1
|
Ge W, Mu Z, Yang S, Zeng Y, Deng Y, Lin Y, Xie P, Li G. Biosensor-based methods for exosome detection with applications to disease diagnosis. Biosens Bioelectron 2025; 279:117362. [PMID: 40157151 DOI: 10.1016/j.bios.2025.117362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/09/2025] [Accepted: 03/09/2025] [Indexed: 04/01/2025]
Abstract
Exosomes are nanoscale extracellular vesicles (EVs) secreted by most eukaryotic cells and can be found in nearly all human body fluids. Increasing evidence has revealed their pivotal roles in intercellular communication, and their active participation in myriad physiological and pathological activities. Exosomes' functions rely on their contents that are closely correlated with the biological characteristics of parental cells, which may provide a rich resource of molecular information for accurate and detailed diagnosis of a diverse array of diseases, such as differential diagnosis of Alzheimer's disease, early detection and subtyping of various tumors. As a category of sensitive detection devices, biosensors can fully reveal the molecular information and convert them into actionable clinical information. In this review, recent advances in biosensor-based methods for the detection of exosomes are summarized. We have described the fabrication of various biosensors based on the analysis of exosomal proteins, RNAs or glycans for accurate diagnosis, with respect to their elaborate recognition designs, signal amplification strategies, sensing properties, as well as their application potential. The challenges along with corresponding technologies in the future development and clinical translation of these biosensors are also discussed.
Collapse
Affiliation(s)
- Weikang Ge
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing 210023, People's Republic of China
| | - Zheying Mu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing 210023, People's Republic of China
| | - Shiao Yang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing 210023, People's Republic of China
| | - Yujing Zeng
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, People's Republic of China
| | - Ying Deng
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing 210023, People's Republic of China
| | - Yifan Lin
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| | - Ping Xie
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China.
| | - Genxi Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing 210023, People's Republic of China; Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, People's Republic of China.
| |
Collapse
|
2
|
Orange F, Pagnotta S, Pierre O, de Almeida Engler J. Application of array tomography to elucidate nuclear clustering architecture in giant-feeding cells induced by root-knot nematodes. THE NEW PHYTOLOGIST 2025; 246:2346-2369. [PMID: 40186428 DOI: 10.1111/nph.70066] [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: 09/13/2024] [Accepted: 02/13/2025] [Indexed: 04/07/2025]
Abstract
Plant-parasitic nematodes like root-knot nematodes (RKN; Meloidogyne spp.) cause great losses in agriculture by inducing root swellings, named galls, in host roots disturbing plant growth and development. Previous two-dimensional studies using different microscopy techniques revealed the presence of numerous nuclear clusters in nematode-induced giant cells within galls. Here, we show in three dimensions (3D) that nuclear clustering occurring in giant cells is revealed to be much more complex, illustrating subclusters built of multiple nuclear lobes. These nuclear subclusters are unveiled to be interconnected and likely communicate via nucleotubes, highlighting the potential relevance of this nuclear transfer for disease. In addition, microtubules and microtubule organizing centers are profusely present between the densely packed nuclear lobes, suggesting that the cytoskeleton might be involved in anchoring nuclear clusters in giant cells. This study illustrates that it is possible to apply volume electron microscopy (EM) approaches such as array tomography (AT) to roots infected by nematodes using basic equipment found in most EM facilities. The application of AT was valuable to observe the cellular ultrastructure in 3D, revealing the remarkable nuclear architecture of giant cells in the model host Arabidopsis thaliana. The discovery of nucleotubes, as a unique component of nuclear clusters present in giant cells, can be potentially exploited as a novel strategy to develop alternative approaches for RKN control in crop species.
Collapse
Affiliation(s)
- François Orange
- Centre Commun de Microscopie Appliquée (CCMA), Université Côte d'Azur, 06108, Nice, France
| | - Sophie Pagnotta
- Centre Commun de Microscopie Appliquée (CCMA), Université Côte d'Azur, 06108, Nice, France
| | - Olivier Pierre
- INRAE, Université Côte d'Azur, CNRS, ISA, 06903, Sophia Antipolis, France
| | | |
Collapse
|
3
|
Yu X, Cao S, Deng X, Chen Y, Sun M, Zhao P, Zhang Q, Chen J, Chen JX. Enhancing early breast cancer detection with APE1-triggered oligonucleotide probes and graphene oxide: The impact of variable AP site modification on sensitivity and specificity. Talanta 2025; 287:127505. [PMID: 39862516 DOI: 10.1016/j.talanta.2024.127505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 12/24/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025]
Abstract
There is a critical need for inclusive diagnostic platforms to enhance the accuracy of early breast cancer detection. Dysregulated microRNA-1246 (miR-1246), closely linked to the disease progression and recurrence, has emerged as a promising diagnostic and prognostic biomarker for BC. However, achieving simple, rapid, and ultrasensitive quantification of serum miRNAs remains significant challenge. In this study, we present an innovative detection platform triggered by endogenous DNA repair enzyme apurinic/apyrimidinic endonuclease 1 (APE1). This platform utilizes an oligonucleotide probe with variable modified AP sites (denoted as AOP) coupled with graphene oxide (GO) for quantifying miR-1246. Our in vitro experiments reveal that the proposed method employing the AOP2 probe with two AP sites exhibits exceptional selectivity and sensitivity. The method achieves a detection limit as low as 2.3 pM towards miR-1246, which is approximately 260-fold more sensitive than the enzyme-free system. RT-qPCR experiments further validate the accuracy and practicability of the AOP2-based platform. In clinical trials, our platform has successfully differentiated between BC patients and normal healthy controls. In conclusion, we have established an integrated biosensing technology for PCR-free, non-invasive liquid biopsies of miR-1246, offering a promising approach for BC diagnosis.
Collapse
Affiliation(s)
- Xuezhao Yu
- Office of Clinical Trial of Drug, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510663, China
| | - Sujian Cao
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, 510005, China
| | - Xuexian Deng
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Yanyan Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Mengxu Sun
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Pei Zhao
- Office of Clinical Trial of Drug, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510663, China
| | - Qun Zhang
- Office of Clinical Trial of Drug, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510663, China.
| | - Jun Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| | - Jin-Xiang Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
4
|
Doghish AS, Mansour RM, Mageed SSA, Moustafa HAM, Mohammed OA, Elesawy AE, Rizk NI. Role and Significance of MicroRNAs in the Relationship Between Obesity and Cancer. Balkan Med J 2025; 42:188-200. [PMID: 40326803 PMCID: PMC12060578 DOI: 10.4274/balkanmedj.galenos.2025.2025-3-60] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Accepted: 03/26/2025] [Indexed: 05/07/2025] Open
Abstract
MicroRNAs (miRNAs) are small, noncoding RNAs that are essential for regulating gene expression at the posttranscriptional stage. Recent research shows that miRNAs are crucial in the development of two major global health issues: obesity and cancer, two significant health issues worldwide. This study examines the complex mechanisms by which miRNAs govern vital biological processes, including adipogenesis, cancer, and metabolic dysregulation. We highlight the dual function of miRNAs as oncogenes and tumor suppressors in obesity-related malignancies and investigate their potential as prognostic and diagnostic markers. To demonstrate their varied roles, specific examples of vital miRNAs are underscored, such as miR-21, which promotes adipogenesis and is overexpressed in various cancers, and miR-34a, a tumor suppressor involved in cell cycle arrest and apoptosis. In addition, we examined the recent developments in miRNA-based therapies, which include miRNA inhibitors, mimics, and novel delivery vehicles and have the potential for treating obesity-related malignancies. This review aims to clarify, within the framework of miRNA biology, the therapeutic potential of miRNAs in addressing the interrelation between obesity and cancer.
Collapse
Affiliation(s)
- Ahmed S. Doghish
- Department of Biochemistry and Molecular Biology, Al-Azhar University Faculty of Pharmacy, Cairo, Egypt
- Department of Biochemistry, Badr University in Cairo Faculty of Pharmacy, Cairo, Egypt
| | - Reda M. Mansour
- Department of Zoology and Entomology, Helwan University Faculty of Science, Heiwan, Egypt
- Department of Molecular Biology and Biotechnology, Badr University in Cairo School of Biotechnology, Cairo, Egypt
| | - Sherif S. Abdel Mageed
- Department of Pharmacology and Toxicology, Badr University in Cairo Faculty of Pharmacy, Cairo, Egypt
| | | | - Osama A. Mohammed
- Department of Pharmacology, University of Bisha College of Medicine, Bisha, Saudi Arabia
| | - Ahmed E. Elesawy
- Department of Biochemistry, Badr University in Cairo Faculty of Pharmacy, Cairo, Egypt
| | - Nehal I. Rizk
- Department of Biochemistry, Egyptian Chinese University Faculty of Pharmacy and Drug Technology, Cairo, Egypt
| |
Collapse
|
5
|
Guerra-Andrés M, Fernández ÁF, Fontanil T. Exosomes, autophagy, and cancer: A complex triad. Int J Cancer 2025. [PMID: 40318053 DOI: 10.1002/ijc.35388] [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: 11/22/2024] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 05/07/2025]
Abstract
Cancer remains one of the leading causes of death worldwide. Despite remarkable progress in prevention, diagnosis, and therapy, the incidence of certain types of cancer persists, urging the identification of clinically relevant biomarkers and the development of novel therapeutic strategies to improve clinical outcomes and overcome treatment resistance. Exosomes, small extracellular vesicles released by diverse types of cells, have attracted interest in biomedical research due to their potential as carriers for different treatments. Moreover, exosomes play a pivotal role in intercellular communication, modulating various cellular processes. One of those is autophagy, a pro-survival pathway that is essential for human cells. Even though autophagy is traditionally described as a catabolic route, its machinery is intricately involved in various cellular responses, including vesicle formation and secretion. In this regard, the link between autophagy and exosomes is complex, bidirectional, and highly dependent on the cellular context. Interestingly, both processes have been extensively implicated in cancer pathogenesis, highlighting their potential as therapeutic targets. This review updates our understanding of how exosomes can participate in cancer development and progression, with a specific focus on their influence on tumor growth, angiogenesis, and metastasis. Additionally, the interplay between these extracellular vesicles and autophagy is minutely reviewed and discussed, as we hypothesize that this crosstalk may hold valuable clues for biomarker discovery and the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- María Guerra-Andrés
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Oviedo, Spain
| | - Álvaro F Fernández
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Tania Fontanil
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
- Instituto Ordoñez (Astracime S.L), Oviedo, Spain
- Lovinium Biocell CO LTD., Bangkok, Thailand
| |
Collapse
|
6
|
Jamouss KT, Damanakis AI, Cornwell AC, Jongepier M, Trujillo MA, Pflüger MJ, Kawalerski R, Maalouf A, Hirose K, Datta S, Sipes A, Pedro BA, Gudmundsson E, Assarzadegan N, Engle L, Scharpf RB, Kawamoto S, Thompson ED, Wood LD. Tumor immune microenvironment alterations associated with progression in human intraductal papillary mucinous neoplasms. J Pathol 2025; 266:40-50. [PMID: 40001347 DOI: 10.1002/path.6402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 11/26/2024] [Accepted: 01/08/2025] [Indexed: 02/27/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) poses a significant challenge due to late-stage diagnoses. To improve patient outcomes, early intervention in precursor lesions such as intraductal papillary mucinous neoplasm (IPMN) is crucial. However, early intervention must be balanced against overtreatment of low-risk lesions that are unlikely to progress, underscoring the need to better understand molecular alterations in neoplastic cells and changes in the tumor microenvironment (TME) that drive the progression of IPMNs. In this study, we characterized alterations in the TME of IPMNs as they progressed to high-grade dysplasia, using immunohistochemistry to quantify immune cell density and activation status in more than 100 well-characterized human IPMN samples. Analyses revealed progression to a more immunosuppressive TME in high-grade IPMN compared with low-grade IPMN, characterized by elevated expression of immune checkpoint molecules (PD-L1, TIM3, VISTA), increased density of macrophages, and decreased density of cytotoxic T cells. Intriguingly, the alterations in macrophages were limited to focal regions of high-grade dysplasia, while T-cell alterations affected the entire IPMN. Additionally, elevated VISTA expression was associated with poorer clinical outcome after IPMN resection in an independent cohort. These findings provide important insights into the interplay between the immune microenvironment and IPMN progression, highlighting potential targets to modify the TME for cancer interception. © 2025 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Kevin T Jamouss
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alexander Ioannis Damanakis
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Abigail C Cornwell
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Martine Jongepier
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Maria A Trujillo
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael Johannes Pflüger
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Graduate School of Life Sciences, Utrecht University, Utrecht, The Netherlands
| | - Ryan Kawalerski
- Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alexandre Maalouf
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Katsuya Hirose
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shalini Datta
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Abigail Sipes
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Brian A Pedro
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Emma Gudmundsson
- Department of Physiology, University of Maryland, Baltimore, MD, USA
| | - Naziheh Assarzadegan
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pathology, University of Florida, Gainesville, FL, USA
| | - Logan Engle
- Bloomberg Kimmel Institute, Tumor Microenvironment Technology Development Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert B Scharpf
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Satomi Kawamoto
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elizabeth D Thompson
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Laura D Wood
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
7
|
Caluwé AD, Bellal S, Cao K, Peignaux K, Remouchamps V, Baten A, Longton E, Bessieres I, Vu-Bezin J, Kirova Y, Gestel DV, Desmoulins I, Ignatiadis M, Romano E, Buisseret L, Piccart M, Vandekerkhove C, Gulyban A, Poortmans P. Adapting radiation therapy to immunotherapy: Delineation and treatment planning of pre-operative immune-modulating breast iSBRT in 151 patients treated in the randomized phase II Neo-CheckRay trial. Radiother Oncol 2025; 206:110836. [PMID: 40057199 DOI: 10.1016/j.radonc.2025.110836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 02/17/2025] [Accepted: 03/01/2025] [Indexed: 04/20/2025]
Abstract
BACKGROUND AND PURPOSE The randomized multicentric phase II Neo-CheckRay trial investigated preoperative immune-modulating stereotactic body radiation therapy (iSBRT) 8 Gy x 3 fractions in combination with chemotherapy with or without the anti PD-L1 durvalumab and the anti-CD73 oleclumab in early-stage, high-risk, luminal B breast cancer. iSBRT was solely delivered to the primary breast cancer leveraging on its immune modulating potential to sustain an anti-tumour response. To avoid immunosuppression induced by radiation therapy (RT), the tumour draining lymph nodes (TDLN) were spared. Here, we present the constraints used in the Neo-CheckRay trial and a dosimetric analysis of all delivered treatment plans with a special focus on the dose to the TDLN. MATERIALS AND METHODS Main constraints were the skin (D0.1 cc < 19.2 Gy), chest wall (D1cc < 15 Gy) and ipsilateral uninvolved breast (V24Gy < 30 %). The dose to the TDLN was reduced by avoiding beams entering or exiting the TDLN. In the present work, the DICOM-RT data of all the patients treated in the Neo-CheckRay trial were collected (n = 151) to describe doses to the target volume, to the organs at risk and the TDLN. The TDLN volumes consisted of the internal mammary nodes (IMN) and the axilla levels I-IV including the interpectoral nodes. RESULTS In 151 patients, the median V95% of the gross target volume (GTV) and planning target volume (PTV) were 97.4 % (90 % CI 26.5-100) and 95.5 % (56.1-100). The mean dose (dMean) to all TDLN volumes was < 1 Gy. The highest dMean were to the IMN and axilla level 1: 0.8 Gy (90 % CI 0.1-2.7) and 0.6 Gy (0.0-3.9), respectively. The dMean to the involved lymph nodes, if present, was 0.3 Gy (0.0-5.0). CONCLUSION In the Neo-CheckRay trial, the predefined organs at risk dose constraints were feasible and the TDLN were adequately spared.
