGAS), Trav. Choupana s/n, Santiago de Compostela, 15706, Spain
| | - Victoria Sampayo
- Department of Gynecology, University Hospital of Santiago de Compostela (SERGAS), Trav. Choupana s/n, Santiago de Compostela, 15706, Spain
| | - Eva Diaz
- MD Anderson Cancer Center Foundation, C/Gómez Hemans 2, Madrid, 28033, Spain
| | - Sara S Oltra
- MD Anderson Cancer Center Foundation, C/Gómez Hemans 2, Madrid, 28033, Spain
| | - Cristian Pablo Moiola
- Department of Gynecologic Oncology, Biomedical Research Group in Gynecology, Vall Hebron Research Institute (VHIR), Universitat Autónoma de Barcelona, 119-129 Pg. Vall d'Hebron, Barcelona, 08035, Spain
- Department of Basic Sciences, Faculty of Health Sciences at Manresa, University of Vic - Central University of Catalonia (UVic-UCC), Av. Universitària, 4-6, Manresa, 08242, Spain
| | - Silvia Cabrera
- Department of Gynecologic Oncology, Biomedical Research Group in Gynecology, Vall Hebron Research Institute (VHIR), Universitat Autónoma de Barcelona, 119-129 Pg. Vall d'Hebron, Barcelona, 08035, Spain
| | - Alexandra Cortegoso
- Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago de Compostela (IDIS), University Hospital of Santiago de Compostela (SERGAS), Trav. Choupana s/n, Santiago de Compostela, 15706, Spain
- Department of Medical Oncology, University Clinical Hospital of Santiago de Compostela, University of Santiago de Compostela (USC), Santiago de Compostela, 15706, Spain
| | - Teresa Curiel
- Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago de Compostela (IDIS), University Hospital of Santiago de Compostela (SERGAS), Trav. Choupana s/n, Santiago de Compostela, 15706, Spain
- Department of Medical Oncology, University Clinical Hospital of Santiago de Compostela, University of Santiago de Compostela (USC), Santiago de Compostela, 15706, Spain
| | - Alicia Abalo
- Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago de Compostela (IDIS), University Hospital of Santiago de Compostela (SERGAS), Trav. Choupana s/n, Santiago de Compostela, 15706, Spain
| | - Mónica Pamies Serrano
- Department of Gynecologic Oncology, Biomedical Research Group in Gynecology, Vall Hebron Research Institute (VHIR), Universitat Autónoma de Barcelona, 119-129 Pg. Vall d'Hebron, Barcelona, 08035, Spain
| | - Santiago Domingo
- Department of Gynecologic Oncology, La Fe University and Polytechnic Hospital, Valencia, Spain
| | - Pablo Padilla-Iserte
- Department of Gynecologic Oncology, La Fe University and Polytechnic Hospital, Valencia, Spain
| | - Marta Arnaez de la Cruz
- Department of Gynecologic Oncology, La Fe University and Polytechnic Hospital, Valencia, Spain
| | - Alicia Hernández
- Department of Gynecology, University Hospital "La Paz", Madrid, Spain
| | | | - Juan Ruiz-Bañobre
- Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago de Compostela (IDIS), University Hospital of Santiago de Compostela (SERGAS), Trav. Choupana s/n, Santiago de Compostela, 15706, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Department of Medical Oncology, University Clinical Hospital of Santiago de Compostela, University of Santiago de Compostela (USC), Santiago de Compostela, 15706, Spain
| | - Rafael López
- Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago de Compostela (IDIS), University Hospital of Santiago de Compostela (SERGAS), Trav. Choupana s/n, Santiago de Compostela, 15706, Spain
- University of Santiago de Compostela (USC), Praza do Obradoiro, 0, Santiago de Compostela, 15705, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Department of Medical Oncology, University Clinical Hospital of Santiago de Compostela, University of Santiago de Compostela (USC), Santiago de Compostela, 15706, Spain
| | - Xavier Matias-Guiu
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Department of Pathology, Instituto de Investigación Biomédica de Bellvitge, Centro de Investigación Biomédica en Red de Cáncer, Hospital U de Bellvitge and Hospital U Arnau de Vilanova, Universities of Lleida and Barcelona, Institut de Recerca Biomèdica de Lleida, Barcelona, Spain
| | - Eva Colás
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Department of Gynecologic Oncology, Biomedical Research Group in Gynecology, Vall Hebron Research Institute (VHIR), Universitat Autónoma de Barcelona, 119-129 Pg. Vall d'Hebron, Barcelona, 08035, Spain
| | - Antonio Gil-Moreno
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Department of Gynecologic Oncology, Biomedical Research Group in Gynecology, Vall Hebron Research Institute (VHIR), Universitat Autónoma de Barcelona, 119-129 Pg. Vall d'Hebron, Barcelona, 08035, Spain
| | - Miguel Abal
- Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago de Compostela (IDIS), University Hospital of Santiago de Compostela (SERGAS), Trav. Choupana s/n, Santiago de Compostela, 15706, Spain
- University of Santiago de Compostela (USC), Praza do Obradoiro, 0, Santiago de Compostela, 15705, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Gema Moreno-Bueno
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.
- MD Anderson Cancer Center Foundation, C/Gómez Hemans 2, Madrid, 28033, Spain.
- Instituto de Investigaciones Biomedicas "Sols-Morreale" CSIC-UAM, C/Arturo Dupurier 4, Madrid, 28029, Spain.
| | - Laura Muinelo-Romay
- Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago de Compostela (IDIS), University Hospital of Santiago de Compostela (SERGAS), Trav. Choupana s/n, Santiago de Compostela, 15706, Spain.
- University of Santiago de Compostela (USC), Praza do Obradoiro, 0, Santiago de Compostela, 15705, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.
| |
Collapse
|
24
|
Mauri G, Patelli G, Sartore-Bianchi A, Abrignani S, Bodega B, Marsoni S, Costanzo V, Bachi A, Siena S, Bardelli A. Early-onset cancers: Biological bases and clinical implications. Cell Rep Med 2024; 5:101737. [PMID: 39260369 PMCID: PMC11525030 DOI: 10.1016/j.xcrm.2024.101737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/02/2024] [Accepted: 08/19/2024] [Indexed: 09/13/2024]
Abstract
Since the nineties, the incidence of sporadic early-onset (EO) cancers has been rising worldwide. The underlying reasons are still unknown. However, identifying them is vital for advancing both prevention and intervention. Here, we exploit available knowledge derived from clinical observations to formulate testable hypotheses aimed at defining the causal factors of this epidemic and discuss how to experimentally test them. We explore the potential impact of exposome changes from the millennials to contemporary young generations, considering both environmental exposures and enhanced susceptibilities to EO-cancer development. We emphasize how establishing the time required for an EO cancer to develop is relevant to defining future screening strategies. Finally, we discuss the importance of integrating multi-dimensional data from international collaborations to generate comprehensive knowledge and translate these findings back into clinical practice.
Collapse
Affiliation(s)
- Gianluca Mauri
- IFOM ETS - The AIRC Institute of Molecular Oncology, Milan, Italy; Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy; Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Giorgio Patelli
- IFOM ETS - The AIRC Institute of Molecular Oncology, Milan, Italy; Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy; Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Andrea Sartore-Bianchi
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy; Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Sergio Abrignani
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy; Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Beatrice Bodega
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy; Department of Biosciences, University of Milan, Milan, Italy
| | - Silvia Marsoni
- IFOM ETS - The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Vincenzo Costanzo
- IFOM ETS - The AIRC Institute of Molecular Oncology, Milan, Italy; Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Angela Bachi
- IFOM ETS - The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Salvatore Siena
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy; Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Alberto Bardelli
- IFOM ETS - The AIRC Institute of Molecular Oncology, Milan, Italy; Department of Oncology, Molecular Biotechnology Center, University of Torino, Torino, Italy.
| |
Collapse
|
25
|
Li J, Yang B, Teng Z, Liu Y, Li D, Qu X. Efficacy and safety of first-line treatments for advanced hepatocellular carcinoma patients: a systematic review and network meta-analysis. Front Immunol 2024; 15:1430196. [PMID: 39355238 PMCID: PMC11442238 DOI: 10.3389/fimmu.2024.1430196] [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: 05/09/2024] [Accepted: 08/28/2024] [Indexed: 10/03/2024] Open
Abstract
Background The first-line treatment for advanced hepatocellular carcinoma has evolved significantly. This study aimed to identify the most beneficial regimen. Methods A systematic search was conducted from July 2012 to August 2024 across the following four databases: PubMed, Embase, Cochrane Library, and ClinicalTrials.gov. This search focused on phase III prospective randomized controlled trials that compared first-line treatment for advanced hepatocellular carcinoma. Results Seventeen studies involving 10322 patients were included in this network meta-analysis. Of the studies we included, twelve studies were global multicenter clinical studies, four were initiated in China, and one was initiated in Korea. The results of our statistical analysis suggest that Hepatic artery infusion chemotherapy with oxaliplatin plus fluorouracil (HAIC-FO) demonstrated significant overall survival (OS) benefits compared with most treatments, including various immune checkpoint inhibitors (ICIs) and anti-vascular endothelial growth factor tyrosine kinase inhibitors (VEGF-TKIs). In terms of OS, HAIC had shown similar efficacy with sorafenib plus FOLFOX (HR, 0.88; 95% CI: 0.37-2.09) and transcatheter arterial chemoembolization (TACE) combined with lenvatinib (HR, 0.69; 95% CI: 0.30-1.56). Notably, immune-related treatments, such as ICIs combined with anti-VEGF therapies, also showed improved OS compared with anti-VEGF-TKIs alone. In terms of progression-free survival (PFS), HAIC-FO outperformed anti-VEGF-TKI monotherapy, ICI monotherapy, and several ICI combinations. However, it was not superior to lenvatinib plus TACE or lenvatinib plus pembrolizumab. Based on the Surface Under the Cumulative Ranking Curve (SUCRA) values, HAIC-FO was ranked the most effective in terms of OS (SUCRA = 0.961) and objective response rate (ORR) (SUCRA = 0.971). The results of the subgroup analysis suggested that HAIC-FO achieved the best OS benefit in the macrovascular invasion (MVI) and extrahepatic spread (EHS) subgroup (SUCRA = 0.99) and that tremelimumab combined with durvalumab achieved the best OS benefit in the Asian subgroup (SUCRA = 0.88). Conclusion This systematic review and network meta-analysis suggest that HAIC-based therapies may become a potential first-line treatment option for advanced HCC, especially for patients in Mainland China with MVI and EHS. Additionally, immune-related treatments may be more suitable for Asian populations.