Collapse
Affiliation(s)
- A De Caluwé
- Jules Bordet Institute, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium.
| | - S Bellal
- Jules Bordet Institute, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - K Cao
- Institut Curie, Paris, France
| | - K Peignaux
- Centre George François Leclerc, Dijon, France
| | - V Remouchamps
- Clinique Hospitalier Universitaire CHU UCL St. Elisabeth, Namur, Belgium
| | - A Baten
- Universitaire Ziekenhuizen Leuven, Leuven, Belgium
| | - E Longton
- Cliniques Universitaires St. Luc, Brussels, Belgium
| | - I Bessieres
- Centre George François Leclerc, Dijon, France
| | | | | | - D Van Gestel
- Jules Bordet Institute, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | | | - M Ignatiadis
- Jules Bordet Institute, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | | | - L Buisseret
- Jules Bordet Institute, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - M Piccart
- Jules Bordet Institute, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - C Vandekerkhove
- Jules Bordet Institute, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - A Gulyban
- Jules Bordet Institute, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - P Poortmans
- Iridium Netwerk and University of Antwerp, Antwerp, Belgium
| |
Collapse
|
8
|
Wu H, Liu J, Zhang XH, Jin S, Li P, Liu H, Zhao L, Wang J, Zhao S, Tian HD, Lai JR, Hao Y, Liu GR, Hou K, Yan M, Liu SL, Pang D. The combination of flaxseed lignans and PD-1/ PD-L1 inhibitor inhibits breast cancer growth via modulating gut microbiome and host immunity. Drug Resist Updat 2025; 80:101222. [PMID: 40048957 DOI: 10.1016/j.drup.2025.101222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 02/22/2025] [Accepted: 02/22/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND Patients with breast cancer (BC) who benefit from the PD-1/PD-L1 inhibitor (PDi) is limited, necessitating novel strategies to improve immunotherapy efficacy of BC. Here we aimed to investigate the inhibitory effects of flaxseed lignans (FL) on the biological behaviors of BC and evaluate the roles of FL in enhancing the anticancer effects of PDi. METHODS HPLC was used to detect the content of enterolactone (ENL), the bacterial transformation product of FL. Transcript sequencing was performed and identified CD38 as a downstream target gene of ENL. CD38-overexpressing cells were constructed and cell proliferation, colony formation, wound healing and transwell assays were used to assess the function of ENL/CD38 axis on BC cells in vitro. Multiplexed immunohistochemistry (mIHC) and CyTOF were used to detect the changes of the tumor immune microenvironment (TIM). 16S rDNA sequencing was used to explore the changes of gut microbiota in mice. A series of in vivo experiments were conducted to investigate the anticancer effects and mechanisms of FL and PDi. RESULTS FL was converted to ENL by gut microbiota and FL administration inhibited the progression of BC. ENL inhibited the malignant behaviors of BC by downregulating CD38, a key gene associated with immunosuppression and PD-1/PD-L1 blockade resistance. The mIHC assay revealed that FL administration enhanced CD3+, CD4+ and CD8+ cells and reduced F4/80+ cells in TIM. CyTOF confirmed the regulatory effects of FL and FL in combination with PDi (FLcPDi) on TIM. In addition, 16S rDNA analysis demonstrated that FLcPDi treatment significantly elevated the abundance of Akkermansia and, importantly, Akkermansia administration enhanced the response to PDi in mice treated with antibiotics. CONCLUSIONS The FL/ENL/CD38 axis inhibited BC progression. FL enhanced the anticancer effects of PDi by modulating gut microbiota and host immunity.
Collapse
Affiliation(s)
- Hao Wu
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China.
| | - Jiena Liu
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Xing-Hua Zhang
- Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150081, China
| | - Shengye Jin
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ping Li
- The Third Affiliated Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Huidi Liu
- Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150081, China; Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
| | - Liuying Zhao
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Jianyu Wang
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Shilu Zhao
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Hong-Da Tian
- Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150081, China
| | - Jin-Ru Lai
- Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150081, China
| | - Yi Hao
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Gui-Rong Liu
- Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150081, China
| | - Kaijian Hou
- School of Public Health, Shantou University, Shantou, China; Longhu People's Hospital, Shantou, China.
| | - Meisi Yan
- Department of Pathology, Harbin Medical University, Harbin, China.
| | - Shu-Lin Liu
- Genomics Research Center, State Key Laboratory of Biomedicine-Pharmaceutics of China, College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, Harbin Medical University, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China; National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin 150081, China; Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada.
| | - Da Pang
- Heilongjiang Clinical Research Center for Breast Cancer, Harbin Medical University Cancer Hospital, Harbin, China; Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
9
|
Kang Y, Cao X, Fan Y, Li Y, Xu T, Zhou Q, He B. Exosome biomarkers in breast cancer: Systematic review and meta-analysis. Clin Chim Acta 2025; 574:120342. [PMID: 40311726 DOI: 10.1016/j.cca.2025.120342] [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: 03/31/2025] [Revised: 04/28/2025] [Accepted: 04/28/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND Breast cancer (BC) has become the primary cancer that threatens women's health and life expectancy. Early diagnosis is crucial for effective treatment and favourable prognosis. As a non-invasive and valuable liquid biopsy method, exosomes are promising for the diagnosis and prognosis of BC. The aim of this meta-analysis is to evaluate the diagnostic and prognostic value of exosome biomarkers in BC. METHODS A systematic search of relevant English literature was conducted in PubMed, Web of Science, and Cochrane library until August 2024 (diagnosis) and October 2024 (prognosis). QUADAS-2 and QUAPAS were used to assess the quality of the literature. Summary statistics and analyses of relevant effect sizes were conducted using STATA software. Subgroup analysis and sensitivity analysis were performed to identify potential sources of heterogeneity. RESULTS For diagnosis, a total of 31 articles with 3,778 patients and 2,722 controls were included, the pooled sensitivity (SEN), specificity (SPE), and area under the receiver operating characteristic curve (AUC) of overall exosome biomarkers were 0.89 (95 %CI: 0.86-0.91), 0.87 (95 %CI: 0.85-0.90), and 0.94 (95 %CI: 0.92-0.96), respectively, indicating a high diagnostic value of exosomes in BC patients. Subgroup analysis suggested that miRNAs in exosomes exhibited better diagnostic value compared to proteins and non-miRNAs, the SEN, SPE, and AUC were 0.89 (95 %CI: 0.82-0.93), 0.86 (95 %CI: 0.80-0.90), and 0.92 (95 %CI: 0.90-0.94), respectively. Among all miRNAs, the pooled SEN, SPE, and AUC of miR-21 were 0.86 (95 %CI: 0.67-0.95), 0.90 (95 %CI: 0.78-0.96), and 0.95 (95 %CI: 0.92-0.96), respectively. The diagnostic efficiency was improved when biomarkers were combined as a panel (SEN 0.91 versus 0.87, SPE 0.89 versus 0.86, AUC 0.96 versus 0.91). In terms of prognosis, we retrieved 14 articles with 2,781 patients. The pooled HR of overall survival (OS) and progression-free survival (PFS) were 1.41 (95 %CI: 0.92-1.90) and 4.39 (95 %CI: 1.87-6.91), respectively, indicating exosome biomarkers like soluble HLA-G, miR-1246, miR-155, and PSMA were a predictor of poor PFS in BC patients. Subgroup analysis in OS group revealed a significant association between the overexpression of exosome proteins (soluble HLA-G, AnxA2, NGF, CXCL13) and worse OS in BC patients (HR = 2.91, 95 %CI: 1.36-4.47). Similarly, the overexpression of miR-1246 and miR-155 was associated with worse PFS in BC patients (HR = 4.13, 95 %CI: 1.24-7.03). Moreover, when biomarkers were combined as a panel, the prognostic efficiency significantly improved in OS (HR = 4.05, 95 %CI: 2.26-5.84) outcome. CONCLUSION The meta-analysis revealed that exosome miR-21 might serve as a promising diagnostic biomarker in BC. Dysregulated exosome proteins and miRNAs could predict poor OS and PFS outcomes, respectively.
Collapse
Affiliation(s)
- Yurou Kang
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xiaoqing Cao
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yujing Fan
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China; Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yimin Li
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China; Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Tao Xu
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.
| | - Qing Zhou
- NHC Key Laboratory of Contraceptives Vigilance and Fertility Surveillance, Jiangsu Health Development Research Center, Jiangsu Provincial Medical Key Laboratory of Fertility Protection and Health Technology Assessment, NO.277 Fenghuang West Street, Nanjing, China.
| | - Bangshun He
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
10
|
Ntalouka MP, Symeonidis D, Kotsi P, Petinaki E, Matsagkas M, Tepetes K, Zacharoulis D, Arnaoutoglou EM. Venous thromboembolism in patients undergoing pancreatic cancer surgery (PaTR-VTE) with curative intent; protocol of a prospective observational study. BMC Surg 2025; 25:183. [PMID: 40287701 PMCID: PMC12032706 DOI: 10.1186/s12893-024-02665-z] [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: 07/03/2024] [Accepted: 11/11/2024] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Pancreatic cancer is probably the most thrombotic malignancy, with an incidence of venous thromboembolism (VTE) of up to 18%. However, the exact pathophysiological mechanisms involved in the development of VTE in the setting of pancreatic cancer are not yet well understood. The primary endpoint of the study is to evaluate the neutrophil/lymphocyte ratio (NLR) and other coagulation biomarkers as predictors of VTE in patients with pancreatic cancer undergoing surgery with curative intent. The exact incidence of VTE, perioperative coagulation status of patients and the possible determinants of VTE in the aforementioned population are the secondary study objectives. METHODS This prospective, non-interventional observational study is conducted according to the STROBE concept. It has been approved by the ethical committee and registered (NCT05964621) and will include eligible patients with primary pancreatic cancer with resectable or borderline resectable disease undergoing surgery with curative intent. Exclusion criteria are: Refusal to participate, previous thromboembolic event < 6 months, ASA score > 3, patients deemed inoperable intraoperatively and the concurrent presence of a second primary malignancy. Three blood samples are taken from all patients (preoperatively, immediately after the operation, and before discharge) and the serum values of the following parameters are determined: Haemoglobin, white blood cells, INR, liver and kidney function tests, von Willebrand factor, factors VIII and XI, D-dimers, fibrinogen, platelet function, Adamts 13 and anti-Xa. One month after the procedure, scheduled screening for asymptomatic deep vein thrombosis (DVT) is performed with a lower extremity ultrasound triplex study. In addition, thromboembolic events (DVT, pulmonary embolism (PE)) diagnosed during the hospital stay period are recorded. Low molecular weight heparin will be routinely administered from the first postoperative day, with the dosage, i.e. prophylactic or therapeutic titrated according to the patient's history of cardiovascular disease. According to the literature, the pooled specificity of the admission NLR for 30-day VTE and PE prediction is 80.5%, while the VTE rates after pancreatectomy is 1.5%. Based on a 95% confidence level and a precision of 0.1, the estimated sample size for the specificity outcome is 62 patients. DISCUSSION The aim of this study is to identify predictors of postoperative VTE in patients undergoing pancreatic cancer surgery. The results could lead to an optimization of perioperative care. TRIAL REGISTRATION NCT05964621. Registered on July, 2023 clinicaltrials.org.
Collapse
Affiliation(s)
- Maria P Ntalouka
- Department of Anaesthesiology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa University Hospital, Thessaly, Greece.
| | - Dimitrios Symeonidis
- Department of Surgery, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa University Hospital, Thessaly, Greece.
| | - Paraskevi Kotsi
- Department of Transfusion Medicine, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa University Hospital, Thessaly, Greece
| | - Efthymia Petinaki
- Department of Microbiology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa University Hospital, Thessaly, Greece
| | - Miltiadis Matsagkas
- Department of Vascular Surgery, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa University Hospital, Thessaly, Greece
| | - Konstantinos Tepetes
- Department of Surgery, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa University Hospital, Thessaly, Greece
| | - Dimitrios Zacharoulis
- Department of Surgery, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa University Hospital, Thessaly, Greece
| | - Eleni M Arnaoutoglou
- Department of Anaesthesiology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa University Hospital, Thessaly, Greece
| |
Collapse
|
11
|
Li H, Chiang CL, Kwak KJ, Lee HL, Wang X, Romano G, Saviana M, Toft R, Cheng TS, Chang Y, Hsiang BD, Liu GW, Mo X, Ma Y, Pan J, Rima XY, Kim TN, Reategui E, Shen CN, Chu YS, Croce C, Chang PMH, Yeh YC, Carbone DP, Huang CYF, Chiang CL, Nana-Sinkam P, Lee LJ. Extracellular Vesicular Delta-Like Ligand 3 and Subtype Transcription Factors for Small Cell Lung Cancer Diagnosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2416711. [PMID: 40285610 DOI: 10.1002/advs.202416711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 03/15/2025] [Indexed: 04/29/2025]
Abstract
Small cell lung cancer (SCLC) is associated with high mortality and limited therapeutic options. There is increasing recognition that SCLC harbors molecular heterogeneity. Using a new liquid biopsy assay, it is demonstrated that SCLC subtypes, as determined by patient tumor tissue staining and cell lines, can be accurately identified by measuring the mRNA expression of subtype transcription factors (ASCL1, POU2F3, and NEUROD1) in circulating exosome-rich extracellular vesicles (Exo). Additionally, upregulation of Delta-like ligand 3 (DLL3) mRNA in Exo and its membrane protein (mProtein) in extracellular vesicles associated with tumor (tEV) may distinguish both limited- and extensive-stage SCLC patients from high-risk smokers, with AUC/ROC values of 0.836 and 0.839, respectively. By incorporating Exo-ASCL1 and Exo-POU2F3 mRNA expression with DLL3 Exo-mRNA/tEV-mProtein expression, the classifier enhances the AUC/ROC to 0.912 and 0.963 for limited- and extensive-stage SCLC patients, respectively.
Collapse
Affiliation(s)
- Hong Li
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Chi-Ling Chiang
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, 43210, USA
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | | | - Hsin-Lun Lee
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei, 11031, Taiwan
- Genomic Research Center, Academia Sinica, Taipei, 11529, Taiwan
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, 11031, Taiwan
| | - Xinyu Wang
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Giulia Romano
- Division of Pulmonary Diseases and Critical Care Medicine, Virginia Commonwealth University, Richmond, VA, 23284, USA
| | - Michela Saviana
- Division of Pulmonary Diseases and Critical Care Medicine, Virginia Commonwealth University, Richmond, VA, 23284, USA
| | - Robin Toft
- Division of Pulmonary Diseases and Critical Care Medicine, Virginia Commonwealth University, Richmond, VA, 23284, USA
| | - Tai-Shan Cheng
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Yuehshih Chang
- Division of Hematology and Oncology, Department of Internal Medicine, Keelung Chang Gung Memorial Hospital, Keelung, 20401, Taiwan
- School of Medicine, College of Traditional Chinese Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
| | - Bi-Da Hsiang
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Guan-Wan Liu
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Xiaokui Mo
- Center for Biostatistics, The Ohio State University, Columbus, OH, 43210, USA
| | - Yifan Ma
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Junjie Pan
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Xilal Y Rima
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Truc Nguyen Kim
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Eduardo Reategui
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Chia-Ning Shen
- Genomic Research Center, Academia Sinica, Taipei, 11529, Taiwan
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, 11031, Taiwan
| | - Yeh-Shiu Chu
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Carlo Croce
- College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Peter Mu-Hsin Chang
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
- Department of Oncology, Taipei Veterans General Hospital, Taipei, 11217, Taiwan
| | - Yi-Chen Yeh
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, 11217, Taiwan
| | - David P Carbone
- College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Chi-Ying F Huang
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| | - Chi-Lu Chiang
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, 11217, Taiwan
| | - Patrick Nana-Sinkam
- Division of Pulmonary Diseases and Critical Care Medicine, Virginia Commonwealth University, Richmond, VA, 23284, USA
| | - L James Lee
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, 43210, USA
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan
| |
Collapse
|
12
|
Acar B, Yilmaz T, Yapar A. Optimization of microwave hyperthermia system for focused breast cancer treatment: A study using realistic digital breast phantoms. Med Phys 2025. [PMID: 40270084 DOI: 10.1002/mp.17836] [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: 11/08/2024] [Revised: 02/25/2025] [Accepted: 03/18/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Microwave breast hyperthermia is a noninvasive treatment method for breast cancer that utilizes microwave energy (ME) sources to raise tissue temperatures above 42∘ C $^{\circ }{\rm C}$ , inducing tumor cell necrosis. The efficiency of ME deposition depends on the electric field magnitude and tissue conductivity, with antenna phase and amplitude adjustments used to maximize the electric field magnitude within tumors. Achieving precise ME focusing in the complex and heterogeneous breast tissue is challenging and can lead to unwanted hot spots in normal tissue. This study presents a novel method for optimizing ME focusing on the center of target tumors, using a simplified calculation of antenna phases, heuristic optimization for antenna amplitudes, and realistic breast phantoms for performance evaluation. PURPOSE In this work, we propose an approach to optimize the microwave hyperthermia system, employing phase and amplitude modulation techniques to concentrate the electric field at the center of a malignant tumor within a breast medium. The approach uses line sources arranged in a circular pattern around realistic breast models. The method begins by determining the phase, followed by adjusting the amplitudes of each source in order to maximize the total electric field at the tumor's center. The goal is to maximize the electric field at the tumor center while minimizing the optimization cost and complexity. METHODS Simulations are performed at 4 GHz frequency using two different types of digital breast phantoms (fatty and dense breasts) as test beds. The algorithm is tested by using three quantities; that is, the electric field distribution, the power density distribution, and the temperature distribution inside the whole breast region. The electric field and power density are calculated using an in-house method of moments (MoM) algorithm, while the temperature distributions are obtained with computer simulation technology (CST) software. To further evaluate the method with quantitative measures of success, thermal indices are calculated for each phantom and method. RESULTS The specific absorbtion rate (SAR) results and corresponding temperature distributions for each breast type and optimization demonstrate that effective focusing is achieved in both cases. However, the combined phase-amplitude optimization provides more precise focusing by eliminating hot spots. Among thermal indices, the TC75 and T90 values obtained from the phase-amplitude combined optimization for both breast types outperform the results found in the literature. The T50 values obtained using the combined optimization are above 42C ∘ ${\rm C}^\circ$ . CONCLUSIONS This study presents an optimization method for focusing ME within breast tissue, performed in two steps: first phase optimization, followed by amplitude optimization. The electric field calculations are performed using both the MoM and Finite Difference Time Domain methods. The technique is numerically tested on two realistic breast models, with thermal indices calculated for each phantom and optimization process. Results show T90 values exceeding 40∘ C $^\circ{\rm C}$ and T50 values above 42∘ C $^\circ{\rm C}$ . While the study employs a 2D applicator, it provides a strong foundation for future development in 3D applications.
Collapse
Affiliation(s)
- Burak Acar
- Department of Electronics and Communication Engineering, Istanbul Technical University, Istanbul, Turkey
| | - Tuba Yilmaz
- Department of Electronics and Communication Engineering, Istanbul Technical University, Istanbul, Turkey
| | - Ali Yapar
- Department of Electronics and Communication Engineering, Istanbul Technical University, Istanbul, Turkey
| |
Collapse
|
13
|
Lee H, Lee J, Ko M, Lee KN, Kim Y, Seo B, Lee J, Jeong S, Heo K, Lee YK, Jung I, Do YR. Advanced Exosome Isolation through Electrophoretic Oscillation-Assisted Tangent-Flow Ultrafiltration with a PVDF-Fiber-Coated SiN x Nanofilter. ACS APPLIED BIO MATERIALS 2025; 8:2965-2976. [PMID: 40063836 DOI: 10.1021/acsabm.4c01821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
This study introduces a comprehensive approach to enhancing SiNx nanofilters for exosome isolation from bovine milk using the electrophoretic oscillation-assisted tangent-flow ultrafiltration (EPOTF) process. Reinforcing the nanofilter with electro-spun poly(vinylidene fluoride) (PVDF) fibers significantly improved durability under high-pressure conditions, withstanding nearly 2.8 times greater pressures than nonreinforced nanofilters. The PVDF-fiber-coated nanofilters achieved a flow rate of over 70 mL min-1, compared to just 25 mL min-1 for nonreinforced nanofilters. A filter housing system with copper electrodes isolated from the solution flow path further enhanced the electrical stability of the entire system, widening the EPO voltage range while reducing the risk of corrosion and contamination. The PVDF-fiber-coated nanofilter with the electrode in a separated housing efficiently prevented clogging and bioparticle agglomeration, maintaining constant filtration performance across various voltages and duty cycles. Biochemical analyses confirmed the high concentration and structural integrity of exosomes isolated at high flow rates. Long-term tests verified the superior performance of PVDF-coated filters, successfully filtering 3400 mL of milk over 24 h. These results demonstrate the potential of these advances for highly efficient exosome isolation while maintaining the integrity and shape of exosomes, offering promise for the future of exosome isolation research.
Collapse
Affiliation(s)
- Hansol Lee
- Department of Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Jaehyuk Lee
- Department of Mechanical Engineering, Kyung Hee University, Yongin 17104, Republic of Korea
- Advanced Institutes of Convergence Technology 8F, R&D Center, Metapore Co., Ltd., Suwon 16229, Republic of Korea
| | - Minji Ko
- Department of Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Keyong Nam Lee
- Department of Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Yeonjae Kim
- R&D Center, Incospharm Corp., Daejeon 34000, Republic of Korea
| | - Bosung Seo
- Advanced Institutes of Convergence Technology 8F, R&D Center, Metapore Co., Ltd., Suwon 16229, Republic of Korea
| | - Jungwon Lee
- Advanced Institutes of Convergence Technology 8F, R&D Center, Metapore Co., Ltd., Suwon 16229, Republic of Korea
| | - Sekyoo Jeong
- R&D Center, Incospharm Corp., Daejeon 34000, Republic of Korea
| | - Kyun Heo
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Young Kwang Lee
- Department of Chemistry and Biochemistry, San Diego State University, San Diego, California 92182, United States
| | - Inhwa Jung
- Department of Mechanical Engineering, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Young Rag Do
- Department of Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| |
Collapse
|
14
|
Xiao G, Wang X, Xu Z, Liu Y, Jing J. Lung-specific metastasis: the coevolution of tumor cells and lung microenvironment. Mol Cancer 2025; 24:118. [PMID: 40241074 PMCID: PMC12001740 DOI: 10.1186/s12943-025-02318-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 03/31/2025] [Indexed: 04/18/2025] Open
Abstract
The vast majority of cancer-related deaths are attributed to metastasis. The lung, being a common site for cancer metastasis, is highly prone to being a target for multiple cancer types and causes a heavy disease burden. Accumulating evidence has demonstrated that tumor metastasis necessitates continuous interactions between tumor cells and distant metastatic niches. Nevertheless, a comprehensive elucidation of the underlying mechanisms governing lung-specific metastasis still poses a formidable challenge. In this review, we depict the lung susceptibility and the molecular profiles of tumors with the potential for lung metastasis. Under the conceptual framework of "Reciprocal Tumor-Lung Metastatic Symbiosis" (RTLMS), we mechanistically delineate the bidirectional regulatory dynamics and coevolutionary adaptation between tumor cells and distal pulmonary niches during lung-specific metastasis, including the induction of pre-metastatic-niches, positive responses of the lung, tumor colonization, dormancy, and reawakening. An enhanced understanding of the latest mechanisms is essential for developing targeted strategies to counteract lung-specific metastasis.
Collapse
Affiliation(s)
- Guixiu Xiao
- Breast Disease Center and Institute for Breast Health Medicine, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xinmin Wang
- Institute of Breast Health Medicine, West China Hospital, Sichuan University Chengdu, Sichuan, 610041, China
| | - Zihan Xu
- Institute of Breast Health Medicine, West China Hospital, Sichuan University Chengdu, Sichuan, 610041, China
- Department of Medical Oncology, West China Hospital, Sichuan University, Cancer Center, Chengdu, Sichuan, 610041, China
| | - Yanyang Liu
- Department of Medical Oncology, West China Hospital, Sichuan University, Cancer Center, Chengdu, Sichuan, 610041, China.
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Jing Jing
- Breast Disease Center and Institute for Breast Health Medicine, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
15
|
Wang J, Tang J, Liang A, Cui Z, Huo J, Li Q, Ke B, Yang D, Yao C. A Smart DNA Network-Based Diagnostic System for Enrichment and Detection of Circulating Tumor Cells in Cancer Liquid Biopsy. Anal Chem 2025; 97:8065-8072. [PMID: 40185686 DOI: 10.1021/acs.analchem.5c00648] [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: 04/07/2025]
Abstract
Circulating tumor cells (CTCs) have emerged as critical biomarkers in liquid biopsy for noninvasive tumor diagnosis and real-time monitoring of cancer progression. However, the isolation of CTCs is often required before detection due to their ultralow abundance in peripheral blood. These isolation processes are typically time-consuming and prone to cell loss, which limits the utility of CTC-based liquid biopsy. Herein, we present a DNA network-based diagnostic system that enables specific recognition, selective enrichment, and accurate detection of CTCs directly from blood samples. The DNA network comprises ultralong DNA chains embedded with polyvalent aptamers and fluorescence detection modules. The polyvalent aptamers selectively bind to the epithelial cell adhesion molecule (EpCAM) on a CTC membrane, facilitating their enrichment through base pairing-driven DNA network formation. This system semiquantitatively detects the expression level of cancer-associated microRNA within CTCs using ratiometric fluorescence imaging based on the chemical assembly of two fluorescence modules. In clinical blood samples, this diagnostic system achieves 100% precision and 96% accuracy in distinguishing breast cancer patients from healthy donors, highlighting its promising potential for clinical breast cancer diagnosis.
Collapse
Affiliation(s)
- Jing Wang
- State Key Laboratory of Synthetic Biology, Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P.R. China
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, College of Chemistry and Materials, Fudan University, Shanghai 200438, P.R. China
| | - Jianpu Tang
- State Key Laboratory of Synthetic Biology, Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P.R. China
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, College of Chemistry and Materials, Fudan University, Shanghai 200438, P.R. China
| | - Aiqi Liang
- State Key Laboratory of Synthetic Biology, Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P.R. China
| | - Zhen Cui
- State Key Laboratory of Synthetic Biology, Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P.R. China
| | - Jiale Huo
- State Key Laboratory of Synthetic Biology, Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P.R. China
| | - Qian Li
- State Key Laboratory of Synthetic Biology, Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P.R. China
| | - Bin Ke
- Department of Gastric Surgery, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Dayong Yang
- State Key Laboratory of Synthetic Biology, Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P.R. China
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, College of Chemistry and Materials, Fudan University, Shanghai 200438, P.R. China
| | - Chi Yao
- State Key Laboratory of Synthetic Biology, Frontiers Science Center for Synthetic Biology, Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P.R. China
| |
Collapse
|
16
|
Delshad M, Sanaei MJ, Mohammadi MH, Sadeghi A, Bashash D. Exosomal Biomarkers: A Comprehensive Overview of Diagnostic and Prognostic Applications in Malignant and Non-Malignant Disorders. Biomolecules 2025; 15:587. [PMID: 40305328 PMCID: PMC12024574 DOI: 10.3390/biom15040587] [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: 02/19/2025] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 05/02/2025] Open
Abstract
Exosomes are small extracellular vesicles, ranging from 30 to 150 nm, that are essential in cell biology, mediating intercellular communication and serving as biomarkers due to their origin from cells. Exosomes as biomarkers for diagnosing various illnesses have gained significant investigation due to the high cost and invasive nature of current diagnostic procedures. Exosomes have a clear advantage in the diagnosis of diseases because they include certain signals that are indicative of the genetic and proteomic profile of the ailment. This feature gives them the potential to be useful liquid biopsies for real-time, noninvasive monitoring, enabling early cancer identification for the creation of individualized treatment plans. According to our analysis, the trend toward utilizing exosomes as diagnostic and prognostic tools has raised since 2012. In this regard, the proportion of malignant indications is higher compared with non-malignant ones. To be precise, exosomes have been used the most in gastrointestinal, thoracic, and urogenital cancers, along with cardiovascular, diabetic, breathing, infectious, and brain disorders. To the best of our knowledge, this is the first research to examine all registered clinical trials that look at exosomes as a diagnostic and prognostic biomarker.
Collapse
Affiliation(s)
- Mahda Delshad
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran; (M.D.); (M.-J.S.); (M.H.M.)
- Department of Laboratory Sciences, School of Allied Medical Sciences, Zanjan University of Medical Sciences, Zanjan 1411718541, Iran
| | - Mohammad-Javad Sanaei
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran; (M.D.); (M.-J.S.); (M.H.M.)
| | - Mohammad Hossein Mohammadi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran; (M.D.); (M.-J.S.); (M.H.M.)
| | - Amir Sadeghi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran 1985717411, Iran;
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran; (M.D.); (M.-J.S.); (M.H.M.)
| |
Collapse
|
17
|
Richard V, Lee K, Kerin MJ. MicroRNAs as Endocrine Modulators of Breast Cancer. Int J Mol Sci 2025; 26:3449. [PMID: 40244378 PMCID: PMC11989600 DOI: 10.3390/ijms26073449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/03/2025] [Accepted: 04/04/2025] [Indexed: 04/18/2025] Open
Abstract
Breast cancer is an aggressive disease of multiple subtypes with varying phenotypic, hormonal, and clinicopathological features, offering enhanced resistance to conventional therapeutic regimens. There is an unmet need for reliable molecular biomarkers capable of detecting the malignant transformation from the early stages of the disease to enhance diagnosis and treatment outcomes. A subset of small non-coding nucleic acid molecules, micro ribonucleic acids (microRNAs/miRNAs), have emerged as promising biomarkers due to their role in gene regulation and cancer pathogenesis. This review discusses, in detail, the different origins and hormone-like regulatory functionalities of miRNAs localized in tumor tissue and in the circulation, as well as their inherent stability and turnover that determines the utility of miRNAs as biomarkers for disease detection, monitoring, prognosis, and therapeutic targets.
Collapse
Affiliation(s)
- Vinitha Richard
- Discipline of Surgery, Lambe Institute for Translational Research, University of Galway, H91 V4AY Galway, Ireland
| | - Kevin Lee
- School of Medicine, University of Galway, H91 V4AY Galway, Ireland;
| | - Michael Joseph Kerin
- Discipline of Surgery, Lambe Institute for Translational Research, University of Galway, H91 V4AY Galway, Ireland
- School of Medicine, University of Galway, H91 V4AY Galway, Ireland;
| |
Collapse
|
18
|
Libring S, Reinhart-King CA. Premetastatic niche mechanics and organotropism in breast cancer. NPJ BIOLOGICAL PHYSICS AND MECHANICS 2025; 2:11. [PMID: 40191104 PMCID: PMC11968405 DOI: 10.1038/s44341-025-00015-5] [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: 09/05/2024] [Accepted: 02/14/2025] [Indexed: 04/09/2025]
Abstract
Numerous physical and mechanical changes occur in the premetastatic niche. Here, we review the mechanics of the premetastatic niche and how the altered extracellular matrix and cancer cell mechanics may play a role in organotropism in breast cancer. Future research into premetastatic niche development and organotropic cell behavior should address physical alterations and biomechanical effects to the same rigor that biochemical alterations are studied.
Collapse
Affiliation(s)
- Sarah Libring
- Department of Bioengineering, Rice University, Houston, TX USA
| | | |
Collapse
|
19
|
Dinh KN, Vázquez-García I, Chan A, Malhotra R, Weiner A, McPherson AW, Tavaré S. CINner: Modeling and simulation of chromosomal instability in cancer at single-cell resolution. PLoS Comput Biol 2025; 21:e1012902. [PMID: 40179124 PMCID: PMC11990800 DOI: 10.1371/journal.pcbi.1012902] [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: 06/03/2024] [Revised: 04/11/2025] [Accepted: 02/24/2025] [Indexed: 04/05/2025] Open
Abstract
Cancer development is characterized by chromosomal instability, manifesting in frequent occurrences of different genomic alteration mechanisms ranging in extent and impact. Mathematical modeling can help evaluate the role of each mutational process during tumor progression, however existing frameworks can only capture certain aspects of chromosomal instability (CIN). We present CINner, a mathematical framework for modeling genomic diversity and selection during tumor evolution. The main advantage of CINner is its flexibility to incorporate many genomic events that directly impact cellular fitness, from driver gene mutations to copy number alterations (CNAs), including focal amplifications and deletions, missegregations and whole-genome duplication (WGD). We apply CINner to find chromosome-arm selection parameters that drive tumorigenesis in the absence of WGD in chromosomally stable cancer types from the Pan-Cancer Analysis of Whole Genomes (PCAWG, [Formula: see text]). We found that the selection parameters predict WGD prevalence among different chromosomally unstable tumors, hinting that the selective advantage of WGD cells hinges on their tolerance for aneuploidy and escape from nullisomy. Analysis of inference results using CINner across cancer types in The Cancer Genome Atlas ([Formula: see text]) further reveals that the inferred selection parameters reflect the bias between tumor suppressor genes and oncogenes on specific genomic regions. Direct application of CINner to model the WGD proportion and fraction of genome altered (FGA) in PCAWG uncovers the increase in CNA probabilities associated with WGD in each cancer type. CINner can also be utilized to study chromosomally stable cancer types, by applying a selection model based on driver gene mutations and focal amplifications or deletions (chronic lymphocytic leukemia in PCAWG, [Formula: see text]). Finally, we used CINner to analyze the impact of CNA probabilities, chromosome selection parameters, tumor growth dynamics and population size on cancer fitness and heterogeneity. We expect that CINner will provide a powerful modeling tool for the oncology community to quantify the impact of newly uncovered genomic alteration mechanisms on shaping tumor progression and adaptation.