Collapse
Affiliation(s)
- Jingyi Li
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, the First Hospital of China Medical University, Shenyang, China
- Liaoning Province Clinical Research Center for Cancer, the First Hospital of China Medical University, Shenyang, China
- Clinical Cancer Treatment and Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, China
| | - Bowen Yang
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, the First Hospital of China Medical University, Shenyang, China
- Liaoning Province Clinical Research Center for Cancer, the First Hospital of China Medical University, Shenyang, China
- Clinical Cancer Treatment and Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, China
| | - Zan Teng
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, the First Hospital of China Medical University, Shenyang, China
- Liaoning Province Clinical Research Center for Cancer, the First Hospital of China Medical University, Shenyang, China
- Clinical Cancer Treatment and Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, China
| | - Yunpeng Liu
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, the First Hospital of China Medical University, Shenyang, China
- Liaoning Province Clinical Research Center for Cancer, the First Hospital of China Medical University, Shenyang, China
- Clinical Cancer Treatment and Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, China
| | - Danni Li
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, the First Hospital of China Medical University, Shenyang, China
- Liaoning Province Clinical Research Center for Cancer, the First Hospital of China Medical University, Shenyang, China
- Clinical Cancer Treatment and Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, China
| | - Xiujuan Qu
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, the First Hospital of China Medical University, Shenyang, China
- Liaoning Province Clinical Research Center for Cancer, the First Hospital of China Medical University, Shenyang, China
- Clinical Cancer Treatment and Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
26
|
Golara A, Kozłowski M, Cymbaluk-Płoska A. The Role of Circulating Tumor DNA in Ovarian Cancer. Cancers (Basel) 2024; 16:3117. [PMID: 39335089 PMCID: PMC11430586 DOI: 10.3390/cancers16183117] [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: 08/03/2024] [Revised: 09/05/2024] [Accepted: 09/08/2024] [Indexed: 09/30/2024] Open
Abstract
Ovarian cancer is the deadliest of all gynecological diseases because its diagnosis and treatment still pose many problems. Surgical excision, hormone therapy, radiation, chemotherapy, or targeted therapy for eradicating the main tumor and halting the spread of metastases are among the treatment options available to individuals with ovarian cancer, depending on the disease's stage. Tumor DNA that circulates in a patient's bodily fluids has been studied recently as a possible novel biomarker for a number of cancers, as well as a means of quantifying tumor size and evaluating the efficacy of cancer therapy. The most significant alterations that we could find in the ctDNA of ovarian cancer patients-such as chromosomal instability, somatic mutations, and methylation-are discussed in this review. Additionally, we talk about the utility of ctDNA in diagnosis, prognosis, and therapy response prediction for these patients.
Collapse
Affiliation(s)
- Anna Golara
- Department of Reconstructive Surgery and Gynecological Oncology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland;
| | - Mateusz Kozłowski
- Department of Reconstructive Surgery and Gynecological Oncology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland;
| | | |
Collapse
|
27
|
He W, Huang W, Zhang L, Wu X, Zhang S, Zhang B. Radiogenomics: bridging the gap between imaging and genomics for precision oncology. MedComm (Beijing) 2024; 5:e722. [PMID: 39252824 PMCID: PMC11381657 DOI: 10.1002/mco2.722] [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: 03/23/2024] [Revised: 08/06/2024] [Accepted: 08/18/2024] [Indexed: 09/11/2024] Open
Abstract
Genomics allows the tracing of origin and evolution of cancer at molecular scale and underpin modern cancer diagnosis and treatment systems. Yet, molecular biomarker-guided clinical decision-making encounters major challenges in the realm of individualized medicine, consisting of the invasiveness of procedures and the sampling errors due to high tumor heterogeneity. By contrast, medical imaging enables noninvasive and global characterization of tumors at a low cost. In recent years, radiomics has overcomes the limitations of human visual evaluation by high-throughput quantitative analysis, enabling the comprehensive utilization of the vast amount of information underlying radiological images. The cross-scale integration of radiomics and genomics (hereafter radiogenomics) has the enormous potential to enhance cancer decoding and act as a catalyst for digital precision medicine. Herein, we provide a comprehensive overview of the current framework and potential clinical applications of radiogenomics in patient care. We also highlight recent research advances to illustrate how radiogenomics can address common clinical problems in solid tumors such as breast cancer, lung cancer, and glioma. Finally, we analyze existing literature to outline challenges and propose solutions, while also identifying future research pathways. We believe that the perspectives shared in this survey will provide a valuable guide for researchers in the realm of radiogenomics aiming to advance precision oncology.
Collapse
Affiliation(s)
- Wenle He
- Department of Radiology The First Affiliated Hospital of Jinan University Guangzhou Guangdong China
| | - Wenhui Huang
- Department of Radiology The First Affiliated Hospital of Jinan University Guangzhou Guangdong China
| | - Lu Zhang
- Department of Radiology The First Affiliated Hospital of Jinan University Guangzhou Guangdong China
| | - Xuewei Wu
- Department of Radiology The First Affiliated Hospital of Jinan University Guangzhou Guangdong China
| | - Shuixing Zhang
- Department of Radiology The First Affiliated Hospital of Jinan University Guangzhou Guangdong China
| | - Bin Zhang
- Department of Radiology The First Affiliated Hospital of Jinan University Guangzhou Guangdong China
| |
Collapse
|
28
|
Francis JH, Arcila ME, Sigler A, Bossert DF, Abramson DH, Diamond EL. Plasma-Derived Cell-Free DNA for the Diagnosis of Ocular-Involving Histiocytosis. OPHTHALMOLOGY SCIENCE 2024; 4:100530. [PMID: 39071917 PMCID: PMC11283146 DOI: 10.1016/j.xops.2024.100530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/04/2024] [Accepted: 04/08/2024] [Indexed: 07/30/2024]
Abstract
Purpose Circulating tumor DNA (ctDNA) is released into the plasma by many cancers and offers clinical applications including noninvasive diagnostics. Histiocytosis results from myelogenous clonal expansion of histiocytes, predominantly driven by mutations in the mitogen-activated protein kinase pathway that are potentially detectable by ctDNA-based sequencing assays. However, ocular-involving histiocytosis is often a diagnostic challenge leading to delayed diagnosis and the need for invasive biopsy of sensitive ocular structures. The purpose of this study is to determine whether sequencing of plasma-derived ctDNA can noninvasively diagnose ocular-involving histiocytosis. Design Single tertiary cancer referral center. Participants Twenty-four adult patients with ocular-involving histiocytosis and ctDNA sequencing. Methods Circulating tumor DNA was analyzed (via digital droplet polymerase chain reaction for BRAF V600E, and/or next-generation sequencing) and variant allele frequency was measured at initial presentation to our center. Patient demographics, clinical characteristics, and oncogenic mutations identified from tumor-based sequencing were recorded. Main Outcome Measures Plasma-derived ctDNA detectability of pertinent driver mutations of histiocytosis. Results At the initial presentation of 14 patients with ocular-involving histiocytosis, sequencing of plasma-derived ctDNA detected driver mutations for histiocytosis (BRAF V600E [10], KRAS [2], ARAF [1], and concurrent MAP2K1/KRAS [1]). Mutations found in circulating cell-free DNA were 100% concordant in 11 of 11 patients with mutations identified by solid tumor sequencing. Of 10 patients without driver mutation detected in ctDNA, 3 patients had alterations (CBL mutation or kinase fusion) not captured in the ctDNA sequencing assay, 3 were wildtype even by tumor sequencing; in 4 patients, tumor-based sequencing identified mutations (BRAF [2], MAP2K1 [2]) not detected in ctDNA. Detectable mutations in ctDNA were significantly more likely in patients with uveal infiltration (P = 0.036). Conclusions In this cohort, plasma-derived ctDNA was detectable and diagnostic in the majority of patients with ocular-involving histiocytosis. This suggests that if ocular histiocytosis is suspected (particularly if involving the uvea), noninvasive plasma-derived ctDNA analysis is a helpful diagnostic tool that may obviate the need to invasively biopsy sensitive ocular structures. Financial Disclosures Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.
Collapse
Affiliation(s)
- Jasmine H. Francis
- Ophthalmic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Ophthalmology, Weill Cornell Medical College, New York, New York
| | - Maria E. Arcila
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Allison Sigler
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Dana F. Bossert
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David H. Abramson
- Ophthalmic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Ophthalmology, Weill Cornell Medical College, New York, New York
| | - Eli L. Diamond
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York
- Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
29
|
Ruglioni M, Crucitta S, Luculli GI, Tancredi G, Del Giudice ML, Mechelli S, Galimberti S, Danesi R, Del Re M. Understanding mechanisms of resistance to FLT3 inhibitors in adult FLT3-mutated acute myeloid leukemia to guide treatment strategy. Crit Rev Oncol Hematol 2024; 201:104424. [PMID: 38917943 DOI: 10.1016/j.critrevonc.2024.104424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/06/2024] [Accepted: 06/16/2024] [Indexed: 06/27/2024] Open
Abstract
The presence of FLT3 mutations, including the most common FLT3-ITD (internal tandem duplications) and FLT3-TKD (tyrosine kinase domain), is associated with an unfavorable prognosis in patients affected by acute myeloid leukemia (AML). In this setting, in recent years, new FLT3 inhibitors have demonstrated efficacy in improving survival and treatment response. Nevertheless, the development of primary and secondary mechanisms of resistance poses a significant obstacle to their efficacy. Understanding these mechanisms is crucial for developing novel therapeutic approaches to overcome resistance and improve the outcomes of patients. In this context, the use of novel FLT3 inhibitors and the combination of different targeted therapies have been studied. This review provides an update on the molecular alterations involved in the resistance to FLT3 inhibitors, and describes how the molecular monitoring may be used to guide treatment strategy in FLT3-mutated AML.
Collapse
Affiliation(s)
- Martina Ruglioni
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Stefania Crucitta
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Giovanna Irene Luculli
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Gaspare Tancredi
- Unit of Hematology, Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Maria Livia Del Giudice
- Unit of Hematology, Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Sandra Mechelli
- Unit of Internal Medicine 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Sara Galimberti
- Unit of Hematology, Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Romano Danesi
- Department of Oncology and Hemato-Oncology, University of Milan, Italy.
| | - Marzia Del Re
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Italy
| |
Collapse
|
30
|
Huang G, Zheng W, Zhou Y, Wan M, Hu T. Recent advances to address challenges in extracellular vesicle-based applications for lung cancer. Acta Pharm Sin B 2024; 14:3855-3875. [PMID: 39309489 PMCID: PMC11413688 DOI: 10.1016/j.apsb.2024.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 05/14/2024] [Accepted: 05/28/2024] [Indexed: 09/25/2024] Open
Abstract
Lung cancer, highly prevalent and the leading cause of cancer-related death globally, persists as a significant challenge due to the lack of definitive tumor markers for early diagnosis and personalized therapeutic interventions. Recently, extracellular vesicles (EVs), functioning as natural carriers for intercellular communication, have received increasing attention due to their ability to traverse biological barriers and deliver diverse biological cargoes, including cytosolic proteins, cell surface proteins, microRNA, lncRNA, circRNA, DNA, and lipids. EVs are increasingly recognized as a valuable resource for non-invasive liquid biopsy, as well as drug delivery platforms, and anticancer vaccines for precision medicine in lung cancer. Herein, given the diagnostic and therapeutic potential of tumor-associated EVs for lung cancer, we discuss this topic from a translational standpoint. We delve into the specific roles that EVs play in lung cancer carcinogenesis and offer a particular perspective on how advanced engineering technologies can overcome the current challenges and expedite and/or enhance the translation of EVs from laboratory research to clinical settings.
Collapse
Affiliation(s)
- Gaigai Huang
- Department of Clinical Laboratory, the First People's Hospital of Shuangliu District (West China Airport Hospital of Sichuan University), Chengdu 610200, China
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Wenshu Zheng
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Yu Zhou
- Department of Clinical Laboratory, the First People's Hospital of Shuangliu District (West China Airport Hospital of Sichuan University), Chengdu 610200, China
| | - Meihua Wan
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu 610200, China
- The First People's Hospital of Shuangliu District (West China Airport Hospital of Sichuan University), Chengdu 610200, China
| | - Tony Hu
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
31
|
Nesic M, Rasmussen MH, Henriksen TV, Demuth C, Frydendahl A, Nordentoft I, Dyrskjøt L, Andersen CL. Beyond basics: Key mutation selection features for successful tumor-informed ctDNA detection. Int J Cancer 2024; 155:925-933. [PMID: 38623608 DOI: 10.1002/ijc.34964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/26/2024] [Accepted: 03/28/2024] [Indexed: 04/17/2024]
Abstract
Tumor-informed mutation-based approaches are frequently used for detection of circulating tumor DNA (ctDNA). Not all mutations make equally effective ctDNA markers. The objective was to explore if prioritizing mutations using mutational features-such as cancer cell fraction (CCF), multiplicity, and error rate-would improve the success rate of tumor-informed ctDNA analysis. Additionally, we aimed to develop a practical and easily implementable analysis pipeline for identifying and prioritizing candidate mutations from whole-exome sequencing (WES) data. We analyzed WES and ctDNA data from three tumor-informed ctDNA studies, one on bladder cancer (Cohort A) and two on colorectal cancer (Cohorts I and N). The studies included 390 patients. For each patient, a unique set of mutations (median mutations/patient: 6, interquartile 13, range: 1-46, total n = 4023) were used as markers of ctDNA. The tool PureCN was used to assess the CCF and multiplicity of each mutation. High-CCF mutations were detected more frequently than low-CCF mutations (Cohort A: odds ratio [OR] 20.6, 95% confidence interval [CI] 5.72-173, p = 1.73e-12; Cohort I: OR 2.24, 95% CI 1.44-3.52, p = 1.66e-04; and Cohort N: OR 1.78, 95% CI 1.14-2.79, p = 7.86e-03). The detection-likelihood was additionally improved by selecting mutations with multiplicity of two or above (Cohort A: OR 1.55, 95% CI 1. 14-2.11, p = 3.85e-03; Cohort I: OR 1.78, 95% CI 1.23-2.56, p = 1.34e-03; and Cohort N: OR 1.94, 95% CI 1.63-2.31, p = 2.83e-14). Furthermore, selecting the mutations for which the ctDNA detection method had the lowest error rates, additionally improved the detection-likelihood, particularly evident when plasma cell-free DNA tumor fractions were below 0.1% (p = 2.1e-07). Selecting mutational markers with high CCF, high multiplicity, and low error rate significantly improve ctDNA detection likelihood. We provide free access to the analysis pipeline enabling others to perform qualified prioritization of mutations for tumor-informed ctDNA analysis.
Collapse
Affiliation(s)
- Marijana Nesic
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Mads H Rasmussen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Tenna V Henriksen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Christina Demuth
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Amanda Frydendahl
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Iver Nordentoft
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Lars Dyrskjøt
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Claus L Andersen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
32
|
Gristina V, Pepe F, Genova C, Bazan Russo TD, Gottardo A, Russo G, Incorvaia L, Galvano A, Badalamenti G, Bazan V, Troncone G, Russo A, Malapelle U. Harnessing the potential of genomic characterization of mutational profiles to improve early diagnosis of lung cancer. Expert Rev Mol Diagn 2024; 24:793-802. [PMID: 39267426 DOI: 10.1080/14737159.2024.2403081] [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: 05/21/2024] [Accepted: 09/08/2024] [Indexed: 09/17/2024]
Abstract
INTRODUCTION Lung Cancer (LC) continues to be a leading cause of cancer-related mortality globally, largely due to the asymptomatic nature of its early stages and the limitations of current diagnostic methods such as Low-Dose Computed Tomography (LDCT), whose often result in late diagnosis, highlighting an urgent need for innovative, minimally invasive diagnostic techniques that can improve early detection rates. AREAS COVERED This review delves into the potential of genomic characterization and mutational profiling to enhance early LC diagnosis, exploring the current state and limitations of traditional diagnostic approaches and the revolutionary role of Liquid Biopsies (LB), including cell-free DNA (cfDNA) analysis through fragmentomics and methylomics. New genomic technologies that allow for earlier detection of LC are scrutinized, alongside a detailed discussion on the literature that shaped our understanding in this field. EXPERT OPINION Despite the promising advancements in genomic characterization techniques, several challenges remain, such as the heterogeneity of LC mutations, the high cost, and limited accessibility of Next-Generation Sequencing (NGS) technologies. Additionally, there is a critical need of standardized protocols for interpreting mutational data. Future research should focus on overcoming these barriers to integrate these novel diagnostic methods into standard clinical practice, potentially revolutionizing the management of LC patients.
Collapse
Affiliation(s)
- Valerio Gristina
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Francesco Pepe
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Carlo Genova
- Department of Internal Medicine and Medical Specialties, University of Genoa, Genoa, Italy
- Academic Oncology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Tancredi Didier Bazan Russo
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Andrea Gottardo
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Gianluca Russo
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Lorena Incorvaia
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Antonio Galvano
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Giuseppe Badalamenti
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Viviana Bazan
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, Palermo, Italy
| | - Giancarlo Troncone
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Antonio Russo
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Naples, Italy
| |
Collapse
|
33
|
Zhao J, Ma Y, Zheng X, Sun Z, Lin H, Du C, Cao J. Bladder cancer: non-coding RNAs and exosomal non-coding RNAs. Funct Integr Genomics 2024; 24:147. [PMID: 39217254 DOI: 10.1007/s10142-024-01433-9] [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: 07/28/2024] [Revised: 08/15/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Bladder cancer (BCa) is a highly prevalent type of cancer worldwide, and it is responsible for numerous deaths and cases of disease. Due to the diverse nature of this disease, it is necessary to conduct significant research that delves deeper into the molecular aspects, to potentially discover novel diagnostic and therapeutic approaches. Lately, there has been a significant increase in the focus on non-coding RNAs (ncRNAs), including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), due to their growing recognition for their involvement in the progression and manifestation of BCa. The interest in exosomes has greatly grown due to their potential for transporting a diverse array of active substances, including proteins, nucleic acids, carbohydrates, and lipids. The combination of these components differs based on the specific cell and its condition. Research indicates that using exosomes could have considerable advantages in identifying and forecasting BCa, offering a less invasive alternative. The distinctive arrangement of the lipid bilayer membrane found in exosomes is what makes them particularly effective for administering treatments aimed at managing cancer. In this review, we have tried to summarize different ncRNAs that are involved in BCa pathogenesis. Moreover, we highlighted the role of exosomal ncRNAs in BCa.
Collapse
Affiliation(s)
- Jingang Zhao
- Department of Urology, Hangzhou Mingzhou Hospital, Hangzhou, 311215, Zhe'jiang, China
| | - Yangyang Ma
- Department of Urology, Hangzhou Mingzhou Hospital, Hangzhou, 311215, Zhe'jiang, China
| | - Xiaodong Zheng
- Department of the First Surgery, Zhejiang Provincial Corps Hospital of Chinese People's Armed Police Force, Hangzhou, 310051, Zhe'jiang, China
| | - Zhen Sun
- Department of the First Surgery, Zhejiang Provincial Corps Hospital of Chinese People's Armed Police Force, Hangzhou, 310051, Zhe'jiang, China
| | - Hongxiang Lin
- Department of Urology, Ganzhou Donghe Hospital, Ganzhou, 341000, Jiang'xi, China
| | - Chuanjun Du
- Department of Urology, Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, 310009, Zhe'jiang, China
| | - Jing Cao
- Department of Urology, Hangzhou Mingzhou Hospital, Hangzhou, 311215, Zhe'jiang, China.
| |
Collapse
|
34
|
Singh J, Peters NJ, Avti P, Trehan A, Mahajan JK, Menon P, Bansal D, Kanojia RP. The Role of Liquid Biopsy in Neuroblastoma: A Scoping Review. J Pediatr Surg 2024:161887. [PMID: 39294087 DOI: 10.1016/j.jpedsurg.2024.161887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/16/2024] [Accepted: 08/27/2024] [Indexed: 09/20/2024]
Abstract
BACKGROUND Neuroblastoma (NBL), is the most common, non-CNS solid tumor of childhood. This disease presents with unique biological and clinical challenges necessitating accurate diagnosis, prognosis assessment, treatment, and vigilant monitoring. Liquid biopsy is an upcoming, innovative, and non-invasive diagnostic modality. It has the potential to detect tumors and perform therapeutic monitoring through the analysis of circulating biomarkers in blood, urine, saliva, and other bodily fluids. METHODOLOGY This scoping review offers an in-depth exploration, of the current landscape of liquid biopsy-based biomarkers in NBL. The review looks at the clinical implications, prevalent challenges, and future outlook of their clinical applications in NBL. The scoping review adhered to the guidelines of the PRISMA extension for scoping reviews, known as PRISMA-ScR, as the skeletal framework. The review involved comprehensive searches for liquid biopsy-based biomarkers in NBL across multiple databases, including PUBMED, EMBASE, SCOPUS, and WEB of Science, without restrictions. RESULTS The scoping review process uncovered a significant body of literature (n = 201) that underwent meticulous scrutiny, ultimately leading to the final selection of studies (n = 15). The liquid biopsy biomarkers included circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), exosomes, and other entities in bodily fluids. Their evaluation focused on associations with clinical outcomes such as overall survival, event-free survival, and risk stratification in NBL. CONCLUSION Our findings highlight the potential of liquid biopsy biomarkers to revolutionize NBL diagnosis and therapeutic monitoring. This rapidly evolving frontier in pediatric oncology suggests significant advancements in precision medicine for the management of NBL.