Collapse
Affiliation(s)
- Khanh N. Dinh
- Irving Institute for Cancer Dynamics, Columbia University, New York, New York, United States of America
- Department of Statistics, Columbia University, New York, New York, United States of America
| | - Ignacio Vázquez-García
- Irving Institute for Cancer Dynamics, Columbia University, New York, New York, United States of America
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
- Department of Pathology, Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Andrew Chan
- Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Rhea Malhotra
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
- Stanford University, Palo Alto, California, United States of America
| | - Adam Weiner
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
- Tri-Institutional PhD Program in Computational Biology and Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Andrew W. McPherson
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Simon Tavaré
- Irving Institute for Cancer Dynamics, Columbia University, New York, New York, United States of America
- Department of Statistics, Columbia University, New York, New York, United States of America
| |
Collapse
|
20
|
Karaca Dogan B, Salman Yilmaz S, Izgi GN, Ozen M. Circulating non-coding RNAs as a tool for liquid biopsy in solid tumors. Epigenomics 2025; 17:335-358. [PMID: 40040488 PMCID: PMC11970797 DOI: 10.1080/17501911.2025.2467021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 02/10/2025] [Indexed: 03/06/2025] Open
Abstract
Solid tumors are significant causes of global mortality and morbidity. Recent research has primarily concentrated on finding pathology-specific molecules that can be acquired non-invasively and that can change as the disease progresses or in response to treatment. The focus of research has moved to RNA molecules that are either freely circulating in body fluids or bundled in microvesicles and exosomes because of their great stability in challenging environments, ease of accessibility, and changes in level in response to therapy. In this context, there are many non-coding RNAs that can be used for this purpose in liquid biopsies. Out of these, microRNAs have been extensively studied. However, there has been an increase of interest in studying long non-coding RNAs, piwi interacting RNAs, circular RNAs, and other small non-coding RNAs. In this article, an overview of the most researched circulating non-coding RNAs in solid tumors will be reviewed, along with a discussion of the significance of these molecules for early diagnosis, prognosis, and therapeutic targets. The publications analyzed were extracted from the PubMed database between 2008 and June 2024.
Collapse
Affiliation(s)
- Beyza Karaca Dogan
- Department of Medical Genetics, Cerrahpaşa Faculty of Medicine, Istanbul University-Cerrahpaşa, Istanbul, Turkiye
| | - Seda Salman Yilmaz
- Department of Medical Genetics, Cerrahpaşa Faculty of Medicine, Istanbul University-Cerrahpaşa, Istanbul, Turkiye
- Department of Medical Services and Techniques Medical Monitoring Techniques Pr. Vocational School of Health Services, Istanbul University-Cerrahpaşa, Istanbul, Turkiye
| | - Gizem Nur Izgi
- Department of Medical Genetics, Cerrahpaşa Faculty of Medicine, Istanbul University-Cerrahpaşa, Istanbul, Turkiye
| | - Mustafa Ozen
- Department of Medical Genetics, Cerrahpaşa Faculty of Medicine, Istanbul University-Cerrahpaşa, Istanbul, Turkiye
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
21
|
Wu L, Yang J, Chen Y, Lin J, Huang W, Li M. Association of circulating metabolic biomarkers with risk of lung cancer: a population-based prospective cohort study. BMC Med 2025; 23:176. [PMID: 40140895 PMCID: PMC11948749 DOI: 10.1186/s12916-025-03993-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND There is emerging evidence that metabolites might be associated with risk of lung cancer, but their relationships have not been fully characterized. We aimed to investigate the association between circulating metabolic biomarkers and lung cancer risk and the potential underlying pathways. METHODS Nuclear magnetic resonance metabolomic profiling was conducted on baseline plasma samples from 91,472 UK Biobank participants without cancer and pregnancy. Multivariate Cox regression models were employed to assess the hazard ratios (HRs) of 164 metabolic biomarkers (including metabolites and lipoprotein subfractions) and 9 metabolic biomarker principal components (PCs) for lung cancer, after adjusting for covariates and false discovery rate (FDR). Pathway analysis was conducted to investigate the potential metabolic pathways. RESULTS During a median follow-up of 11.0 years, 702 participants developed lung cancer. A total of 109 metabolic biomarkers (30 metabolites and 79 lipoprotein subfractions) were associated with the risk of lung cancer. Glycoprotein acetyls demonstrated a positive association with lung cancer risk [HR = 1.13 (95%CI: 1.04, 1.22)]. Negative associations with lung cancer were found for albumin [0.78 (95%CI: 0.72, 0.83)], acetate [0.91 (95%CI: 0.85, 0.97)], valine [0.90 (95%CI: 0.83, 0.98)], alanine [0.88 (95%CI: 0.82, 0.95)], glucose [0.91 (95%CI: 0.85, 0.99)], citrate [0.91 (95%CI: 0.85, 0.99)], omega-3 fatty acids [0.83 (95%CI: 0.77, 0.90)], linoleic acid [0.83 (95%CI: 0.77, 0.89)], etc. Nine PCs represented over 90% of the total variances, and among those with statistically significant estimates, PC1 [0.85 (95%CI: 0.80, 0.92)], PC2 [0.88 (95%CI: 0.82, 0.95)], and PC9 [0.87 (95%CI: 0.80, 0.93)] were negatively associated with lung cancer risk, whereas PC7 [1.08 (95%CI: 1.00, 1.16)] and PC8 [1.16 (95%CI: 1.08, 1.26)] showed positive associations with lung cancer risk. The pathway analysis showed that the "linoleic acid metabolism" was statistically significant after the FDR adjustment (p value 0.0496). CONCLUSIONS Glycoprotein acetyls had a positive association with lung cancer risk while other metabolites and lipoprotein subfractions showed negative associations. Certain metabolites and lipoprotein subfractions might be independent risk factors for lung cancer. Our findings shed new light on the etiology of lung cancer and might aid the selection of high-risk individuals for lung cancer screening.
Collapse
Affiliation(s)
- Lan Wu
- Department of Cancer Prevention, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Jun Yang
- School of Public Health, Guangzhou Medical University, Guangzhou, China
| | - Yu Chen
- Department of Cancer Prevention, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Jiahao Lin
- Department of Cancer Prevention, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Wenkai Huang
- National Central Cancer Registry Office, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mengmeng Li
- Department of Cancer Prevention, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
22
|
Odehnalová N, Šandriková V, Hromadka R, Skaličková M, Dytrych P, Hoskovec D, Kejík Z, Hajduch J, Vellieux F, Vašáková MK, Martásek P, Jakubek M. The potential of exosomes in regenerative medicine and in the diagnosis and therapies of neurodegenerative diseases and cancer. Front Med (Lausanne) 2025; 12:1539714. [PMID: 40182844 PMCID: PMC11966052 DOI: 10.3389/fmed.2025.1539714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 02/06/2025] [Indexed: 04/05/2025] Open
Abstract
Exosomes, nanosized extracellular vesicles released by various cell types, are intensively studied for the diagnosis and treatment of cancer and neurodegenerative diseases, and they also display high usability in regenerative medicine. Emphasizing their diagnostic potential, exosomes serve as carriers of disease-specific biomarkers, enabling non-invasive early detection and personalized medicine. The cargo loading of exosomes with therapeutic agents presents an innovative strategy for targeted drug delivery, minimizing off-target effects and optimizing therapeutic interventions. In regenerative medicine, exosomes play a crucial role in intercellular communication, facilitating tissue regeneration through the transmission of bioactive molecules. While acknowledging existing challenges in standardization and scalability, ongoing research efforts aim to refine methodologies and address regulatory considerations. In summary, this review underscores the transformative potential of exosomes in reshaping the landscape of medical interventions, with a particular emphasis on cancer, neurodegenerative diseases, and regenerative medicine.
Collapse
Affiliation(s)
- Nikola Odehnalová
- NEXARS Research and Development Center C2P s.r.o, Chlumec nad Cidlinou, Czechia
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
| | - Viera Šandriková
- NEXARS Research and Development Center C2P s.r.o, Chlumec nad Cidlinou, Czechia
| | - Róbert Hromadka
- NEXARS Research and Development Center C2P s.r.o, Chlumec nad Cidlinou, Czechia
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
| | - Markéta Skaličková
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Petr Dytrych
- Department of Surgery-Department of Abdominal, Thoracic Surgery and Traumatology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - David Hoskovec
- Department of Surgery-Department of Abdominal, Thoracic Surgery and Traumatology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Zdeněk Kejík
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
- Department of Analytical Chemistry, University of Chemistry and Technology, Prague, Czechia
| | - Jan Hajduch
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
- The Department of Chemistry of Natural Compounds, University of Chemistry and Technology, Prague, Czechia
| | - Frédéric Vellieux
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Martina Koziar Vašáková
- Department of Respiratory Medicine, First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czechia
| | - Pavel Martásek
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Milan Jakubek
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
- Department of Analytical Chemistry, University of Chemistry and Technology, Prague, Czechia
| |
Collapse
|
23
|
Kersch CN, Grossberg AJ. Perioperative Radiation for Patients with Resectable Pancreatic Cancer: an Updated Review After the Initial RTOG 0848 Results. J Gastrointest Cancer 2025; 56:70. [PMID: 39987276 DOI: 10.1007/s12029-025-01185-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2025] [Indexed: 02/24/2025]
Abstract
PURPOSE Pancreatic cancer remains one of the most lethal malignancies, with limited long-term survival despite advances in treatment strategies. While surgical resection offers the best chance for cure in localized disease, high rates of recurrence underscore the need for effective adjuvant therapies. Over four decades, the role of adjuvant chemoradiation (CRT) has been the subject of significant debate, with numerous trials yielding mixed outcomes regarding its impact on survival. Improvements in chemotherapy regimens and radiotherapy techniques have prompted renewed efforts to define the value of CRT, particularly in comparison to chemotherapy alone. The recent initial results of RTOG 0848 mark a critical milestone in this ongoing discussion, providing contemporary evidence that challenges established assumptions and refines patient selection criteria. By identifying specific subgroups-such as lymph node-negative patients-which may benefit from CRT, the trial offers clarity while highlighting the limitations of CRT in other populations. METHODS Herein, we review prior prospective and retrospective trials that investigated the role of perioperative CRT, in particular radiation therapy, for resectable pancreatic cancer. RESULTS This review examines the trajectory of research on CRT in pancreatic cancer, assesses the implications of RTOG 0848 for current clinical practice, and underscores the importance of further studies to optimize the integration of multimodal therapy in the management of this aggressive disease. CONCLUSION The combination of results from RTOG 0848 in conjunction with the results of prior prospective and retrospective trials lend support for the use of adjuvant RT for patients with both lymph node-negative and lymph node-positive disease. However, several open questions remain about the role of this therapy in select patient cohorts, and whether neoadjuvant versus advent radiation is optimal.
Collapse
Affiliation(s)
- Cymon N Kersch
- Department of Radiation Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Aaron J Grossberg
- Department of Radiation Medicine, Oregon Health & Science University, Portland, OR, USA.
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR, USA.
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR, USA.
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
24
|
Elliott MJ, Echelard P, Pipinikas C, Main S, Fuentes Antrás J, Dou A, Veitch Z, Amir E, Nadler MB, Meti N, Atenafu E, Shah E, Yu C, Campbell N, Ventura R, Siu LL, Bedard PL, Berman HK, Cescon DW. Longitudinal evaluation of circulating tumor DNA in patients undergoing neoadjuvant therapy for early breast cancer using a tumor-informed assay. Nat Commun 2025; 16:1837. [PMID: 39984446 PMCID: PMC11845481 DOI: 10.1038/s41467-025-56658-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 01/21/2025] [Indexed: 02/23/2025] Open
Abstract
Circulating tumor DNA (ctDNA) is an emerging biomarker for the treatment of early breast cancer (EBC). We sought to evaluate a highly sensitive tumor-informed ctDNA assay in a real-world cohort of patients receiving neoadjuvant therapy (NAT) to assess clinical validity and explore prognostic outcomes. ctDNA is detected in 77.2% (88/114) of participants at baseline, with 18/88 (20.5%) having a baseline estimated variant allele frequency (eVAF) of <0.01%. Persistent detection of ctDNA, measured midway through NAT (mid-NAT), is associated with disease recurrence in all participants, reaching statistical significance in those with HER2-negative disease. Stratified analyses demonstrate that ctDNA detected mid-NAT enhances the prognostic accuracy of the residual cancer burden (RCB) score for disease recurrence. Postoperative or follow-up detection of ctDNA demonstrates a 100% positive predictive value for disease recurrence, with a median lead time of 374 days (range: 13-1010 days). These data suggest that assays with high analytical sensitivity may improve baseline ctDNA detection in patients with EBC. The ability to replicate the prognostic association of ctDNA dynamics in a real-world cohort supports further investigation. Prospective trials incorporating ctDNA testing are warranted to assess and develop the clinical utility of ctDNA-guided treatment strategies.
Collapse
Affiliation(s)
- Mitchell J Elliott
- Division of Medical Oncology & Hematology, Department of Medicine, Princess Margaret Cancer Centre and University of Toronto, Toronto, ON, Canada.
| | - Philippe Echelard
- Department of Pathology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | | | - Sasha Main
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Jesús Fuentes Antrás
- NEXT Oncology Early Drug Development Unit, Hospital Universitario Quirónsalud, Madrid, Spain
| | - Aaron Dou
- Division of Medical Oncology & Hematology, Department of Medicine, Princess Margaret Cancer Centre and University of Toronto, Toronto, ON, Canada
| | - Zachary Veitch
- Royal Victoria Regional Health Centre, Barrie, ON, Canada
| | - Eitan Amir
- Division of Medical Oncology & Hematology, Department of Medicine, Princess Margaret Cancer Centre and University of Toronto, Toronto, ON, Canada
| | - Michelle B Nadler
- Division of Medical Oncology & Hematology, Department of Medicine, Princess Margaret Cancer Centre and University of Toronto, Toronto, ON, Canada
| | - Nicholas Meti
- Gerald Bronfman Department of Oncology, St. Mary's Hospital Center, McGill University, Montréal, QC, Canada
| | - Eshetu Atenafu
- Biostatistics Department, University Health Network, Toronto, ON, Canada
| | - Elizabeth Shah
- Division of Medical Oncology & Hematology, Department of Medicine, Princess Margaret Cancer Centre and University of Toronto, Toronto, ON, Canada
| | - Celeste Yu
- Division of Medical Oncology & Hematology, Department of Medicine, Princess Margaret Cancer Centre and University of Toronto, Toronto, ON, Canada
| | | | | | - Lillian L Siu
- Division of Medical Oncology & Hematology, Department of Medicine, Princess Margaret Cancer Centre and University of Toronto, Toronto, ON, Canada
| | - Philippe L Bedard
- Division of Medical Oncology & Hematology, Department of Medicine, Princess Margaret Cancer Centre and University of Toronto, Toronto, ON, Canada
| | - Hal K Berman
- Department of Pathology and Laboratory Medicine, University Health Network, Toronto, ON, Canada
| | - David W Cescon
- Division of Medical Oncology & Hematology, Department of Medicine, Princess Margaret Cancer Centre and University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
25
|
Allepot K, Morel-Journel N, Boucher F. Abdominal wall complications: An unknown aspect of morbidity in phalloplasty. A comprehensive analysis and clinical implications. ANN CHIR PLAST ESTH 2025:S0294-1260(25)00007-X. [PMID: 39979157 DOI: 10.1016/j.anplas.2025.01.007] [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: 08/19/2024] [Revised: 01/09/2025] [Accepted: 01/14/2025] [Indexed: 02/22/2025]
Abstract
BACKGROUND Phalloplasty is a complex reconstructive procedure with complications broadly categorized as urinary, vascular, or donor-site related. This study investigates abdominal wall complications, such as bulging and lateral hernias, associated with the use of the inferior epigastric artery as the recipient vessel in microsurgical phalloplasty-a rare and underreported complication. METHODS A retrospective review was conducted on 37 patients who underwent microsurgical phalloplasty at a university hospital from January 2016 to February 2020. The most commonly employed technique was forearm flap phalloplasty, followed by the MSLD flap technique. The inferior epigastric artery was accessed via a 7cm oblique incision. Data collected included demographic details, BMI, smoking status, surgical technique, recipient vessel used, and postoperative complications. Follow-up evaluations were performed at 15 days, 3 months, 6 months, and 1 year postoperatively. RESULTS Of the 37 patients, the mean age was 33 years, with 84% undergoing phalloplasty for gender affirmation. Abdominal wall complications occurred in 11% of patients (n=4). Three patients developed parietal complications (hernia or bulging), all requiring surgical revision. All three were smokers, one had a BMI>28, and none had prior abdominal surgeries. Contributing factors included musculoaponeurotic disruption, relative denervation from vessel exposure, and smoking-related wound healing impairment. CONCLUSIONS The use of the inferior epigastric artery in microsurgical phalloplasty may increase the risk of abdominal wall complications, particularly in smokers and patients with elevated BMI. Optimizing preoperative risk factors, including smoking cessation and careful vessel selection, is essential for reducing these complications. To reduce the occurrence of these complications, the choice of the recipient vessel should be considered alongside the surgical technique during operative planning. Further studies should explore technical refinements to minimize abdominal wall morbidity.