Collapse
Affiliation(s)
- Jitender Singh
- Department of Pediatric Surgery, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Nitin J Peters
- Department of Pediatric Surgery, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India.
| | - Pramod Avti
- Department of Biophysics, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Amita Trehan
- Pediatric Oncology Unit, Department of Paediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - J K Mahajan
- Department of Pediatric Surgery, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Prema Menon
- Department of Pediatric Surgery, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Deepak Bansal
- Pediatric Oncology Unit, Department of Paediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Ravi Prakash Kanojia
- Department of Pediatric Surgery, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| |
Collapse
|
35
|
Rocchetti F, Tenore G, Macali F, Vicidomini T, Podda GM, Fantozzi PJ, Silvestri V, Porzio V, Valentini V, Ottini L, Richetta AG, Valentini V, Della Monaca M, Grenga C, Polimeni A, Romeo U. Expression Analysis of Circulating microRNAs in Saliva and Plasma for the Identification of Clinically Relevant Biomarkers for Oral Squamous Cell Carcinoma and Oral Potentially Malignant Disorders. Cancers (Basel) 2024; 16:2990. [PMID: 39272848 PMCID: PMC11394426 DOI: 10.3390/cancers16172990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/10/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
This study aims to evaluate the expression of salivary and plasmatic miRNAs as diagnostic biomarkers in patients with oral squamous cell carcinoma (OSCC) and oral potentially malignant disorders (OPMDs). A total of 25 patients were divided into three groups, according to their diagnosis: OSCC patients (n = 14); OPMDs patients (n = 6); and healthy controls (n = 5). At the time at diagnosis/enrolment, patients underwent salivary and plasmatic collection. The expression of miRNA -21, -31, -138, -145, -184, and -424 were evaluated by real-time PCR. An F-test and ANOVA test were performed to evaluate the miRNA levels (significance at p < 0.05). By comparing miRNA expression levels from saliva, a statistically significant difference emerged in the expression of miR-138 and miR-424 between the three groups (p < 0.05). In particular, these two miRNAs showed decreased expression levels in saliva samples from OSCC and OPMD patients compared to those from healthy controls. On the other hand, miRNA expression levels in plasma were low in all the groups, and no statistically significant differences were found. Overall, our results showed that liquid biopsy from saliva may be a useful tool for the identification of diagnostic molecular biomarkers in OSCC and OPMDs.
Collapse
Affiliation(s)
- Federica Rocchetti
- Department of Oral and Maxillofacial Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Gianluca Tenore
- Department of Oral and Maxillofacial Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Federica Macali
- Department of Oral and Maxillofacial Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Teresa Vicidomini
- Department of Oral and Maxillofacial Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Gian Marco Podda
- Department of Oral and Maxillofacial Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Paolo Junior Fantozzi
- Department of Oral and Maxillofacial Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Valentina Silvestri
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Virginia Porzio
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Virginia Valentini
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Laura Ottini
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Antonio Giovanni Richetta
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, 00161 Rome, Italy
| | - Valentino Valentini
- Department of Oral and Maxillofacial Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Marco Della Monaca
- Department of Oral and Maxillofacial Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Camilla Grenga
- Department of Oral and Maxillofacial Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Antonella Polimeni
- Department of Oral and Maxillofacial Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Umberto Romeo
- Department of Oral and Maxillofacial Sciences, Sapienza University of Rome, 00161 Rome, Italy
| |
Collapse
|
36
|
Karp JM, Modrek AS, Ezhilarasan R, Zhang ZY, Ding Y, Graciani M, Sahimi A, Silvestro M, Chen T, Li S, Wong KK, Ramkhelawon B, Bhat KP, Sulman EP. Deconvolution of the tumor-educated platelet transcriptome reveals activated platelet and inflammatory cell transcript signatures. JCI Insight 2024; 9:e178719. [PMID: 39190500 PMCID: PMC11466191 DOI: 10.1172/jci.insight.178719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 08/13/2024] [Indexed: 08/29/2024] Open
Abstract
Tumor-educated platelets (TEPs) are a potential method of liquid biopsy for the diagnosis and monitoring of cancer. However, the mechanism underlying tumor education of platelets is not known, and transcripts associated with TEPs are often not tumor-associated transcripts. We demonstrated that direct tumor transfer of transcripts to circulating platelets is an unlikely source of the TEP signal. We used CDSeq, a latent Dirichlet allocation algorithm, to deconvolute the TEP signal in blood samples from patients with glioblastoma. We demonstrated that a substantial proportion of transcripts in the platelet transcriptome are derived from nonplatelet cells, and the use of this algorithm allows the removal of contaminant transcripts. Furthermore, we used the results of this algorithm to demonstrate that TEPs represent a subset of more activated platelets, which also contain transcripts normally associated with nonplatelet inflammatory cells, suggesting that these inflammatory cells, possibly in the tumor microenvironment, transfer transcripts to platelets that are then found in circulation. Our analysis suggests a useful and efficient method of processing TEP transcriptomic data to enable the isolation of a unique TEP signal associated with specific tumors.
Collapse
Affiliation(s)
- Jerome M. Karp
- Department of Radiation Oncology, NYU Grossman School of Medicine, New York, New York, USA
| | - Aram S. Modrek
- Department of Radiation Oncology, Keck School of Medicine of University of Southern California, Los Angeles, California, USA
| | - Ravesanker Ezhilarasan
- Department of Radiation Oncology, NYU Grossman School of Medicine, New York, New York, USA
| | - Ze-Yan Zhang
- Department of Radiation Oncology, NYU Grossman School of Medicine, New York, New York, USA
| | - Yingwen Ding
- Department of Radiation Oncology, NYU Grossman School of Medicine, New York, New York, USA
| | - Melanie Graciani
- Department of Radiation Oncology, NYU Grossman School of Medicine, New York, New York, USA
| | - Ali Sahimi
- Department of Radiation Oncology, NYU Grossman School of Medicine, New York, New York, USA
| | | | - Ting Chen
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, New York, USA
| | - Shuai Li
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, New York, USA
| | - Kwok-Kin Wong
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, New York, USA
| | | | | | - Erik P. Sulman
- Department of Radiation Oncology, NYU Grossman School of Medicine, New York, New York, USA
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
37
|
Lobo-Martins S, Corredeira P, Cavaco A, Rodrigues C, Piairo P, Lopes C, Fraga J, Silva M, Alves P, Wachholz Szeneszi L, Barradas A, Castro Duran C, Antunes M, Nogueira-Costa G, Sousa R, Pinto C, Ribeiro L, Abreu C, Torres S, Quintela A, Mata G, Megías D, Ribot J, Serre K, Casimiro S, Silva-Santos B, Diéguez L, Costa L. Effect of Cyclin-Dependent Kinase 4/6 Inhibitors on Circulating Cells in Patients with Metastatic Breast Cancer. Cells 2024; 13:1391. [PMID: 39195280 PMCID: PMC11487375 DOI: 10.3390/cells13161391] [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: 07/19/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024] Open
Abstract
The combination of cyclin-dependent kinase 4/6 inhibitors (CDK4/6i) with endocrine therapy (ET) is the standard-of-care for estrogen receptor (ER)-positive, HER2-negative (ER+/HER2- advanced/metastatic breast cancer (mBC). However, the impact of CDK4/6i on circulating immune cells and circulating tumor cells (CTCs) in patients receiving CDK4/6i and ET (CDK4/6i+ET) remains poorly understood. This was a prospective cohort study including 44 patients with ER+/HER2- mBC treated with CDK4/6i+ET in either first or second line. Peripheral blood samples were collected before (baseline) and 3 months (t2) after therapy. Immune cell's subsets were quantified by flow cytometry, and microfluidic-captured CTCs were counted and classified according to the expression of cytokeratin and/or vimentin. Patients were categorized according to response as responders (progression-free survival [PFS] ≥ 6.0 months; 79.1%) and non-responders (PFS < 6.0 months; 20.9%). CDK4/6i+ET resulted in significant changes in the hematological parameters, including decreased hemoglobin levels and increased mean corpuscular volume, as well as reductions in neutrophil, eosinophil, and basophil counts. Specific immune cell subsets, such as early-stage myeloid-derived suppressor cells, central memory CD4+ T cells, and Vδ2+ T cells expressing NKG2D, decreased 3 months after CDK4/6i+ET. Additionally, correlations between the presence of CTCs and immune cell populations were observed, highlighting the interplay between immune dysfunction and tumor dissemination. This study provides insights into the immunomodulatory effects of CDK4/6i+ET, underscoring the importance of considering immune dynamics in the management of ER+/HER2- mBC.
Collapse
Affiliation(s)
- Soraia Lobo-Martins
- Academic Trials Promoting Team, Institut Jules Bordet, Université Libre de Bruxelles (U.L.B.), 1070 Bruxelles, Belgium;
- Medical Oncology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, 1649-028 Lisbon, Portugal (L.R.)
- Instituto de Medicina Molecular João Lobo Antunes, 1649-028 Lisbon, Portugal; (P.C.); (A.C.); (S.C.)
| | - Patrícia Corredeira
- Instituto de Medicina Molecular João Lobo Antunes, 1649-028 Lisbon, Portugal; (P.C.); (A.C.); (S.C.)
| | - Ana Cavaco
- Instituto de Medicina Molecular João Lobo Antunes, 1649-028 Lisbon, Portugal; (P.C.); (A.C.); (S.C.)
| | - Carolina Rodrigues
- International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga s/n, 4715-330 Braga, Portugal (C.L.)
| | - Paulina Piairo
- International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga s/n, 4715-330 Braga, Portugal (C.L.)
- RUBYnanomed Lda, Praça Conde de Agrolongo, 4700-314 Braga, Portugal
| | - Cláudia Lopes
- International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga s/n, 4715-330 Braga, Portugal (C.L.)
| | - Joana Fraga
- RUBYnanomed Lda, Praça Conde de Agrolongo, 4700-314 Braga, Portugal
| | - Madalena Silva
- RUBYnanomed Lda, Praça Conde de Agrolongo, 4700-314 Braga, Portugal
| | - Patrícia Alves
- START Lisboa-CHULN Hospital Santa Maria, 1649-028 Lisbon, Portugal;
| | - Lisiana Wachholz Szeneszi
- Medical Oncology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, 1649-028 Lisbon, Portugal (L.R.)
| | - Ana Barradas
- Medical Oncology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, 1649-028 Lisbon, Portugal (L.R.)
| | - Camila Castro Duran
- Medical Oncology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, 1649-028 Lisbon, Portugal (L.R.)
| | - Marília Antunes
- Centro de Estatística e Aplicações, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisbon, Portugal
| | - Gonçalo Nogueira-Costa
- Medical Oncology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, 1649-028 Lisbon, Portugal (L.R.)
- Faculdade de Medicina, Universidade de Lisboa, 1649-004 Lisbon, Portugal
| | - Rita Sousa
- Medical Oncology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, 1649-028 Lisbon, Portugal (L.R.)
- Faculdade de Medicina, Universidade de Lisboa, 1649-004 Lisbon, Portugal
| | - Conceição Pinto
- Medical Oncology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, 1649-028 Lisbon, Portugal (L.R.)
| | - Leonor Ribeiro
- Medical Oncology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, 1649-028 Lisbon, Portugal (L.R.)
- Faculdade de Medicina, Universidade de Lisboa, 1649-004 Lisbon, Portugal
| | - Catarina Abreu
- Medical Oncology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, 1649-028 Lisbon, Portugal (L.R.)
- Faculdade de Medicina, Universidade de Lisboa, 1649-004 Lisbon, Portugal
| | - Sofia Torres
- Medical Oncology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, 1649-028 Lisbon, Portugal (L.R.)
- Faculdade de Medicina, Universidade de Lisboa, 1649-004 Lisbon, Portugal
| | - António Quintela
- Medical Oncology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, 1649-028 Lisbon, Portugal (L.R.)
| | - Gadea Mata
- Matemáticas y Computación Department, Universidad de La Rioja, 26006 Logroño, Spain
| | - Diego Megías
- Confocal Microscopy Unit, Centro Nacional de Investigaciones Oncológicas (CNIO-ISCIII), 28029 Madrid, Spain
| | - Julie Ribot
- Instituto de Medicina Molecular João Lobo Antunes, 1649-028 Lisbon, Portugal; (P.C.); (A.C.); (S.C.)
| | - Karine Serre
- Instituto de Medicina Molecular João Lobo Antunes, 1649-028 Lisbon, Portugal; (P.C.); (A.C.); (S.C.)
- Faculdade de Medicina, Universidade de Lisboa, 1649-004 Lisbon, Portugal
- iMM Laço Hub, iMM-CARE, 1649-028 Lisbon, Portugal
| | - Sandra Casimiro
- Instituto de Medicina Molecular João Lobo Antunes, 1649-028 Lisbon, Portugal; (P.C.); (A.C.); (S.C.)
- Faculdade de Medicina, Universidade de Lisboa, 1649-004 Lisbon, Portugal
| | - Bruno Silva-Santos
- Instituto de Medicina Molecular João Lobo Antunes, 1649-028 Lisbon, Portugal; (P.C.); (A.C.); (S.C.)
- Faculdade de Medicina, Universidade de Lisboa, 1649-004 Lisbon, Portugal
| | - Lorena Diéguez
- International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga s/n, 4715-330 Braga, Portugal (C.L.)
- RUBYnanomed Lda, Praça Conde de Agrolongo, 4700-314 Braga, Portugal
| | - Luís Costa
- Medical Oncology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, 1649-028 Lisbon, Portugal (L.R.)
- Instituto de Medicina Molecular João Lobo Antunes, 1649-028 Lisbon, Portugal; (P.C.); (A.C.); (S.C.)
- Faculdade de Medicina, Universidade de Lisboa, 1649-004 Lisbon, Portugal
| |
Collapse
|
38
|
Chen L, Yang G, Qu F. Aptamer-based sensors for fluid biopsies of protein disease markers. Talanta 2024; 276:126246. [PMID: 38796994 DOI: 10.1016/j.talanta.2024.126246] [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: 02/18/2024] [Revised: 05/08/2024] [Accepted: 05/10/2024] [Indexed: 05/29/2024]
Abstract
Fluid biopsy technology, characterized by its minimally invasive nature, speed, and continuity, has become a rapidly advancing and widely applied real-time diagnostic technique. Among various biomarkers, proteins represent the most abundant class of disease indicators. The sensitive and accurate detection of protein markers in bodily fluids is significantly influenced by the control exerted by recognition ligands. Aptamers, which are structurally dynamic functional oligonucleotides, exhibit high affinity, specific recognition of targets, and notable characteristics of high editability and modularity. These features make aptamer universal "recognition-capture" components, contribute to a significant leap in their applications within the biosensor domain. In this context, we provide a comprehensive review of the extensive application of aptamer-based biosensors in fluid biopsy. We systematically compile the characteristics and construction strategies of aptamer-based biosensors tailored for fluid biopsy, including aptamer sequences, affinity (KD), fluid background, sensing technologies, sensor construction strategies, incubation time, detection performance, and influencing factors. Furthermore, a comparative analysis of their advantages and disadvantages was conducted. In conclusion, we delineate and deliberate on prospective research trajectories and challenges that lie ahead in the realm of aptamer-based biosensors for fluid biopsy.
Collapse
Affiliation(s)
- Li Chen
- School of Life Science, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Ge Yang
- CAMS Key Laboratory of Antiviral Drug Research, Beijing Key Laboratory of Antimicrobial Agents, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Feng Qu
- School of Life Science, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, China.
| |
Collapse
|
39
|
Pan J, Wu J, Wang B, Zhu B, Liu X, Gan H, Wei Y, Jin S, Hu X, Wang Q, Song S, Liu C, Ye D, Zhu Y. Interlesional response heterogeneity is associated with the prognosis of abiraterone treatment in metastatic castration-resistant prostate cancer. MED 2024:S2666-6340(24)00302-7. [PMID: 39151419 DOI: 10.1016/j.medj.2024.07.020] [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: 01/02/2024] [Revised: 04/15/2024] [Accepted: 07/23/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND Interlesional response heterogeneity (ILRH) poses challenges to the treatment of metastatic castration-resistant prostate cancer (mCRPC). Currently, there are no prospective clinical trials exploring the prognostic significance of ILRH on paired positron emission tomography/computed tomography (PET/CT) in the context of abiraterone therapy. METHODS In this prospective study, we enrolled patients with mCRPC treated with abiraterone (ClinicalTrials.gov: NCT05188911; ChiCTR.org.cn: ChiCTR2000034708). 68Ga-prostate-specific membrane antigen (PSMA)+18F-fluorodeoxyglucose (FDG) PET/CT and circulating tumor DNA (ctDNA) monitoring were performed at baseline and week 13. Patients were grouped by their early ILRH measurement. The primary endpoint was to evaluate the predictive role of ILRH for conventional progression-free survival (PFS) through the concordance index (C-index) assessment. Conventional PFS was defined as the time from medication to conventional radiographic progression, clinical progression, or death. FINDINGS Ultimately, 33 patients were included with a median follow-up of 28.7 months. Baseline+week 13 PSMA PET/CT revealed that 33.3% of patients showed ILRH. Those patients with hetero-responding disease had significantly different PFS compared to the responding and non-responding groups (hazard ratio: responding group = reference, hetero-responding group = 4.0, non-responding group = 5.8; p < 0.0001). The C-index of ILRH on paired PSMA PET/CT (0.742 vs. 0.660) and FDG PET/CT (0.736 vs. 0.668) for conventional PFS was higher than that of PSA response. In an exploratory analysis, PSMA-/FDG+ lesions at week 13 were identified as a strong surrogate for poor conventional PFS (p = 0.039). CONCLUSIONS ILRH on both baseline+week 13 PSMA and FDG PET/CT strongly associated with conventional PFS. FUNDING This study was funded by the Ministry of Science and Technology of China and Shanghai.
Collapse
Affiliation(s)
- Jian Pan
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China; Shanghai Genitourinary Cancer Institute, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Junlong Wu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China; Shanghai Genitourinary Cancer Institute, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Beihe Wang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China; Shanghai Genitourinary Cancer Institute, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bin Zhu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xiaohang Liu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Hualei Gan
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yu Wei
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China; Shanghai Genitourinary Cancer Institute, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shengming Jin
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China; Shanghai Genitourinary Cancer Institute, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaoxin Hu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Qifeng Wang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Shaoli Song
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China.
| | - Chang Liu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China.
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China; Shanghai Genitourinary Cancer Institute, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Yao Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China; Shanghai Genitourinary Cancer Institute, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
40
|
Ashouri K, Wong A, Mittal P, Torres-Gonzalez L, Lo JH, Soni S, Algaze S, Khoukaz T, Zhang W, Yang Y, Millstein J, Lenz HJ, Battaglin F. Exploring Predictive and Prognostic Biomarkers in Colorectal Cancer: A Comprehensive Review. Cancers (Basel) 2024; 16:2796. [PMID: 39199569 PMCID: PMC11353018 DOI: 10.3390/cancers16162796] [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: 06/28/2024] [Revised: 08/04/2024] [Accepted: 08/07/2024] [Indexed: 09/01/2024] Open
Abstract
Colorectal cancer (CRC) remains the second leading cause of cancer-related mortality worldwide. While immune checkpoint inhibitors have significantly improved patient outcomes, their effectiveness is mostly limited to tumors with microsatellite instability (MSI-H/dMMR) or an increased tumor mutational burden, which comprise 10% of cases. Advancing personalized medicine in CRC hinges on identifying predictive biomarkers to guide treatment decisions. This comprehensive review examines established tissue markers such as KRAS and HER2, highlighting their roles in resistance to anti-EGFR agents and discussing advances in targeted therapies for these markers. Additionally, this review summarizes encouraging data on promising therapeutic targets and highlights the clinical utility of liquid biopsies. By synthesizing current evidence and identifying knowledge gaps, this review provides clinicians and researchers with a contemporary understanding of the biomarker landscape in CRC. Finally, the review examines future directions and challenges in translating promising biomarkers into clinical practice, with the goal of enhancing personalized medicine approaches for colorectal cancer patients.
Collapse
Affiliation(s)
- Karam Ashouri
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; (K.A.); (A.W.)
| | - Alexandra Wong
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; (K.A.); (A.W.)
| | - Pooja Mittal
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; (K.A.); (A.W.)
| | - Lesly Torres-Gonzalez
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; (K.A.); (A.W.)
| | - Jae Ho Lo
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; (K.A.); (A.W.)
| | - Shivani Soni
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; (K.A.); (A.W.)
| | - Sandra Algaze
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; (K.A.); (A.W.)
| | - Taline Khoukaz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; (K.A.); (A.W.)
| | - Wu Zhang
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; (K.A.); (A.W.)
| | - Yan Yang
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; (K.A.); (A.W.)
| | - Joshua Millstein
- Department of Population and Public Health Sciences, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; (K.A.); (A.W.)
| | - Francesca Battaglin
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; (K.A.); (A.W.)
| |
Collapse
|
41
|
Taghizadeh-Teymorloei M, Alizadeh L, Matin S, Jafari-Koshki T, Karimi A. Diagnostic and prognostic significance of ALU-based cell-free DNA in colorectal cancer: a systematic review and meta-analysis. Front Oncol 2024; 14:1398062. [PMID: 39169935 PMCID: PMC11335620 DOI: 10.3389/fonc.2024.1398062] [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: 03/08/2024] [Accepted: 07/18/2024] [Indexed: 08/23/2024] Open
Abstract
Introduction Colorectal cancer (CRC) is a major global health concern. This study aimed to investigate the role of ALU-based cell-free DNA (cfDNA) in the diagnosis and prognosis of CRC. Methods We selected relevant literature from PubMed, Scopus, Web of Science, EMBASE, and Science Direct databases based on strict inclusion and exclusion criteria. 17 eligible studies were included in the final analysis (13 studies for diagnostic and 4 studies for prognostic meta-analysis). The search covered relevant publications up to July 1, 2024. Results The pooled sensitivity, specificity, and diagnostic odds ratios (DOR) of ALU-based cfDNA in CRC diagnosis were 0.81 (95% CI= [0.70, 0.89]), 0.90 (95% CI= [0.70, 0.96]), and 40.58 (95% CI= [17.87, 92.19]), respectively. The area under the ROC curve was 0.92 (95% CI= [0.89, 0.94]). Patients with higher concentrations of plasma/serum ALU-based cfDNA had poorer overall survival (OS) (pooled hazard ratio = 2.33 ([95% CI= [1.80, 3.03]). Conclusion The current evidence supports the utility of circulating ALU as a promising non-invasive diagnostic and prognostic tool for CRC. Furthermore, as a potential biomarker, ALU-based cfDNA could play a significant role in clinical application. Clinical implications The evidence suggests that circulating ALU-based cell-free DNA (cfDNA) holds promise as a non-invasive diagnostic and prognostic tool for colorectal cancer, potentially enhancing clinical decision-making. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier PROSPERO (CRD42023486369).