Collapse
Affiliation(s)
- K Allepot
- Department of Plastic Surgery, Ambroise-Paré, 9, avenue Charles-de-Gaulle, 92100 Boulogne-Billancourt, France.
| | - N Morel-Journel
- Department of Urology, Hospices Civils de Lyon, 165, chemin du Grand Revoyet, 69495 Pierre-Bénite, France
| | - F Boucher
- Department of Plastic Surgery, Hospices Civils de Lyon, 103, grande rue de la Croix Rousse, 69004 Lyon, France
| |
Collapse
|
26
|
Kuang L, Wu L, Li Y. Extracellular vesicles in tumor immunity: mechanisms and novel insights. Mol Cancer 2025; 24:45. [PMID: 39953480 PMCID: PMC11829561 DOI: 10.1186/s12943-025-02233-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: 09/30/2024] [Accepted: 01/14/2025] [Indexed: 02/17/2025] Open
Abstract
Extracellular vesicles (EVs), nanoscale vesicles secreted by cells, have attracted considerable attention in recent years due to their role in tumor immunomodulation. These vesicles facilitate intercellular communication by transporting proteins, nucleic acids, and other biologically active substances, and they exhibit a dual role in tumor development and immune evasion mechanisms. Specifically, EVs can assist tumor cells in evading immune surveillance and attack by impairing immune cell function or modulating immunosuppressive pathways, thereby promoting tumor progression and metastasis. Conversely, they can also transport and release immunomodulatory factors that stimulate the activation and regulation of the immune system, enhancing the body's capacity to combat malignant diseases. This dual functionality of EVs presents promising avenues and targets for tumor immunotherapy. By examining the biological characteristics of EVs and their influence on tumor immunity, novel therapeutic strategies can be developed to improve the efficacy and relevance of cancer treatment. This review delineates the complex role of EVs in tumor immunomodulation and explores their potential implications for cancer therapeutic approaches, aiming to establish a theoretical foundation and provide practical insights for the advancement of future EVs-based cancer immunotherapy strategies.
Collapse
Affiliation(s)
- Liwen Kuang
- School of Medicine, Chongqing University, Chongqing, China
| | - Lei Wu
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Yongsheng Li
- School of Medicine, Chongqing University, Chongqing, China.
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, China.
| |
Collapse
|
27
|
Mohamed AH, Abaza T, Youssef YA, Rady M, Fahmy SA, Kamel R, Hamdi N, Efthimiado E, Braoudaki M, Youness RA. Extracellular vesicles: from intracellular trafficking molecules to fully fortified delivery vehicles for cancer therapeutics. NANOSCALE ADVANCES 2025; 7:934-962. [PMID: 39823046 PMCID: PMC11733735 DOI: 10.1039/d4na00393d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 12/22/2024] [Indexed: 01/19/2025]
Abstract
Extracellular vesicles (EVs) are emerging as viable tools in cancer treatment due to their ability to carry a wide range of theranostic activities. This review summarizes different forms of EVs such as exosomes, microvesicles, apoptotic bodies, and oncosomes. It also sheds the light onto isolation methodologies, characterization techniques and therapeutic applications of all discussed EVs. Evidence indicates that EVs are particularly effective in delivering chemotherapeutic medications, and immunomodulatory agents. However, the advancement of EV-based therapies into clinical practice is hindered by challenges including EVs heterogeneity, cargo loading efficiency, and in vivo stability. Overall, EVs have the potential to change cancer therapeutic paradigms. Continued research and development activities are critical for improving EV-based medications and increasing their therapeutic impact.
Collapse
Affiliation(s)
- Adham H Mohamed
- Department of Chemistry, Faculty of Science, Cairo University 12613 Giza Egypt
| | - Tasneem Abaza
- Biotechnology and Biomolecular Chemistry Program, Faculty of Science, Cairo University 12613 Giza Egypt
- Université Paris-Saclay, Université d'Evry Val D'Essonne 91000 Évry-Courcouronnes Île-de-France France
| | - Yomna A Youssef
- Department of Physiology, Faculty of Physical Therapy, German International University (GIU) 11835 Cairo Egypt
- Molecular Biology and Biochemistry Department, Faculty of Biotechnology, German International University (GIU) 11835 Cairo Egypt
| | - Mona Rady
- Microbiology, Immunology and Biotechnology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC) 11835 Cairo Egypt
- Faculty of Biotechnology, German International University New Administrative Capital 11835 Cairo Egypt
| | - Sherif Ashraf Fahmy
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg Robert-Koch-Str. 4 35037 Marburg Germany
| | - Rabab Kamel
- Pharmaceutical Technology Department, National Research Centre 12622 Cairo Egypt
| | - Nabila Hamdi
- Pharmacology and Toxicology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC) 11835 Cairo Egypt
| | - Eleni Efthimiado
- Inorganic Chemistry Laboratory, Chemistry Department, National and Kapodistrian University of Athens Athens Greece
| | - Maria Braoudaki
- Department of Clinical, Pharmaceutical, and Biological Science, School of Life and Medical Sciences, University of Hertfordshire Hatfield AL10 9AB UK
| | - Rana A Youness
- Molecular Biology and Biochemistry Department, Faculty of Biotechnology, German International University (GIU) 11835 Cairo Egypt
| |
Collapse
|
28
|
Zhao M, Lin Y, Zeng Y, Lv Z, Liang J, Tang P, Zhen X, Han L. Biomimetic membrane-coated nanoparticles specially permeate the inflammatory blood-brain barrier to deliver plasmin therapy for brain metastases. J Control Release 2025; 378:763-775. [PMID: 39732369 DOI: 10.1016/j.jconrel.2024.12.066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 12/13/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
Many brain-targeting drug delivery strategies have been reported to permeate the blood-brain barrier (BBB) via hijacking receptor-mediated transport. However, these receptor-based strategies could mediate whole-brain BBB crossing due to the wide intracranial expression of target receptors and lead to unwanted accumulation and side effects on healthy brain tissues. Inspired by brain metastatic processes and the selectivity of brain metastatic cancer cells for the inflammatory BBB, a biomimetic nanoparticle was developed by coating drug-loaded core with the inflammatory BBB-seeking erythrocyte-brain metastatic hybrid membrane, which can resist homotypic aggregation and specially bind and permeate the inflammatory BBB for specific drug delivery. Dexamethasone and embelin were used as model drugs to be loaded in the biomimetic nanoparticles to restore plasmin-mediated attacks against brain metastases. The drug-loaded nanoparticles were proved to inhibit tumor serpin secretion to restore local plasmin production, which could inactivate tumor cell surface L1CAM to inhibit vessel-spreading-dependent tumor growth and produce lethal soluble factor-related apoptosis ligands (sFasL) to induce tumor cell apoptosis, leading to the suppression of the intracranial metastatic nodule development and prolonged survival of mice with brain metastases. The inflammatory BBB-seeking biomimetic approach represents an effective regimen for potent therapy against brain metastases.
Collapse
Affiliation(s)
- Mei Zhao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yuanyuan Lin
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yuteng Zeng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Ziyan Lv
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Jiayu Liang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Puxian Tang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Xuechu Zhen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Liang Han
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou 215123, China.
| |
Collapse
|
29
|
Levine EA, Cos H, Votanopoulos KI, Shen P, Russell G, Mansfield P, Fournier K, Bartlett D, Stewart JH. Ten-Year Outcome of a Randomized Trial: Cytoreduction and HIPEC with Mitomycin C Versus Oxaliplatin for Appendiceal Neoplasm with Peritoneal Dissemination. Ann Surg Oncol 2025; 32:679-686. [PMID: 39531120 PMCID: PMC11698756 DOI: 10.1245/s10434-024-16441-z] [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: 06/21/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Appendiceal cancer is a rare disease that has proven difficult to study in prospectively. Our initial report of this trial showed minor hematologic toxicity with both mitomycin C and oxaliplatin and similar 3-year survival. We now report an update of the first prospective randomized trial for appendiceal cancer with 10-year follow up. PATIENTS AND METHODS Patients with mucinous appendiceal neoplasms and evidence of peritoneal dissemination were enrolled in the Multicenter Randomized Trial to evaluating HIPEC for 120 min with oxaliplatin (200 mg/M2) or mitomycin C (40 mg). Overall survival and disease-free survival were calculated at 10 years and compared between the groups. RESULTS A total of 121 patients were included in the study. The patients were 57% female, with a mean age of 55.3 years (range 22-82 years). The disease was low grade in 71% and high grade in 29%. The average peritoneal cancer index (PCI) score was 18 (SD 10) in the mitomycin C group and 17.9 (SD 9.4) in the oxaliplatin group (p = 0.94). The 10-year survival rate was 56.2% (SE 7.2) with mitomycin C and 47.5% (SE 8.4) with oxaliplatin, p = 0.83. The 10-year progression-free survival rate in the mitomycin C group was 45.2% (SE 8.4) compared with 50.4% (SE 6.7) in the oxaliplatin group, p = 0.95. Median survival was 9.1 years after HIPEC with oxaliplatin, and median not reached for the mitomycin C group (> 5.6 years). CONCLUSIONS Oxaliplatin and mitomycin C have similar long-term efficacy for hyperthermic intraperitoneal chemotherapy (HIPEC) in patients with appendiceal neoplasms and peritoneal dissemination. Long-term survival is experienced by most patients after cytoreduction surgery (CRS) and HIPEC for appendiceal neoplasms.
Collapse
Affiliation(s)
- Edward A Levine
- Section of Surgical Oncology, Department of General Surgery, Wake Forest Baptist Health, Winston-Salem, NC, USA.
| | - Heidy Cos
- Section of Surgical Oncology, Department of General Surgery, Wake Forest Baptist Health, Winston-Salem, NC, USA
| | - Konstantinos I Votanopoulos
- Section of Surgical Oncology, Department of General Surgery, Wake Forest Baptist Health, Winston-Salem, NC, USA
| | - Perry Shen
- Section of Surgical Oncology, Department of General Surgery, Wake Forest Baptist Health, Winston-Salem, NC, USA
| | - Greg Russell
- Department of Biostatistics and Data Science, Wake Forest Baptist Health, Winston-Salem, NC, USA
| | | | | | | | - John H Stewart
- Section of Surgical Oncology, Department of General Surgery, Wake Forest Baptist Health, Winston-Salem, NC, USA
| |
Collapse
|
30
|
Nie L, Ma J, Yu Y, Tao Y, Song Z, Li J. Exosomes as carriers to stimulate an anti-cancer immune response in immunotherapy and as predictive markers. Biochem Pharmacol 2025; 232:116699. [PMID: 39647605 DOI: 10.1016/j.bcp.2024.116699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/26/2024] [Accepted: 12/03/2024] [Indexed: 12/10/2024]
Abstract
During this era of rapid advancements in cancer immunotherapy, the application of cell-released small vesicles that activate the immune system is of considerable interest. Exosomes are cell-derived nanovesicles that show great promise for the immunological treatment of cancer because of their immunogenicity and molecular transfer capacity. Recent technological advancements have enabled the identification of functional functions that exosome cargoes perform in controlling immune responses. Exosomes are originated specifically from immune cells and tumor cells and they show unique composition patterns directly related to the immunotherapy against cancer. Exosomes can also deliver their cargo to particular cells, which can affect the phenotypic and immune-regulatory functions of those cells. Exosomes can influence the course of cancer and have therapeutic benefits by taking part in several cellular processes; as a result, they have the dual properties of activating and restraining cancer. Exosomes have tremendous potential for cancer immunotherapy; they may develop into the most powerful cancer vaccines and carriers of targeted antigens and drugs. Comprehending the potential applications of exosomes in immune therapy is significant for regulating cancer progression. This review offers an analysis of the function of exosomes in immunotherapy, specifically as carriers that function as diagnostic indicators for immunological activation and trigger an anti-cancer immune response. Moreover, it summarizes the fundamental mechanism and possible therapeutic applications of exosome-based immunotherapy for human cancer.
Collapse
Affiliation(s)
- Lili Nie
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun, China
| | - Jingru Ma
- Department of Clinical Laboratory, the Second Hospital of Jilin University, Changchun, China
| | - Yang Yu
- Department of Emergency and Critical Care, the Second Hospital of Jilin University, Changchun, China
| | - Ying Tao
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zhidu Song
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun, China
| | - Jian Li
- Department of Emergency and Critical Care, the Second Hospital of Jilin University, Changchun, China.
| |
Collapse
|
31
|
Marchio V, Augimeri G, Morelli C, Vivacqua A, Giordano C, Catalano S, Sisci D, Barone I, Bonofiglio D. Omega-3 fatty acids: molecular weapons against chemoresistance in breast cancer. Cell Mol Biol Lett 2025; 30:11. [PMID: 39863855 PMCID: PMC11762563 DOI: 10.1186/s11658-025-00694-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Breast cancer is the most commonly diagnosed type of cancer and the leading cause of cancer-related death in women worldwide. Highly targeted therapies have been developed for different subtypes of breast cancer, including hormone receptor (HR)-positive and human epidermal growth factor receptor 2 (HER2)-positive breast cancer. However, triple-negative breast cancer (TNBC) and metastatic breast cancer disease are primarily treated with chemotherapy, which improves disease-free and overall survival, but does not offer a curative solution for these aggressive forms of breast cancer. Moreover, the development of chemoresistance is a major cause of therapeutic failure in this neoplasia, leading to disease relapse and patient death. In addition, chemotherapy's adverse side effects may substantially worsen health-related quality of life. Therefore, to improve the outcome of patients with breast cancer who are undergoing chemotherapy, several therapeutic options are under investigation, including the combination of chemotherapeutic drugs with natural compounds. Omega-3 (ω-3) polyunsaturated fatty acids (PUFAs), including docosahexaenoic and eicosapentaenoic acids, have drawn attention for their antitumoral properties and their preventive activities against chemotherapy-induced toxicities in breast cancer. A literature review was conducted on PubMed using keywords related to breast cancer, omega-3, chemoresistance, and chemotherapy. This review aims to provide an overview of the molecular mechanisms driving breast cancer chemoresistance, focusing on the role of ω-3 PUFAs in these recognized cellular paths and presenting current findings on the effects of ω-3 PUFAs combined with chemotherapeutic drugs in breast cancer management.