Collapse
Affiliation(s)
- Mohammad Taghizadeh-Teymorloei
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Alizadeh
- Gastroenterology and Liver Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Somaieh Matin
- Department of Internal Medicine, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Tohid Jafari-Koshki
- Molecular Medicine Research Center (MMRC), Department of Statistics and Epidemiology, Faculty of Health, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abbas Karimi
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Epidemiology and Biostatistics, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
42
|
Werner R, Crosbie R, Dorney M, Connolly A, Collins D, Hand CK, Burke L. Implementation of an ISO 15189 accredited next generation sequencing service for cell-free total nucleic acid (cfTNA) analysis to facilitate driver mutation reporting in blood: the experience of a clinical diagnostic laboratory. J Clin Pathol 2024:jcp-2024-209514. [PMID: 38914446 DOI: 10.1136/jcp-2024-209514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 06/11/2024] [Indexed: 06/26/2024]
Abstract
AIMS Next generation sequencing (NGS) on tumour tissue is integral to the delivery of personalised medicine and targeted therapy. NGS on liquid biopsy, a much less invasive technology, is an emerging clinical tool that has rapidly expanded clinical utility. Gene mutations in cell-free total nucleic acids (cfTNA) circulating in the blood are representative of whole tumour biology and can reveal different mutations from different tumour sites, thus addressing tumour heterogeneity challenges. METHODS The novel Ion Torrent Genexus NGS system with automated sample preparation, onboard library preparation, templating, sequencing, data analysis and Oncomine Reporter software was used. cfTNA extracted from plasma was verified with the targeted pan-cancer (~50 genes) Oncomine Precision Assay (OPA). Assessment criteria included analytical sensitivity, specificity, limits of detection (LOD), accuracy, repeatability, reproducibility and the establishment of performance metrics. RESULTS An ISO 15189 accredited, minimally invasive cfTNA NGS diagnostic service has been implemented. High sensitivity (>83%) and specificity between plasma and tissue were observed. A sequencing LOD of 1.2% was achieved when the depth of coverage was >22 000×. A reduction (>68%) in turnaround time (TAT) of liquid biopsy results was achieved: 5 days TAT for in-house analysis from sample receipt to a final report issued to oncologists as compared with >15 days from reference laboratories. CONCLUSION Tumour-derived somatic variants can now be reliably assessed from plasma to provide minimally invasive tumour profiling. Successful implementation of this accredited service resulted in:Appropriate molecular profiling of patients where tumour tissue is unavailable or inaccessible.Rapid TAT of plasma NGS results.
Collapse
Affiliation(s)
- Reiltin Werner
- Pathology Department, Cork University Hospital, Cork, Ireland
- Department of Pathology, School of Medicine, University College Cork College of Medicine and Health, Cork, Ireland
| | - Ruth Crosbie
- Pathology Department, Cork University Hospital, Cork, Ireland
| | - Mairead Dorney
- Pathology Department, Cork University Hospital, Cork, Ireland
| | - Amy Connolly
- Pathology Department, Cork University Hospital, Cork, Ireland
| | | | - Collette K Hand
- Department of Pathology, School of Medicine, University College Cork College of Medicine and Health, Cork, Ireland
| | - Louise Burke
- Pathology Department, Cork University Hospital, Cork, Ireland
- Department of Pathology, School of Medicine, University College Cork College of Medicine and Health, Cork, Ireland
| |
Collapse
|
43
|
Sun M, Wu Y, Zhang J, Zhang H, Liu Z, Li M, Wang C, Sitti M. Versatile, modular, and customizable magnetic solid-droplet systems. Proc Natl Acad Sci U S A 2024; 121:e2405095121. [PMID: 39088393 PMCID: PMC11317579 DOI: 10.1073/pnas.2405095121] [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: 03/12/2024] [Accepted: 06/21/2024] [Indexed: 08/03/2024] Open
Abstract
Magnetic miniature robotic systems have attracted broad research interest because of their precise maneuverability in confined spaces and adaptability to diverse environments, holding significant promise for applications in both industrial infrastructures and biomedical fields. However, the predominant construction methodology involves the preprogramming of magnetic components into the system's structure. While this approach allows for intricate shape transformations, it exhibits limited flexibility in terms of reconfiguration and presents challenges when adapting to diverse materials, combining, and decoupling multiple functionalities. Here, we propose a construction strategy that facilitates the on-demand assembly of magnetic components, integrating ferrofluid droplets with the system's structural body. This approach enables the creation of complex solid-droplet robotic systems across a spectrum of length scales, ranging from 0.8 mm to 1.5 cm. It offers a diverse selection of materials and structural configurations, akin to assembling components like building blocks, thus allowing for the seamless integration of various functionalities. Moreover, it incorporates decoupling mechanisms to enable selective control over multiple functions, leveraging the fluidity, fission/fusion, and magneto-responsiveness properties inherent in the ferrofluid. Various solid-droplet systems have validated the feasibility of this strategy. This study advances the complexity and functionality achievable in small-scale magnetic robots, augmenting their potential for future biomedical and other applications.
Collapse
Affiliation(s)
- Mengmeng Sun
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, Stuttgart70569, Germany
| | - Yingdan Wu
- State Key Laboratory of Robotics and Systems, Harbin Institute of Technology, Harbin150001, China
| | - Jianhua Zhang
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, Stuttgart70569, Germany
| | - Hongchuan Zhang
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, Stuttgart70569, Germany
| | - Zemin Liu
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, Stuttgart70569, Germany
- School of Medicine and College of Engineering, Koç University, Istanbul34450, Türkiye
| | - Mingtong Li
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, Stuttgart70569, Germany
| | - Chunxiang Wang
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, Stuttgart70569, Germany
- School of Medicine and College of Engineering, Koç University, Istanbul34450, Türkiye
| | - Metin Sitti
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, Stuttgart70569, Germany
- School of Medicine and College of Engineering, Koç University, Istanbul34450, Türkiye
| |
Collapse
|
44
|
Slusher N, Jones N, Nonaka T. Liquid biopsy for diagnostic and prognostic evaluation of melanoma. Front Cell Dev Biol 2024; 12:1420360. [PMID: 39156972 PMCID: PMC11327088 DOI: 10.3389/fcell.2024.1420360] [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: 04/20/2024] [Accepted: 07/03/2024] [Indexed: 08/20/2024] Open
Abstract
Melanoma is the most aggressive form of skin cancer, and the majority of cases are associated with chronic or intermittent sun exposure. The incidence of melanoma has grown exponentially over the last 50 years, especially in populations of fairer skin, at lower altitudes and in geriatric populations. The gold standard for diagnosis of melanoma is performing an excisional biopsy with full resection or an incisional tissue biopsy. However, due to their invasiveness, conventional biopsy techniques are not suitable for continuous disease monitoring. Utilization of liquid biopsy techniques represent substantial promise in early detection of melanoma. Through this procedure, tumor-specific components shed into circulation can be analyzed for not only diagnosis but also treatment selection and risk assessment. Additionally, liquid biopsy is significantly less invasive than tissue biopsy and offers a novel way to monitor the treatment response and disease relapse, predicting metastasis.
Collapse
Affiliation(s)
- Nicholas Slusher
- School of Medicine, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - Nicholas Jones
- School of Medicine, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - Taichiro Nonaka
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA, United States
- Feist-Weiller Cancer Center, Louisiana State University Health Shreveport, Shreveport, LA, United States
| |
Collapse
|
45
|
Suri S, Boora GS, Kaur R, Chauhan A, Ghoshal S, Pal A. Recent advances in minimally invasive biomarkers of OSCC: from generalized to personalized approach. FRONTIERS IN ORAL HEALTH 2024; 5:1426507. [PMID: 39157206 PMCID: PMC11327221 DOI: 10.3389/froh.2024.1426507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 07/16/2024] [Indexed: 08/20/2024] Open
Abstract
Oral cancer is the 6th most common type of cancer worldwide, and oral squamous cell carcinoma (OSCC) accounts for >90% of oral cancers. It is a major health problem, particularly in low- and middle-income countries (LMICs), due to both its high incidence and significant mortality and morbidity. Despite being a global burden, and even with the significant advancement in the management of OSCC, the overall outcome of the disease is still abysmal. With the advent of time, advanced diagnostic and treatment approaches have come into practice, but the burden of the disease has not improved significantly. Major reasons attributed to the poor outcome are delay in diagnosis, locoregional recurrence and resistance to the currently available treatment regimen. In this review, we have highlighted the existing challenges in the diagnosis and have emphasized the advancements in minimally invasive biomarkers. Additionally, the importance of collaborative multidimensional approaches involving clinicians and researchers has been discussed, as well as the need to redefine and establish better utility and management of existing diagnostic and treatment protocols along with the minimally invasive/non-invasive biomarkers.
Collapse
Affiliation(s)
- Smriti Suri
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh,India
| | - Geeta S. Boora
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh,India
| | - Rajandeep Kaur
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh,India
| | - Anshika Chauhan
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh,India
| | - Sushmita Ghoshal
- Department of Radiotherapy, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Arnab Pal
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh,India
| |
Collapse
|
46
|
Ferchiou S, Caza F, Sinha K, Sauvageau J, St-Pierre Y. Assessing marine ecosystem health using multi-omic analysis of blue mussel liquid biopsies: A case study within a national marine park. CHEMOSPHERE 2024; 362:142714. [PMID: 38950751 DOI: 10.1016/j.chemosphere.2024.142714] [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: 04/09/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/03/2024]
Abstract
Marine ecosystems are under escalating threats from myriad environmental stressors, necessitating a deeper understanding of their impact on biodiversity and the health of sentinel organisms. In this study, we carried out a spatiotemporal multi-omic analysis of liquid biopsies collected from mussels (Mytilus spp.) in marine ecosystems of a national park. We delved into the epigenomic, transcriptomic, glycomic, proteomic, and microbiomic profiles to unravel the intricate interplay between ecosystem biodiversity and mussels' biological response to their environments. Our analysis revealed temporal fluctuations in the alpha diversity of the circulating microbiome associated with human activities. Analysis of the hemolymphatic circulating cell-free DNA (ccfDNA) provided information on the biodiversity and the presence of potential pathogens. Epigenomic analysis revealed widespread hypomethylation sites within the mitochondrial (mtDNA). Comparative transcriptomic and glycomic analyses highlighted differences in metabolic pathways and genes associated with immune and wound healing functions. This study demonstrates the potential of multi-omic analysis of liquid biopsy in sentinel to provide a holistic view of human activities' environmental impacts on marine coastal ecosystems. Overall, this approach has the potential to enhance the effectiveness and efficiency of various conservation efforts, leading to more informed decision-making and better outcomes for biodiversity and ecosystem conservation.