Collapse
Affiliation(s)
- Vittoria Marchio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende, 87036, Cosenza, Italy
| | - Giuseppina Augimeri
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende, 87036, Cosenza, Italy
| | - Catia Morelli
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende, 87036, Cosenza, Italy
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
| | - Adele Vivacqua
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende, 87036, Cosenza, Italy
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
| | - Cinzia Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende, 87036, Cosenza, Italy
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
| | - Stefania Catalano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende, 87036, Cosenza, Italy
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
| | - Diego Sisci
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende, 87036, Cosenza, Italy
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
| | - Ines Barone
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende, 87036, Cosenza, Italy.
| | - Daniela Bonofiglio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende, 87036, Cosenza, Italy
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
| |
Collapse
|
32
|
Zhao M, Li L, Wang B, Gao S, Wang J, Liu J, Song Y, Liu H. Comparing survival outcomes between neoadjuvant and adjuvant chemotherapy within hormone receptor-positive, human epidermal growth factor receptor 2-negative early breast cancer among young women (≤35): a retrospective cohort study based on SEER database and TJMUCH registry. Am J Cancer Res 2025; 15:390-405. [PMID: 39949935 PMCID: PMC11815373 DOI: 10.62347/ezgv9302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 01/17/2025] [Indexed: 02/16/2025] Open
Abstract
Breast cancer is a leading cause of cancer morbidity and mortality among young women, who often experience more aggressive disease, which may impact their treatment responses and long-term prognoses. Understanding the effectiveness of neoadjuvant chemotherapy (NAC) versus adjuvant chemotherapy (AC) in this specific population is critical for optimizing treatment strategies and improving prognoses. This research was conducted to compare the prognoses of young women (≤35 years old) with early-stage hormone receptor (HR)-positive, human epidermal growth factor receptor 2 (HER2)-negative breast cancer, who were treated with NAC versus those treated with AC. This study retrospectively analyzed data from young women with HR+/HER2- breast cancer, with complete follow-up information, sourced from the Surveillance, Epidemiology, and End Results (SEER) database (2010-2018) and the Tianjin Medical University Cancer Institute and Hospital (TJMUCH) (2014-2018). Patients from both cohorts were allocated to NAC and AC groups based on their treatment regimens. Categorical variables were compared using chi-square, whereas the Kaplan-Meier method was utilized to generate survival curves; additionally, the log-rank test was employed for survival analysis. Propensity score matching (PSM) was employed to control baseline differences. Analysis of the SEER and TJMUCH cohorts revealed that patients treated with NAC had significantly worse overall survival (OS) compared to those treated with AC, as indicated by Kaplan-Meier curves both before and after PSM. The disease-free survival analysis of the TJMUCH cohort yielded similar results, indicating that patients treated with AC experienced longer periods without disease recurrence compared to their counterparts receiving NAC. Statistically significant differences were observed across both survival metrics, reinforcing the robustness of our findings. Overall, among young women (≤35 years old) with early-stage HR+/HER2- breast cancer, patients treated with AC exhibited a more favorable prognosis and improved survival outcomes compared to those treated with NAC. These findings could potentially influence clinical decision-making and treatment guidelines, advocating for a more tailored approach in managing young women with HR+/HER2- breast cancer.
Collapse
Affiliation(s)
- Mengjun Zhao
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerTianjin 300060, China
- Tianjin’s Clinical Research Center for CancerTianjin 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of EducationTianjin 300060, China
| | - Linwei Li
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerTianjin 300060, China
- Tianjin’s Clinical Research Center for CancerTianjin 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of EducationTianjin 300060, China
| | - Bin Wang
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerTianjin 300060, China
- Tianjin’s Clinical Research Center for CancerTianjin 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of EducationTianjin 300060, China
| | - Shuang Gao
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerTianjin 300060, China
- Tianjin’s Clinical Research Center for CancerTianjin 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of EducationTianjin 300060, China
| | - Jinhui Wang
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerTianjin 300060, China
- Tianjin’s Clinical Research Center for CancerTianjin 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of EducationTianjin 300060, China
| | - Jianing Liu
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerTianjin 300060, China
- Tianjin’s Clinical Research Center for CancerTianjin 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of EducationTianjin 300060, China
| | - Yixuan Song
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerTianjin 300060, China
- Tianjin’s Clinical Research Center for CancerTianjin 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of EducationTianjin 300060, China
| | - Hong Liu
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerTianjin 300060, China
- Tianjin’s Clinical Research Center for CancerTianjin 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of EducationTianjin 300060, China
| |
Collapse
|
33
|
Cao L, Duan Q, Zhu Z, Xu X, Liu J, Li B. Liquid biopsy technologies: innovations and future directions in breast cancer biomarker detection. Biomed Microdevices 2025; 27:4. [PMID: 39849252 DOI: 10.1007/s10544-025-00734-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2025] [Indexed: 01/25/2025]
Abstract
Globally, breast cancer is the most frequent type of cancer, and its early diagnosis and screening can significantly improve the probability of survival and quality of life of those affected. Liquid biopsy-based targets such as circulating tumor cells, circulating tumor DNA, and exosomes have been instrumental in the early discovery of cancer, and have been found to be effective in stage therapy, recurrence monitoring, and drug selection. Biosensors based on these target related biomarkers convert the tested substances into quantifiable signals such as electrical and optical signals through signal transduction, which has the advantages of high sensitivity, simple operation, and low invasiveness. This review provides an overview of the latest progress of liquid biopsy biomarkers in the diagnosis, prognosis and treatment of breast cancer, compares the application and advantages of different biosensors based on these biomarkers in the diagnosis of breast cancer, and analyzes the limitations and solutions of biosensor based methods.
Collapse
Affiliation(s)
- Linhong Cao
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou, 646000, Sichuan, People's Republic of China
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, China
- Molecular Diagnosis of Clinical Diseases Key Laboratory, Luzhou, Sichuan, China
| | - Qingli Duan
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou, 646000, Sichuan, People's Republic of China
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, China
- Molecular Diagnosis of Clinical Diseases Key Laboratory, Luzhou, Sichuan, China
| | - Zixin Zhu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou, 646000, Sichuan, People's Republic of China
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, China
- Molecular Diagnosis of Clinical Diseases Key Laboratory, Luzhou, Sichuan, China
| | - Xuejing Xu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou, 646000, Sichuan, People's Republic of China
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, China
- Molecular Diagnosis of Clinical Diseases Key Laboratory, Luzhou, Sichuan, China
| | - Jinbo Liu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou, 646000, Sichuan, People's Republic of China.
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, China.
- Molecular Diagnosis of Clinical Diseases Key Laboratory, Luzhou, Sichuan, China.
| | - Baolin Li
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou, 646000, Sichuan, People's Republic of China.
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, China.
- Molecular Diagnosis of Clinical Diseases Key Laboratory, Luzhou, Sichuan, China.
| |
Collapse
|
34
|
Ahn JY, Lee DK, Kim MG, Kim WJ, Park SH. Temperature-Responsive Hybrid Composite with Zero Temperature Coefficient of Resistance for Wearable Thermotherapy Pads. MICROMACHINES 2025; 16:108. [PMID: 39858763 PMCID: PMC11767656 DOI: 10.3390/mi16010108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/13/2025] [Accepted: 01/17/2025] [Indexed: 01/27/2025]
Abstract
Carbon-based polymer composites are widely used in wearable devices due to their exceptional electrical conductivity and flexibility. However, their temperature-dependent resistance variations pose significant challenges to device safety and performance. A negative temperature coefficient (NTC) can lead to overcurrent risks, while a positive temperature coefficient (PTC) compromises accuracy. In this study, we present a novel hybrid composite combining carbon nanotubes (CNTs) with NTC properties and carbon black (CB) with PTC properties to achieve a near-zero temperature coefficient of resistance (TCR) at an optimal ratio. This innovation enhances the safety and reliability of carbon-based polymer composites for wearable heating applications. Furthermore, a thermochromic pigment layer is integrated into the hybrid composite, enabling visual temperature indication across three distinct zones. This bilayer structure not only addresses the TCR challenge but also provides real-time, user-friendly temperature monitoring. The resulting composite demonstrates consistent performance and high precision under diverse heating conditions, making it ideal for wearable thermotherapy pads. This study highlights a significant advancement in developing multifunctional, temperature-responsive materials, offering a promising solution for safer and more controllable wearable devices.
Collapse
Affiliation(s)
| | | | | | | | - Sung-Hoon Park
- Department of Mechanical Engineering, Soongsil University, 369 Sangdo-ro, Dongjak-Gu, Seoul 06978, Republic of Korea; (J.-Y.A.); (D.-K.L.); (M.-G.K.); (W.-J.K.)
| |
Collapse
|
35
|
Bhandari K, Kong JS, Tina Ho WT, Bourne PC, Mooers BH, Ding WQ. Arginine demethylation of Serine/Arginine-rich splicing factor 1 enhances miRNA enrichment in small extracellular vesicles derived from pancreatic ductal adenocarcinoma cells. FASEB J 2025; 39:e70287. [PMID: 39760222 PMCID: PMC11827091 DOI: 10.1096/fj.202401811rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 01/07/2025]
Abstract
Small extracellular vesicles (sEVs) are enriched in certain miRNAs, impacting the progression of pancreatic ductal adenocarcinoma (PDAC). The mechanisms involved in the selective sEV miRNA enrichment remain to be elucidated. We recently reported that Serine/Arginine-rich splicing factor 1 (SRSF1) regulates selective sEV miRNA enrichment in PDAC cells. SRSF1 is an onco-protein that is overexpressed in PDAC, and its function is dictated by posttranslational modifications such as phosphorylation and arginine methylation. The objective of this study was to examine the role of phosphorylation and arginine methylation in SRSF1-mediated sEV miRNA enrichment in PDAC cells. Treatment of PDAC cells with the protein arginine methyltransferase inhibitors AMI-5 and EPZ015666, but not with the phosphorylation inhibitor SRPIN340, selectively enhanced the level of sEV miR-1246, a miRNA known to be highly enriched in PDAC sEVs. Consistently, overexpression of the mutant SRSF1 with the three arginine residues R93, R97, and R109 being replaced with lysinaugmented sEV miR-1246 levels in both wild-type and SRSF1-knockdown PANC-1 cells. Interestingly, the binding of SRSF1 to miR-1246 was significantly reduced in PDAC cells overexpressing the mutant SRSF1, which was further confirmed using purified wild-type and the mutant SRSF1 proteins. We demonstrate that arginine demethylation of SRSF1 reduces SRSF1-miRNA binding in PDAC cells and enhances selective sEV miRNA enrichment, providing novel insight into SRSF1-mediated sEV miRNA enrichment in PDAC cells and opening up new avenues of investigation on the biology and function of extracellular vesicles in PDAC.
Collapse
Affiliation(s)
- Kritisha Bhandari
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City
| | - Jeng Shi Kong
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City
| | - Wang-Ting Tina Ho
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City
| | - Philip C. Bourne
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, USA
| | - Blaine H.M. Mooers
- Department of Physiology and Biochemistry, University of Oklahoma Health Sciences Center, Oklahoma City
| | - Wei-Qun Ding
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City
| |
Collapse
|
36
|
Xu X, Zhang Y, Liu J, Wei S, Li N, Yao X, Wang M, Su X, Jing G, Xu J, Liu Y, Lu Y, Cheng J, Xu Y. Concurrent Detection of Protein and miRNA at the Single Extracellular Vesicle Level Using a Digital Dual CRISPR-Cas Assay. ACS NANO 2025; 19:1271-1285. [PMID: 39688838 DOI: 10.1021/acsnano.4c13557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
The simultaneous detection of proteins and microRNA (miRNA) at the single extracellular vesicle (EV) level shows great promise for precise disease profiling, owing to the heterogeneity and scarcity of tumor-derived EVs. However, a highly reliable method for multiple-target analysis of single EVs remains to be developed. In this study, a digital dual CRISPR-Cas-powered Single EV Evaluation (ddSEE) system was proposed to enable the concurrent detection of surface protein and inner miRNA of EVs at the single-molecule level. By optimizing simultaneous reaction conditions of CRISPR-Cas12a and CRISPR-Cas13a, the surface protein of EVs was detected by Cas12a using antibody-DNA conjugates to transfer the signal of the protein to DNA, while the inner miRNA was analyzed by Cas13a through EV-liposome fusion. A microfluidic chip containing 188,000 microwells was used to convert the CRISPR-Cas system into a digital assay format to enable the absolute quantification of miRNA/protein-positive EVs without bias through fluorescence imaging, which can detect as few as 214 EVs/μL. Finally, a total of 31 blood samples, 21 from breast cancer patients and 10 from healthy donors, were collected and tested, achieving a diagnostic accuracy of 92% in distinguishing patients with breast cancer from healthy donors. With its absolute quantification, ease of use, and multiplexed detection capability, the ddSEE system demonstrates its great potential for both EV research and clinical applications.
Collapse
Affiliation(s)
- Xun Xu
- School of Biomedical Engineering, Tsinghua University, Beijing 100084, China
| | - Yuanyue Zhang
- School of Biomedical Engineering, Tsinghua University, Beijing 100084, China
| | - Jiajia Liu
- School of Biomedical Engineering, Tsinghua University, Beijing 100084, China
- CapitalBio Technology, Beijing 101111, China
| | - Shujin Wei
- School of Biomedical Engineering, Tsinghua University, Beijing 100084, China
| | - Nan Li
- School of Biomedical Engineering, Tsinghua University, Beijing 100084, China
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China
| | - Xintong Yao
- School of Biomedical Engineering, Tsinghua University, Beijing 100084, China
| | - Muxue Wang
- School of Biomedical Engineering, Tsinghua University, Beijing 100084, China
| | - Xiaohan Su
- School of Biomedical Engineering, Tsinghua University, Beijing 100084, China
| | - Gaoshan Jing
- Institute of Microelectronics of the Chinese Academy of Sciences, Beijing 100029, China
| | - Junquan Xu
- Iomics Biosciences, Beijing 101318, China
| | - Yan Liu
- Iomics Biosciences, Beijing 101318, China
| | - Ying Lu
- School of Biomedical Engineering, Tsinghua University, Beijing 100084, China
- National Engineering Research Center for Beijing Biochip Technology, Beijing 102200, China
| | - Jing Cheng
- School of Biomedical Engineering, Tsinghua University, Beijing 100084, China
- National Engineering Research Center for Beijing Biochip Technology, Beijing 102200, China
| | - Youchun Xu
- School of Biomedical Engineering, Tsinghua University, Beijing 100084, China
- National Engineering Research Center for Beijing Biochip Technology, Beijing 102200, China
| |
Collapse
|
37
|
Vasu S, Johnson V, M A, Reddy KA, Sukumar UK. Circulating Extracellular Vesicles as Promising Biomarkers for Precession Diagnostics: A Perspective on Lung Cancer. ACS Biomater Sci Eng 2025; 11:95-134. [PMID: 39636879 DOI: 10.1021/acsbiomaterials.4c01323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Extracellular vesicles (EVs) have emerged as promising biomarkers in liquid biopsy, owing to their ubiquitous presence in bodily fluids and their ability to carry disease-related cargo. Recognizing their significance in disease diagnosis and treatment, substantial efforts have been dedicated to developing efficient methods for EV isolation, detection, and analysis. EVs, heterogeneous membrane-encapsulated vesicles secreted by all cells, contain bioactive substances capable of modulating recipient cell biology upon internalization, including proteins, lipids, DNA, and various RNAs. Their prevalence across bodily fluids has positioned them as pivotal mediators in physiological and pathological processes, notably in cancer, where they hold potential as straightforward tumor biomarkers. This review offers a comprehensive examination of advanced nanotechnology-based techniques for detecting lung cancer through EV analysis. It begins by providing a brief overview of exosomes and their role in lung cancer progression. Furthermore, this review explores the evolving landscape of EV isolation and cargo analysis, highlighting the importance of characterizing specific biomolecular signatures within EVs for improved diagnostic accuracy in lung cancer patients. Innovative strategies for enhancing the sensitivity and specificity of EV isolation and detection, including the integration of microfluidic platforms and multiplexed biosensing technologies are summarized. The discussion then extends to key challenges associated with EV-based liquid biopsies, such as the standardization of isolation and detection protocols and the establishment of robust analytical platforms for clinical translation. This review highlights the transformative impact of EV-based liquid biopsy in lung cancer diagnosis, heralding a new era of personalized medicine and improved patient care.
Collapse
Affiliation(s)
- Sunil Vasu
- Department of Chemical Engineering, Indian Institute of Technology Tirupati, Tirupati, Andhra Pradesh, India-517 619
| | - Vinith Johnson
- Department of Chemical Engineering, Indian Institute of Technology Tirupati, Tirupati, Andhra Pradesh, India-517 619
| | - Archana M
- Department of Chemical Engineering, Indian Institute of Technology Tirupati, Tirupati, Andhra Pradesh, India-517 619
| | - K Anki Reddy
- Department of Chemical Engineering, Indian Institute of Technology Tirupati, Tirupati, Andhra Pradesh, India-517 619
| | - Uday Kumar Sukumar
- Department of Chemical Engineering, Indian Institute of Technology Tirupati, Tirupati, Andhra Pradesh, India-517 619
| |
Collapse
|
38
|
Kim OH, Jeon TJ, Kang H, Chang ES, Hong SA, Kim MK, Lee HJ. hsa-mir-483-3p modulates delayed breast cancer recurrence. Sci Rep 2025; 15:693. [PMID: 39753688 PMCID: PMC11698896 DOI: 10.1038/s41598-024-84437-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 12/23/2024] [Indexed: 01/06/2025] Open
Abstract
Patients with estrogen receptor-positive breast cancer undergoing continuous adjuvant hormone therapy often experience delayed recurrence with tamoxifen use, potentially causing adverse effects. However, the lack of biomarkers hampers patient selection for extended endocrine therapy. This study aimed to elucidate the molecular mechanisms underlying delayed recurrence and identify biomarkers. When miRNA expression was assessed in luminal breast cancer tissues with and without delayed recurrence using NanoString, a significant increase in the expression of miR483-3p was observed in samples from patients with delayed recurrence compared with those without. miR483-3p expression was elevated in tamoxifen resistant (TAMR) EFM19 cells than in non-resistant EFM19 cells. Notably, genes associated with cancer metastasis (AMOTL2, ANKRD1, CTGF, and VEGF) were upregulated in TAMR EFM19 cells, although cell motility and proliferation were reduced. Transfection of miR483-3p mimics into both non-resistant EFM19 and MCF7 cells resulted in increased expression of cancer metastasis-related genes, but decreased proliferation and migration. Given that miR483-3p can bind to the 3'UTR region of O-GlcNAc transferase (OGT) and potentially affect its protein expression, we examined OGT protein levels and found that transfection with miR483-3p mimics selectively reduced OGT expression. Overall, breast cancer cells subjected to long-term hormone therapy displayed elevated miR483-3p expression, reducing motility and dormancy induction via decreased OGT expression. These findings suggest that miR483-3p is a potential biomarker for long-term endocrine therapy.
Collapse
Affiliation(s)
- Ok-Hyeon Kim
- Department of Anatomy and Cell Biology, College of Medicine, Chung-Ang University, Seoul, 06974, South Korea
| | - Tae Jin Jeon
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, 06974, South Korea
| | - Hana Kang
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, 06974, South Korea
| | - Eun Seo Chang
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, 06974, South Korea
| | - Soon Auck Hong
- Department of Pathology, College of Medicine, Chung-Ang University, Seoul, 06974, South Korea
| | - Min Kyoon Kim
- Department of Surgery, College of Medicine, Chung-Ang University, Seoul, 06974, South Korea.
| | - Hyun Jung Lee
- Department of Anatomy and Cell Biology, College of Medicine, Chung-Ang University, Seoul, 06974, South Korea.
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, 06974, South Korea.
| |
Collapse
|
39
|
Hashimoto K, Ochiya T, Shimomura A. Liquid biopsy using non-coding RNAs and extracellular vesicles for breast cancer management. Breast Cancer 2025; 32:16-25. [PMID: 38512533 DOI: 10.1007/s12282-024-01562-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 02/24/2024] [Indexed: 03/23/2024]
Abstract
This article examines liquid biopsy using non-coding RNAs and extracellular vesicles in detail. Liquid biopsy is emerging as a prominent non-invasive diagnostic tool in the treatment of breast cancer. We will elucidate the roles of these molecules in early detection, monitoring treatment effectiveness, and prognostic assessment of breast cancer. Additionally, the clinical significance of these molecules will be discussed. We aim to delve into the distinct characteristics of these molecules and their possible roles in breast cancer management, with an anticipation of their contribution to future diagnostic and therapeutic advancements.
Collapse
Affiliation(s)
- Kazuki Hashimoto
- Department of Breast Surgical Oncology, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-Ku, Tokyo, 162-8655, Japan
| | - Takahiro Ochiya
- Department of Molecular and Cellular Medicine, Institute of Medical Science, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-Ku, Tokyo, 160-0023, Japan
| | - Akihiko Shimomura
- Department of Breast and Medical Oncology, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-Ku, Tokyo, 162-8655, Japan.
| |
Collapse
|
40
|
Zhang L, Pan J, Wang M, Yang J, Zhu S, Li L, Hu X, Wang Z, Pang L, Li P, Jia F, Ren G, Zhang Y, Xu D, Qiu F, Huang J. Chronic Stress-Induced and Tumor Derived SP1 + Exosomes Polarizing IL-1β + Neutrophils to Increase Lung Metastasis of Breast Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2310266. [PMID: 39630109 PMCID: PMC11789585 DOI: 10.1002/advs.202310266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 11/07/2024] [Indexed: 01/30/2025]
Abstract
Chronic stress can significantly promote breast cancer progression. When exposed to chronic stress, exosomes released from neural and neuroendocrine cells in the central nervous system are enhanced and modified. However, whether tumor-derived exosomes (TDEs) are influenced by chronic stress and participate in chronic stress-mediated distant metastasis remains unclear. Here, it is shown that chronic stress remarkably facilitates the secretion of TDEs and modifies the contents of exosomes by activating the adrenergic β receptor in 4T1 tumor-bearing mice. Exosomes injection and blockade experiments indicate that exosomes play a crucial role in chronic stress-mediated lung metastasis of breast cancer. Chronic stress-induced TDEs are internalized by pulmonary neutrophils and strengthen neutrophil recruitment via the CXCL2 autocrine. In addition, the level of SP1 in TDEs increases, which favors the secretion of IL-1β by neutrophils through the activation of the TLR4-NFκβ pathway, ultimately aggravating lung metastasis of breast cancer. Collectively, this study provides a novel mechanism by which neutrophils within a pre-metastatic niche acquire their inflamed phenotype and establishes an important link among neuroendocrine changes, exosomes, immunity, and metastasis.
Collapse
|
41
|
Wang Y, Xiong J, Ouyang K, Ling M, Luo J, Sun J, Xi Q, Chen T, Zhang Y. Extracellular vesicles: From large-scale production and engineering to clinical applications. J Tissue Eng 2025; 16:20417314251319474. [PMID: 40322740 PMCID: PMC12048759 DOI: 10.1177/20417314251319474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Indexed: 05/08/2025] Open
Abstract
Extracellular vesicles (EVs) have emerged as a promising strategy for treating a wide spectrum of pathologies, as they can deliver their cargo to recipient cells and regulate the signaling pathway of these cells to modulate their fate. Despite the great potential of EVs in clinical applications, their low yield and the challenges of cargo loading remain significant obstacles, hindering their transition from experimental research to clinical practice. Therefore, promoting EV release and enhancing EV cargo-loading are promising fields with substantial research potential and broad application prospects. In this review, we summarize the clinical applications of EVs, the methods and technologies for their large-scale production, engineering, and modification, as well as the challenges that must be addressed during their development. We also discuss the future perspectives of this exciting field of research to facilitate its transformation from bench to clinical reality.
Collapse
Affiliation(s)
- Yuxuan Wang
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, China
| | - Jiali Xiong
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, China
- College of Medicine, Jiaxing University, Jiaxing, Zhejiang, China
| | - Kun Ouyang
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, China
| | - Mingwang Ling
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, China
| | - Junyi Luo
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, China
| | - Jiajie Sun
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, China
| | - Qianyun Xi
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, China
| | - Ting Chen
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, China
| | - Yongliang Zhang
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, China
| |
Collapse
|
42
|
Huang C, Li J, Xie Z, Hu X, Huang Y. Relationship between exosomes and cancer: formation, diagnosis, and treatment. Int J Biol Sci 2025; 21:40-62. [PMID: 39744442 PMCID: PMC11667803 DOI: 10.7150/ijbs.95763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 11/02/2024] [Indexed: 01/11/2025] Open
Abstract
Exosomes are a member of extracellular vesicles. However, their biological characteristics differ from those of other vesicles, and recently, their powerful functions as information molecules, biomarkers, and carriers have been demonstrated. Malignancies are the leading cause of high morbidity and mortality worldwide. The cure rate of malignancies can be improved by improving early screening rates and therapy. Moreover, a close correlation between exosomes and malignancies has been observed. An in-depth study of exosomes can provide new methods for diagnosing and treating tumors. Therefore, this study aimed to review, sort, and summarize such achievements, and present ideas and opinions on the application of exosomes in tumor treatment.
Collapse
Affiliation(s)
- Chen Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jiajin Li
- Sichuan university, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Zichuan Xie
- Sichuan university, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Xiangjun Hu
- Sichuan university, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Yan Huang
- Health Management Center, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
- State Key Laboratory of Respiratory Health and Multimorbidity, China
- Research Laboratory for Prediction and Evaluation of Chronic Diseases in the Elderly, National Clinical Research Center for Geriatric Diseases, China
- General Practice Research Institute, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
43
|
Linares-Rodríguez M, Blancas I, Rodríguez-Serrano F. The Predictive Value of Blood-Derived Exosomal miRNAs as Biomarkers in Breast Cancer: A Systematic Review. Clin Breast Cancer 2025; 25:e48-e55.e15. [PMID: 39054208 DOI: 10.1016/j.clbc.2024.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 06/20/2024] [Accepted: 06/22/2024] [Indexed: 07/27/2024]
Abstract
Breast cancer (BC) remains a widespread disease worldwide, despite advances in its detection and treatment. microRNAs (miRNAs) play a significant role in cancer, and their presence within exosomes may confer several advantages in terms of tumor initiation, propagation, immune evasion, and drug resistance compared to freely circulating miRNAs in the blood. The objective of this study was to conduct a systematic review to analyze the role of exosomal miRNAs present in serum or plasma as biomarkers in BC. Bibliographic sources were collected from various databases with no starting date limit until March 2023. The search terms used were related to "breast cancer," "microRNAs," and "exosomes." Following the search, inclusion and exclusion criteria were applied, resulting in a total of 46 articles. Data were extracted from the selected studies and summarized to indicate the miRNAs, type of dysregulation, sample source, number of patients and controls, and clinical relevance of the miRNAs. We carried out an enrichment study of the microRNAs that appeared in at least 3 studies, those that were suitable for selection were miR-16, miR-21 and miR-155. Exosomal miRNAs isolated from blood samples of patients diagnosed with BC could be valuable in the clinical setting. They could provide information about early diagnosis, disease progression, recurrence, treatment response, and metastases. It is crucial to reach a consensus on the specific exosomal miRNAs to detect and the most appropriate type of sample for comprehensive utilization of miRNAs as biomarkers for BC.
Collapse
Affiliation(s)
- Marina Linares-Rodríguez
- Biopathology and Regenerative Medicine Institute (IBIMER), University of Granada, Granada, Spain
| | - Isabel Blancas
- Department of Medicine, School of Medicine, University of Granada, Granada, Spain; Department of Medical Oncology, San Cecilio University Hospital, Granada, Spain; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain.
| | - Fernando Rodríguez-Serrano
- Biopathology and Regenerative Medicine Institute (IBIMER), University of Granada, Granada, Spain; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain; Department of Human Anatomy and Embryology, University of Granada, Granada, Spain.
| |
Collapse
|
44
|
Meng X, Wang X, Zhang Z, Song L, Chen J. Recent Advancements of Nanomedicine in Breast Cancer Surgery. Int J Nanomedicine 2024; 19:14143-14169. [PMID: 39759962 PMCID: PMC11699852 DOI: 10.2147/ijn.s494364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 11/28/2024] [Indexed: 01/07/2025] Open
Abstract
Breast cancer surgery plays a pivotal role in the multidisciplinary approaches. Surgical techniques and objectives are gradually shifting from tumor complete resection towards prolonging survival, improving cosmetic outcomes, and restoring the social and psychological well-being of patients. However, surgical treatment still faces challenges such as inadequate sensitivity in sentinel lymph node localization, the need to improve intraoperative tumor boundary localization imaging, postoperative scar healing, and the risk of recurrence, necessitating other adjunct measures for improvement. To address these challenges, specificity-optimized nanomedicines have been introduced into the surgical therapeutic landscape of breast cancer. In particular, this review involves starting with an overview of breast structure and the composition of the tumor microenvironment and then introducing the guiding principle and foundation for the design of nanomedicine. Moreover, we will take the order process of breast cancer surgery diagnosis and treatment as the starting point, and adaptively propose the roles and advantages of nanomedicine in addressing the corresponding issues. Furthermore, we also involved the prospects of utilizing advanced technological approaches. Overall, this review seeks to uncover the sophisticated design and strategies of nanomedicine from a clinical standpoint, address the challenges faced in surgical treatment, and provide insights into this subject matter.
Collapse
Affiliation(s)
- Xiangyue Meng
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
- Breast Center, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Xin Wang
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
- Breast Center, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Zhihao Zhang
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
- Breast Center, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Linlin Song
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, 610041, People’s Republic of China
- Department of Ultrasound, Laboratory of Ultrasound Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Jie Chen
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
- Breast Center, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| |
Collapse
|
45
|
Leslie Salewon M, Pathak R, Dooley WC, Squires RA, Rui H, Chervoneva I, Tanaka T. Surgical delay-associated mortality risk varies by subtype in loco-regional breast cancer patients in SEER-Medicare. Breast Cancer Res 2024; 26:191. [PMID: 39736650 DOI: 10.1186/s13058-024-01949-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 12/14/2024] [Indexed: 01/01/2025] Open
Abstract
Substantial evidence supports that delay of surgery after breast cancer diagnosis is associated with increased mortality risk, leading to the introduction of a new Commission on Cancer quality measure for receipt of surgery within 60 days of diagnosis for non-neoadjuvant patients. Breast cancer subtype is a critical prognostic factor and determines treatment options; however, it remains unknown whether surgical delay-associated breast cancer-specific mortality (BCSM) risk differs by subtype. This retrospective cohort study aimed to assess whether the impact of delayed surgery on survival varies by subtype (hormone [HR] + /HER2 -, HR -/HER2 -, and HER2 +) in patients with loco-regional breast cancer who received surgery as their first treatment between 2010 and 2017 using the SEER-Medicare database. Exposure of this study was continuous time to surgery from diagnostic biopsy (TTS; days) in reference to TTS = 30 days. BCSM were evaluated as flexibly dependent on continuous time (days) to surgery from diagnosis (TTS) using Fine and Gray competing-risk regression models, respectively, by HR status. Inverse propensity score-weighting was adjusted for demographic, clinical, and treatment variables impacting TTS. Adjusted BCSM risk grew with increasing TTS across all subtypes; however, the pattern and extent of the association varied. HR + /HER2 - patients exhibited the most pronounced increase in BCSM risk associated with TTS, with approximately exponential growth after 42 days, with adjusted subdistribution hazard ratios (sHR) of 1.21 (95% CI: 1.06-1.37) at TTS = 60 days, 1.79 (95% CI: 1.40-2.29) at TTS = 90 days, and 2.83 (95% CI: 1.76-4.55) at TTS = 120 days. In contrast, both HER2 + and HR -/HER2 - patients showed slower, approximately linear growth in sHR, although non-significant in HR -HER2 -.