Collapse
Affiliation(s)
- Sophia Ferchiou
- INRS-Center Armand-Frappier Santé Technologie, 531 Boul. des Prairies, Laval, QC, Canada, H7V 1B7
| | - France Caza
- INRS-Center Armand-Frappier Santé Technologie, 531 Boul. des Prairies, Laval, QC, Canada, H7V 1B7
| | - Kumardip Sinha
- Human Health Therapeutics, National Research Council, 100 Sussex Dr., K1N 5A2, Ottawa, Ontario, Canada
| | - Janelle Sauvageau
- Human Health Therapeutics, National Research Council, 100 Sussex Dr., K1N 5A2, Ottawa, Ontario, Canada
| | - Yves St-Pierre
- INRS-Center Armand-Frappier Santé Technologie, 531 Boul. des Prairies, Laval, QC, Canada, H7V 1B7.
| |
Collapse
|
47
|
Wang R, Wang B, Zhang H, Liao X, Shi B, Zhou Y, Zhou C, Yan Y, Zhang W, Wang K, Ge G, Ren Y, Tang X, Gan B, He J, Niu L. Early evaluation of circulating tumor DNA as marker of therapeutic efficacy and prognosis in breast cancer patients during primary systemic therapy. Breast 2024; 76:103738. [PMID: 38685149 PMCID: PMC11067540 DOI: 10.1016/j.breast.2024.103738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/17/2024] [Accepted: 04/23/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND We assessed the potential role of serial circulating tumor DNA (ctDNA) as a biomarker to monitor treatment response to primary systemic therapy (PST) in breast cancer and evaluated the predictive value of ctDNA to further identify patients with residual disease. METHODS We prospectively enrolled 208 plasma samples collected at three time points (before PST, after 2 cycles of treatment, before surgery) of 72 patients with stage Ⅱ-III breast cancer. Somatic mutations in plasma samples were identified using a customized 128-gene capture panel with next-generation sequencing. The correlation between early change in ctDNA levels and treatment response or long-term clinical outcomes was assessed. RESULTS 37 of 72 (51.4%) patients harbored detectable ctDNA alterations at baseline. Patients with complete response showed a larger decrease in ctDNA levels during PST. The median relative change of variant allele fraction (VAF) was -97.4%, -46.7%, and +21.1% for patients who subsequently had a complete response (n = 11), partial response (n = 11), and no response (n = 15) (p = 0.0012), respectively. In addition, the relative change of VAF between the pretreatment and first on-treatment blood draw exhibited the optimal predictive value to tumor response after PST (area under the curve, AUC = 0.7448, p = 0.02). More importantly, early change of ctDNA levels during treatment have significant prognostic value for patients with BC, there was a significant correlation between early decrease of VAF and longer recurrence-free survival compared to those with an VAF increase (HR = 12.54; 95% CI, 2.084 to 75.42, p = 0.0063). CONCLUSION Early changes of ctDNA are strongly correlated with therapeutic efficacy to PST and clinical outcomes in BC patients. The integration of preoperative ctDNA evaluation could help improving the perioperative management for BC patients receiving PST.
Collapse
Affiliation(s)
- Ru Wang
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Bin Wang
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Huimin Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Xiaoqin Liao
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Bohui Shi
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Yuhui Zhou
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Can Zhou
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Yu Yan
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Wei Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Ke Wang
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Guanqun Ge
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Yu Ren
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Xiaojiang Tang
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Baoyu Gan
- Biobank of the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Jianjun He
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| | - Ligang Niu
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
48
|
Hsu CC, Su Y, Rittenhouse-Olson K, Attwood KM, Mojica W, Reid ME, Dy GK, Wu Y. Exosomal Thomsen-Friedenreich Glycoantigen: A New Liquid Biopsy Biomarker for Lung and Breast Cancer Diagnoses. CANCER RESEARCH COMMUNICATIONS 2024; 4:1933-1945. [PMID: 39015090 DOI: 10.1158/2767-9764.crc-23-0505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 04/16/2024] [Accepted: 07/12/2024] [Indexed: 07/18/2024]
Abstract
Exosomes are nanosized extracellular vesicles released by cells to transport biomolecules such as proteins and RNAs for intercellular communication. Exosomes play important roles in cancer development and metastasis; therefore, they have emerged as potential liquid biopsy biomarkers for cancer screening, diagnosis, and management. Many exosome cargos, including proteins, RNAs, and lipids, have been extensively investigated as biomarkers for cancer liquid biopsy. However, carbohydrates, an important type of biomolecule, have not yet been explored for this purpose. In this study, we reported a new exosomal carbohydrate biomarker, α-linked Thomsen-Friedenreich glycoantigen (TF-Ag-α; Galβ1-3GalNAc-α). To translate our discovery into clinical settings, we developed a surface plasmon resonance-based assay which utilized a unique mAb, JAA-F11, with high specificity to measure the levels of exosomal TF-Ag-α in blood. To the best of our knowledge, we are the first to demonstrate that exosomes carry TF-Ag-α. We detected exosomal TF-Ag-α in as low as 10 μL serum samples from patients with cancer, but in contrast, levels were negligible in those from normal controls. With a total of 233 patients with cancer and normal controls, we showed that exosomal TF-Ag-α detected lung cancer (n = 60) and breast cancer (n = 95) from normal controls (n = 78) with ≥95% and ≥97% accuracy, respectively. These results demonstrated that exosomal TF-Ag-α is a potential liquid biopsy biomarker for cancer diagnosis. SIGNIFICANCE Exosomes or small extracellular vesicles have emerged as potent biomarkers of cancer liquid biopsy. We discovered a new exosomal carbohydrate marker, TF-Ag-α (Galβ1-3GalNAc-α), and showed that exosomal TF-Ag-α detected both lung and breast cancers with >95% accuracy. Our findings demonstrated that exosomal TF-Ag-α is a promising liquid biopsy biomarker for cancer screening and early detection.
Collapse
Affiliation(s)
- Chang-Chieh Hsu
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, New York
| | - Yafei Su
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, New York
| | | | - Kristopher M Attwood
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Wilfrido Mojica
- Department of Pathology and Anatomical Sciences, University at Buffalo, The State University of New York, Buffalo, New York
| | - Mary E Reid
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Grace K Dy
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Yun Wu
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, New York
| |
Collapse
|
49
|
Linscott JA, Miyagi H, Murthy PB, Yao S, Grass GD, Vosoughi A, Xu H, Wang X, Yu X, Yu A, Zemp L, Gilbert SM, Poch MA, Sexton WJ, Spiess PE, Li R. From Detection to Cure - Emerging Roles for Urinary Tumor DNA (utDNA) in Bladder Cancer. Curr Oncol Rep 2024; 26:945-958. [PMID: 38837106 DOI: 10.1007/s11912-024-01555-0] [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] [Accepted: 05/20/2024] [Indexed: 06/06/2024]
Abstract
PURPOSE OF REVIEW This review sought to define the emerging roles of urinary tumor DNA (utDNA) for diagnosis, monitoring, and treatment of bladder cancer. Building from early landmark studies the focus is on recent studies, highlighting how utDNA could aid personalized care. RECENT FINDINGS Recent research underscores the potential for utDNA to be the premiere biomarker in bladder cancer due to the constant interface between urine and tumor. Many studies find utDNA to be more informative than other biomarkers in bladder cancer, especially in early stages of disease. Points of emphasis include superior sensitivity over traditional urine cytology, broad genomic and epigenetic insights, and the potential for non-invasive, real-time analysis of tumor biology. utDNA shows promise for improving all phases of bladder cancer care, paving the way for personalized treatment strategies. Building from current research, future comprehensive clinical trials will validate utDNA's clinical utility, potentially revolutionizing bladder cancer management.
Collapse
Affiliation(s)
- Joshua A Linscott
- Department of Genitourinary Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| | - Hiroko Miyagi
- Department of Genitourinary Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Prithvi B Murthy
- Department of Genitourinary Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Sijie Yao
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - G Daniel Grass
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Aram Vosoughi
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Hongzhi Xu
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Xuefeng Wang
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Xiaoqing Yu
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Alice Yu
- Department of Genitourinary Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Logan Zemp
- Department of Genitourinary Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Scott M Gilbert
- Department of Genitourinary Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Michael A Poch
- Department of Genitourinary Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Wade J Sexton
- Department of Genitourinary Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Philippe E Spiess
- Department of Genitourinary Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Roger Li
- Department of Genitourinary Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| |
Collapse
|
50
|
Mason CE, Green J, Adamopoulos KI, Afshin EE, Baechle JJ, Basner M, Bailey SM, Bielski L, Borg J, Borg J, Broddrick JT, Burke M, Caicedo A, Castañeda V, Chatterjee S, Chin CR, Church G, Costes SV, De Vlaminck I, Desai RI, Dhir R, Diaz JE, Etlin SM, Feinstein Z, Furman D, Garcia-Medina JS, Garrett-Bakelman F, Giacomello S, Gupta A, Hassanin A, Houerbi N, Irby I, Javorsky E, Jirak P, Jones CW, Kamal KY, Kangas BD, Karouia F, Kim J, Kim JH, Kleinman AS, Lam T, Lawler JM, Lee JA, Limoli CL, Lucaci A, MacKay M, McDonald JT, Melnick AM, Meydan C, Mieczkowski J, Muratani M, Najjar D, Othman MA, Overbey EG, Paar V, Park J, Paul AM, Perdyan A, Proszynski J, Reynolds RJ, Ronca AE, Rubins K, Ryon KA, Sanders LM, Glowe PS, Shevde Y, Schmidt MA, Scott RT, Shirah B, Sienkiewicz K, Sierra MA, Siew K, Theriot CA, Tierney BT, Venkateswaran K, Hirschberg JW, Walsh SB, Walter C, Winer DA, Yu M, Zea L, Mateus J, Beheshti A. A second space age spanning omics, platforms and medicine across orbits. Nature 2024; 632:995-1008. [PMID: 38862027 DOI: 10.1038/s41586-024-07586-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 05/18/2024] [Indexed: 06/13/2024]
Abstract
The recent acceleration of commercial, private and multi-national spaceflight has created an unprecedented level of activity in low Earth orbit, concomitant with the largest-ever number of crewed missions entering space and preparations for exploration-class (lasting longer than one year) missions. Such rapid advancement into space from many new companies, countries and space-related entities has enabled a 'second space age'. This era is also poised to leverage, for the first time, modern tools and methods of molecular biology and precision medicine, thus enabling precision aerospace medicine for the crews. The applications of these biomedical technologies and algorithms are diverse, and encompass multi-omic, single-cell and spatial biology tools to investigate human and microbial responses to spaceflight. Additionally, they extend to the development of new imaging techniques, real-time cognitive assessments, physiological monitoring and personalized risk profiles tailored for astronauts. Furthermore, these technologies enable advancements in pharmacogenomics, as well as the identification of novel spaceflight biomarkers and the development of corresponding countermeasures. In this Perspective, we highlight some of the recent biomedical research from the National Aeronautics and Space Administration, Japan Aerospace Exploration Agency, European Space Agency and other space agencies, and detail the entrance of the commercial spaceflight sector (including SpaceX, Blue Origin, Axiom and Sierra Space) into aerospace medicine and space biology, the first aerospace medicine biobank, and various upcoming missions that will utilize these tools to ensure a permanent human presence beyond low Earth orbit, venturing out to other planets and moons.
Collapse
Affiliation(s)
- Christopher E Mason
- Department of Physiology and Biophysics and Tri-Institutional Computational Biology and Medicine Program, Weill Cornell Medicine, New York, NY, USA.
- The WorldQuant Initiative for Quantitative Prediction, New York, NY, USA.
| | | | - Konstantinos I Adamopoulos
- Blue Marble Space Institute of Science, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
- Biomedical Engineering Laboratory, School of Electrical and Computer Engineering, National University of Athens, Athens, Greece
| | - Evan E Afshin
- Department of Physiology and Biophysics and Tri-Institutional Computational Biology and Medicine Program, Weill Cornell Medicine, New York, NY, USA
| | - Jordan J Baechle
- Buck Artificial Intelligence Platform, Buck Institute for Research on Aging, Novato, CA, USA
| | - Mathias Basner
- Unit for Experimental Psychiatry, Division of Sleep and Chronobiology, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Susan M Bailey
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
| | - Luca Bielski
- Department of Physiology and Biophysics and Tri-Institutional Computational Biology and Medicine Program, Weill Cornell Medicine, New York, NY, USA
| | - Josef Borg
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, Malta
| | - Joseph Borg
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, Malta
| | - Jared T Broddrick
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - Marissa Burke
- Department of Physiology and Biophysics and Tri-Institutional Computational Biology and Medicine Program, Weill Cornell Medicine, New York, NY, USA
- Embry-Riddle Aeronautical University, Department of Human Factors and Behavioral Neurobiology, Daytona Beach, FL, USA
| | - Andrés Caicedo
- Instituto de Investigaciones en Biomedicina iBioMed, Universidad San Francisco de Quito USFQ, Quito, Ecuador
- Escuela de Medicina, Colegio de Ciencias de la Salud COCSA, Universidad San Francisco de Quito USFQ, Quito, Ecuador
- Sistemas Médicos SIME, Universidad San Francisco de Quito USFQ, Quito, Ecuador
- Mito-Act Research Consortium, Quito, Ecuador
| | - Verónica Castañeda
- Faculty of Medicine, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Molecular Biology and Bioinformatics Lab, Program in Molecular Biology and Bioinformatics, Center for Biomedical Research and Innovation (CIIB), Universidad de los Andes, Santiago, Chile
| | | | - Christopher R Chin
- Department of Physiology and Biophysics and Tri-Institutional Computational Biology and Medicine Program, Weill Cornell Medicine, New York, NY, USA
| | | | - Sylvain V Costes
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - Iwijn De Vlaminck
- Department of Physiology and Biophysics and Tri-Institutional Computational Biology and Medicine Program, Weill Cornell Medicine, New York, NY, USA
| | - Rajeev I Desai
- Integrative Neurochemistry Laboratory, Behavioral Biology Program, Department of Psychiatry, Harvard Medical School, Belmont, MA, USA
| | - Raja Dhir
- Seed Health, Venice, CA, USA
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Juan Esteban Diaz
- Data Science Institute, School of Business, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| | - Sofia M Etlin
- Department of Astrobiology, Cornell University, New York, NY, USA
| | - Zachary Feinstein
- Department of Physiology and Biophysics and Tri-Institutional Computational Biology and Medicine Program, Weill Cornell Medicine, New York, NY, USA
| | - David Furman
- Buck Institute for Research on Aging, Novato, CA, USA
- Stanford 1000 Immunomes Project, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Research in Translational Medicine, Universidad Austral, CONICET, Pilar, Argentina
| | - J Sebastian Garcia-Medina
- Department of Physiology and Biophysics and Tri-Institutional Computational Biology and Medicine Program, Weill Cornell Medicine, New York, NY, USA
| | - Francine Garrett-Bakelman
- Department of Physiology and Biophysics and Tri-Institutional Computational Biology and Medicine Program, Weill Cornell Medicine, New York, NY, USA
| | - Stefania Giacomello
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | | | - Amira Hassanin
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Nadia Houerbi
- Department of Physiology and Biophysics and Tri-Institutional Computational Biology and Medicine Program, Weill Cornell Medicine, New York, NY, USA
| | - Iris Irby
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Emilia Javorsky
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Future of Life Institute, Campbell, CA, USA
| | - Peter Jirak
- Paracelsus Medical University, Salzburg, Austria
- Department of Internal Medicine, Hospital Gmünd, Lower Austria, Austria
| | - Christopher W Jones
- Unit for Experimental Psychiatry, Division of Sleep and Chronobiology, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Khaled Y Kamal
- Redox Biology and Cell Signaling Laboratory, Department of Kinesiology and Sport Management, Texas A&M University, College Station, TX, USA
- Department of Kinesiology, Iowa State University, Ames, USA
| | - Brian D Kangas
- Behavioral Biology Program, Department of Psychiatry, Harvard Medical School, Belmont, MA, USA
| | - Fathi Karouia
- Blue Marble Institute of Science, Exobiology Branch NASA Ames Research Center, Moffett Field, CA, USA
- Space Research Within Reach, San Francisco, CA, USA
- Center for Space Medicine, Baylor College of Medicine, Houston, TX, USA
- BioServe Space Technologies, Smead Aerospace Engineering Science Department, University of Colorado Boulder, Boulder, CO, USA
| | - JangKeun Kim
- Department of Physiology and Biophysics and Tri-Institutional Computational Biology and Medicine Program, Weill Cornell Medicine, New York, NY, USA
| | - Joo Hyun Kim
- Redox Biology and Cell Signaling Laboratory, Department of Kinesiology and Sport Management, Texas A&M University, College Station, TX, USA
| | - Ashley S Kleinman
- Department of Physiology and Biophysics and Tri-Institutional Computational Biology and Medicine Program, Weill Cornell Medicine, New York, NY, USA
| | - Try Lam
- Jet Propulsion Laboratory, California Institute of Technology, Pasadena, CA, USA
| | - John M Lawler
- Redox Biology and Cell Signaling Laboratory, Department of Kinesiology and Sport Management, Texas A&M University, College Station, TX, USA
| | - Jessica A Lee
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, CA, USA
| | - Alexander Lucaci
- Department of Physiology and Biophysics and Tri-Institutional Computational Biology and Medicine Program, Weill Cornell Medicine, New York, NY, USA
| | - Matthew MacKay
- Department of Physiology and Biophysics and Tri-Institutional Computational Biology and Medicine Program, Weill Cornell Medicine, New York, NY, USA
| | - J Tyson McDonald
- Department of Radiation Medicine, Georgetown University School of Medicine, Washington, D.C., USA
| | - Ari M Melnick
- Department of Physiology and Biophysics and Tri-Institutional Computational Biology and Medicine Program, Weill Cornell Medicine, New York, NY, USA
| | - Cem Meydan
- Department of Physiology and Biophysics and Tri-Institutional Computational Biology and Medicine Program, Weill Cornell Medicine, New York, NY, USA
| | - Jakub Mieczkowski
- International Research Agenda 3P-Medicine Laboratory, Medical University of Gdansk, Gdansk, Poland
| | - Masafumi Muratani
- Department of Genome Biology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Deena Najjar
- Department of Physiology and Biophysics and Tri-Institutional Computational Biology and Medicine Program, Weill Cornell Medicine, New York, NY, USA
| | - Mariam A Othman
- Redox Biology and Cell Signaling Laboratory, Department of Kinesiology and Sport Management, Texas A&M University, College Station, TX, USA
| | - Eliah G Overbey
- Department of Physiology and Biophysics and Tri-Institutional Computational Biology and Medicine Program, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
- BioAstra, New York, NY, USA
| | - Vera Paar
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University, Salzburg, Austria
| | - Jiwoon Park
- Department of Physiology and Biophysics and Tri-Institutional Computational Biology and Medicine Program, Weill Cornell Medicine, New York, NY, USA
| | - Amber M Paul
- Blue Marble Space Institute of Science, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
- Embry-Riddle Aeronautical University, Department of Human Factors and Behavioral Neurobiology, Daytona Beach, FL, USA
| | - Adrian Perdyan
- International Research Agenda 3P-Medicine Laboratory, Medical University of Gdansk, Gdansk, Poland
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Jacqueline Proszynski
- Department of Physiology and Biophysics and Tri-Institutional Computational Biology and Medicine Program, Weill Cornell Medicine, New York, NY, USA
| | - Robert J Reynolds
- University of Texas Medical Branch, Galveston, TX, USA
- KBR, Inc., Houston, TX, USA
| | - April E Ronca
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
- Wake Forest Medical School, Dept of Obstetrics and Gynecology, Winston-Salem, NC, USA
| | | | - Krista A Ryon
- Department of Physiology and Biophysics and Tri-Institutional Computational Biology and Medicine Program, Weill Cornell Medicine, New York, NY, USA
| | - Lauren M Sanders
- Blue Marble Space Institute of Science, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | | | - Yash Shevde
- Ursa Biotechnology Corporation, Ursa Bio, New York, NY, USA
| | | | - Ryan T Scott
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - Bader Shirah
- Department of Neuroscience, King Faisal Specialist Hospital and Research Centre, Jeddah, Saudi Arabia
| | - Karolina Sienkiewicz
- Department of Physiology and Biophysics and Tri-Institutional Computational Biology and Medicine Program, Weill Cornell Medicine, New York, NY, USA
| | - Maria A Sierra
- Department of Physiology and Biophysics and Tri-Institutional Computational Biology and Medicine Program, Weill Cornell Medicine, New York, NY, USA
| | - Keith Siew
- London Tubular Centre, Department of Renal Medicine, University College London, London, UK
| | | | - Braden T Tierney
- Department of Physiology and Biophysics and Tri-Institutional Computational Biology and Medicine Program, Weill Cornell Medicine, New York, NY, USA
| | | | - Jeremy Wain Hirschberg
- Department of Physiology and Biophysics and Tri-Institutional Computational Biology and Medicine Program, Weill Cornell Medicine, New York, NY, USA
| | - Stephen B Walsh
- London Tubular Centre, Department of Renal Medicine, University College London, London, UK
| | - Claire Walter
- Department of Physiology and Biophysics and Tri-Institutional Computational Biology and Medicine Program, Weill Cornell Medicine, New York, NY, USA
| | - Daniel A Winer
- Buck Institute for Research on Aging, Novato, CA, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Division of Cellular and Molecular Biology, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Min Yu
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Luis Zea
- Smead Aerospace Engineering Sciences Department, University of Colorado Boulder, Boulder, CO, USA
- Jaguar Space, LLC, Erie, CO, USA
| | - Jaime Mateus
- Space Exploration Technologies Corporation (SpaceX), Hawthorne, CA, USA
| | - Afshin Beheshti
- Blue Marble Space Institute of Science, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|