Collapse
Affiliation(s)
- Macall Leslie Salewon
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 975 NE, 10th, Oklahoma City, OK, 73104, USA
| | - Rashmi Pathak
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 975 NE, 10th, Oklahoma City, OK, 73104, USA
| | - William C Dooley
- Department of Surgery, School of Medicine, University of Oklahoma Health Sciences Center, 800 Stanton L. Young Blvd., Oklahoma City, OK, 73104, USA
| | - Ronald A Squires
- Department of Surgery, School of Medicine, University of Oklahoma Health Sciences Center, 800 Stanton L. Young Blvd., Oklahoma City, OK, 73104, USA
| | - Hallgeir Rui
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, 1015 Chestnut St., Suite 520, Philadelphia, PA, 19107, USA
- Division of Biostatistics, Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, 1015 Chestnut St., Suite 520, Philadelphia, PA, 19107, USA
| | - Inna Chervoneva
- Division of Biostatistics, Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, 1015 Chestnut St., Suite 520, Philadelphia, PA, 19107, USA.
| | - Takemi Tanaka
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 975 NE, 10th, Oklahoma City, OK, 73104, USA.
- Department of Pathology, School of Medicine, University of Oklahoma Health Sciences Center, 800 Stanton L. Young Blvd., Oklahoma City, OK, 73104, USA.
| |
Collapse
|
46
|
Kisar Tunca S, Unal R. Adipocyte-derived fatty acid uptake induces obesity-related breast cancer progression: a review. Mol Biol Rep 2024; 52:39. [PMID: 39644365 DOI: 10.1007/s11033-024-10139-x] [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: 04/24/2024] [Accepted: 11/25/2024] [Indexed: 12/09/2024]
Abstract
Obesity is a metabolic disorder that occurs when excess energy taken into the body is stored as fat. It is known that this metabolic imbalance affects the development of other diseases such as cancer, cardiovascular diseases, insulin resistance, and diabetes. The main cellular component of adipose tissue is adipocytes, and the environmental interactions of adipocytes are important to study the mechanism of disorder formation. Breast tissue is rich in adipose tissue and obesity is known to be an important risk factor in the development of breast cancer. Altered adipogenesis and lipogenesis processes in adipocytes in breast tissue support tumor development through the transfer of fatty acids released from adipocytes. We believe that blending adipocyte biology with breast cancer development is important for investigating the mechanisms that regulate breast tumor malignant behavior and providing new targets for treatment. Fatty acids, which are an energy source for breast cancer cells, are discussed from molecular perspectives in this review.
Collapse
Affiliation(s)
- Selin Kisar Tunca
- Faculty of Science, Department of Molecular Biology and Genetics, Mugla Sitki Kocman University, Mugla, Turkey
| | - Resat Unal
- Faculty of Science, Department of Molecular Biology and Genetics, Mugla Sitki Kocman University, Mugla, Turkey.
| |
Collapse
|
47
|
Martínez-Rodríguez A, Fuentes-Antrás J, Lorca V, López de Sá A, Pérez-Segura P, Moreno F, García-Sáenz JA, García-Barberán V. Molecular Profiling of Endocrine Resistance in HR+/HER2-Metastatic Breast Cancer: Insights from Extracellular Vesicles-Derived DNA and ctDNA in Liquid Biopsies. Int J Mol Sci 2024; 25:13045. [PMID: 39684756 DOI: 10.3390/ijms252313045] [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: 10/31/2024] [Revised: 11/27/2024] [Accepted: 12/01/2024] [Indexed: 12/18/2024] Open
Abstract
Standard treatments in hormone receptor-positive (HR+)/HER2-metastatic breast cancer (mBC) typically involve endocrine therapy (ET) combined with CDK4/6 inhibitors, yet resistance to ET remains a persistent challenge in advanced cases. A deeper knowledge of the use of liquid biopsy is crucial for the implementation of precision medicine in mBC with real-time treatment guidance. Our study assesses the prognostic value of PIK3CA and ESR1 mutations in DNA derived from extracellular vesicles (EV-DNA) in longitudinal plasma from 59 HR+/HER2-mBC patients previously exposed to aromatase inhibitors, with a comparative analysis against circulating tumor DNA (ctDNA). Mutations were evaluated by digital PCR. PIK3CA and ESR1 mutations were found in 22 and 25% of patients. Baseline ESR1 mutations in EV-DNA were associated with shorter progression-free survival (PFS) across the cohort, with the Y537S mutation showing a particularly strong impact on the outcome of fulvestrant-treated patients. In contrast, PIK3CA mutations in EV-DNA did not significantly correlate with PFS, whereas in ctDNA, they were linked to poor outcomes. Altogether, this study positions EV-DNA as a valuable biomarker alongside ctDNA, enriching the understanding of different analytes in liquid biopsy and supporting strategies for HR+/HER2-mBC in precision oncology.
Collapse
MESH Headings
- Humans
- Female
- Breast Neoplasms/genetics
- Breast Neoplasms/drug therapy
- Breast Neoplasms/pathology
- Breast Neoplasms/blood
- Extracellular Vesicles/metabolism
- Extracellular Vesicles/genetics
- Circulating Tumor DNA/genetics
- Circulating Tumor DNA/blood
- Liquid Biopsy/methods
- Middle Aged
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/metabolism
- Class I Phosphatidylinositol 3-Kinases/genetics
- Drug Resistance, Neoplasm/genetics
- Aged
- Mutation
- Estrogen Receptor alpha/genetics
- Estrogen Receptor alpha/metabolism
- Biomarkers, Tumor/genetics
- Adult
- Aromatase Inhibitors/therapeutic use
- Aromatase Inhibitors/pharmacology
- Prognosis
- Receptors, Estrogen/metabolism
- Receptors, Estrogen/genetics
- Receptors, Progesterone/metabolism
- Receptors, Progesterone/genetics
- Neoplasm Metastasis
- Antineoplastic Agents, Hormonal/therapeutic use
- Antineoplastic Agents, Hormonal/pharmacology
Collapse
Affiliation(s)
- Ana Martínez-Rodríguez
- "Clinical and Translational Research in Oncology" Group, Molecular Oncology Laboratory, Hospital Clinico San Carlos, Instituto de Investigación Sanitaria Hospital Clinico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Medical Oncology, Hospital Clinico San Carlos, Instituto de Investigación Sanitaria Hospital Clinico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Jesús Fuentes-Antrás
- Department of Medical Oncology, Hospital Clinico San Carlos, Instituto de Investigación Sanitaria Hospital Clinico San Carlos (IdISSC), 28040 Madrid, Spain
- NEXT Oncology Experimental Therapeutics Unit, Hospital Universitario Quironsalud Madrid, 28223 Madrid, Spain
| | - Víctor Lorca
- "Clinical and Translational Research in Oncology" Group, Molecular Oncology Laboratory, Hospital Clinico San Carlos, Instituto de Investigación Sanitaria Hospital Clinico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Alfonso López de Sá
- Department of Medical Oncology, Hospital Clinico San Carlos, Instituto de Investigación Sanitaria Hospital Clinico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Pedro Pérez-Segura
- Department of Medical Oncology, Hospital Clinico San Carlos, Instituto de Investigación Sanitaria Hospital Clinico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Fernando Moreno
- Department of Medical Oncology, Hospital Clinico San Carlos, Instituto de Investigación Sanitaria Hospital Clinico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Jose Angel García-Sáenz
- Department of Medical Oncology, Hospital Clinico San Carlos, Instituto de Investigación Sanitaria Hospital Clinico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Vanesa García-Barberán
- "Clinical and Translational Research in Oncology" Group, Molecular Oncology Laboratory, Hospital Clinico San Carlos, Instituto de Investigación Sanitaria Hospital Clinico San Carlos (IdISSC), 28040 Madrid, Spain
| |
Collapse
|
48
|
Ma L, Guo H, Zhao Y, Liu Z, Wang C, Bu J, Sun T, Wei J. Liquid biopsy in cancer current: status, challenges and future prospects. Signal Transduct Target Ther 2024; 9:336. [PMID: 39617822 PMCID: PMC11609310 DOI: 10.1038/s41392-024-02021-w] [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: 06/07/2024] [Revised: 09/10/2024] [Accepted: 10/14/2024] [Indexed: 12/06/2024] Open
Abstract
Cancer has a high mortality rate across the globe, and tissue biopsy remains the gold standard for tumor diagnosis due to its high level of laboratory standardization, good consistency of results, relatively stable samples, and high accuracy of results. However, there are still many limitations and drawbacks in the application of tissue biopsy in tumor. The emergence of liquid biopsy provides new ideas for early diagnosis and prognosis of tumor. Compared with tissue biopsy, liquid biopsy has many advantages in the diagnosis and treatment of various types of cancer, including non-invasive, quickly and so on. Currently, the application of liquid biopsy in tumor detection has received widely attention. It is now undergoing rapid progress, and it holds significant potential for future applications. Around now, liquid biopsies encompass several components such as circulating tumor cells, circulating tumor DNA, exosomes, microRNA, circulating RNA, tumor platelets, and tumor endothelial cells. In addition, advances in the identification of liquid biopsy indicators have significantly enhanced the possibility of utilizing liquid biopsies in clinical settings. In this review, we will discuss the application, advantages and challenges of liquid biopsy in some common tumors from the perspective of diverse systems of tumors, and look forward to its future development prospects in the field of cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Liwei Ma
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- Key Clinical Laboratory of Henan province, Zhengzhou, Henan, China.
| | - Huiling Guo
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Clinical Laboratory of Henan province, Zhengzhou, Henan, China
| | - Yunxiang Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhibo Liu
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Clinical Laboratory of Henan province, Zhengzhou, Henan, China
| | - Chenran Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Clinical Laboratory of Henan province, Zhengzhou, Henan, China
| | - Jiahao Bu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ting Sun
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- Key Clinical Laboratory of Henan province, Zhengzhou, Henan, China.
| | - Jianwei Wei
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
49
|
Massa D, Vernieri C, Nicolè L, Criscitiello C, Boissière-Michot F, Guiu S, Bobrie A, Griguolo G, Miglietta F, Vingiani A, Lobefaro R, Taurelli Salimbeni B, Pinato C, Schiavi F, Brich S, Pescia C, Fusco N, Pruneri G, Fassan M, Curigliano G, Guarneri V, Jacot W, Dieci MV. Immune and gene-expression profiling in estrogen receptor low and negative early breast cancer. J Natl Cancer Inst 2024; 116:1914-1927. [PMID: 39083015 PMCID: PMC11630536 DOI: 10.1093/jnci/djae178] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 06/25/2024] [Accepted: 07/23/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND The cutoff of <1% positive cells to define estrogen receptor (ER) negativity by immunohistochemistry (IHC) in breast cancer (BC) is debated. We explored the tumor immune microenvironment and gene-expression profile of patients with early-stage HER2-negative ER-low (ER 1%-9%) BC, comparing them to ER-negative (ER <1%) and ER-intermediate (ER 10%-50%) tumors. METHODS Among 921 patients with early-stage I-III, ER ≤50%, HER2-negative BCs, tumors were classified as ER-negative (n = 712), ER-low (n = 128), or ER-intermediate (n = 81). Tumor-infiltrating lymphocytes (TILs) were evaluated. CD8+, FOXP3+ cells, and PD-L1 status were assessed by IHC and quantified by digital pathology. We analyzed 776 BC-related genes in 116 samples. All tests were 2-sided at a <.05 significance level. RESULTS ER-low and ER-negative tumors exhibited similar median TILs, statistically significantly higher than ER-intermediate tumors. CD8/FOXP3 ratio and PD-L1 positivity rates were comparable between ER-low and ER-negative groups. These groups showed similar enrichment in basal-like intrinsic subtypes and comparable expression of immune-related genes. ER-low and ER-intermediate tumors showed significant transcriptomic differences. High TILs (≥30%) were associated with improved relapse-free survival (RFS) in ER-low (5-year RFS 78.6% vs 66.2%, log-rank P = .033, hazard ratio [HR] 0.37 [95% CI = 0.15 to 0.96]) and ER-negative patients (5-year RFS 85.2% vs 69.8%, log-rank P < .001, HR 0.41 [95% CI = 0.27 to 0.60]). CONCLUSIONS ER-low and ER-negative tumors are similar biological and molecular entities, supporting their comparable clinical outcomes and treatment responses, including to immunotherapy. Our findings contribute to the growing evidence calling for a reevaluation of ER-positive BC classification and management, aligning ER-low and ER-negative tumors more closely.
Collapse
Affiliation(s)
- Davide Massa
- Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padova, Padova, Italy
| | - Claudio Vernieri
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- IFOM ETS, The AIRC Institute of Molecular Oncology
| | | | - Carmen Criscitiello
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Division of Early Drug Development for Innovative Therapy, European Institute of Oncology IRCCS, Milan, Italy
| | | | - Séverine Guiu
- Department of Medical Oncology, Institut Régional Du Cancer de Montpellier (ICM), Montpellier, France
- Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Montpellier University, Montpellier, France
| | - Angélique Bobrie
- Department of Medical Oncology, Institut Régional Du Cancer de Montpellier (ICM), Montpellier, France
- Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Montpellier University, Montpellier, France
| | - Gaia Griguolo
- Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padova, Padova, Italy
| | - Federica Miglietta
- Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padova, Padova, Italy
| | - Andrea Vingiani
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Riccardo Lobefaro
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Beatrice Taurelli Salimbeni
- Division of Early Drug Development for Innovative Therapy, European Institute of Oncology IRCCS, Milan, Italy
| | - Claudia Pinato
- UOSD Hereditary Tumors, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Francesca Schiavi
- UOSD Hereditary Tumors, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Silvia Brich
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Carlo Pescia
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy
| | - Nicola Fusco
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy
| | - Giancarlo Pruneri
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Matteo Fassan
- Department of Medicine (DIMED), University of Padua, Padova, Italy
- Veneto Institute of Oncology IOV—IRCCS, Padova, Italy
| | - Giuseppe Curigliano
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Division of Early Drug Development for Innovative Therapy, European Institute of Oncology IRCCS, Milan, Italy
| | - Valentina Guarneri
- Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padova, Padova, Italy
| | - William Jacot
- Translational Research Unit, Institut du Cancer de Montpellier, Montpellier, France
- Department of Medical Oncology, Institut Régional Du Cancer de Montpellier (ICM), Montpellier, France
- Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Montpellier University, Montpellier, France
| | - Maria Vittoria Dieci
- Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padova, Padova, Italy
| |
Collapse
|
50
|
Mazzotta AD, Addeo P, Ielpo B, Ginesini M, Regenet N, Boggi U, Bachellier P, Soubrane O. Pancreatoduodenectomy after Ivor-Lewis Santi oesophagectomy with gastric tube reconstruction. An European multicentre experience. Surg Oncol 2024; 57:102144. [PMID: 39357095 DOI: 10.1016/j.suronc.2024.102144] [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: 06/11/2024] [Revised: 08/31/2024] [Accepted: 09/19/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Pancreaticoduodenectomy (PD) is the standard surgery to treat tumors and other conditions affecting the head of the pancreas. PD involves the division of the gastroduodenal artery (GDA) and its branches, to allow for complete dissection of lymph nodes. However, PD in patients with prior esophageal resection presents challenges due to altered anatomy and risks compromising gastric tube vascularization. GDA preservation becomes crucial to avoid ischemia, although this may pose oncological risks by potentially leaving behind regional lymph nodes. This article reviews European surgical center experiences and techniques for PD in patients with prior esophageal surgery, focusing on short-term outcomes. METHODS We have collected all the experiences carried out in European surgical centers and evaluated the techniques applied for PD in patients who had prior esophageal surgery while analyzing short-term outcomes. RESULTS Eight patients from 5 European centers were identified. Six patients were diagnosed with pancreatic adenocarcinoma, including one borderline case. Intraoperatively, the gastroduodenal artery (GDA) was preserved in all cases, with portal vein reconstruction required in only one instance due to tumor invasion. No ischemia or venous congestion of the gastric tube was observed during the surgical procedure. Post-operative complications that occurred included POPF type C in 1 (12.5 %), PPH type C in 1 (12.5 %). The median number of harvested lymph nodes was 21 [14-24]. with a median of 1.5 positive lymph nodes. R1 resection was present in 62.5 % of cases. CONCLUSION Performing pancreaticoduodenectomy subsequent to Ivor Lewis esophagectomy is a technical challenge, but seems feasiable and safe in selected patients. GDA-preserving pancreaticoduodenectomy emerges as a valuable and time-efficient variation of the conventional procedure, it can be considered oncologically appropriate, but studies confirming its long-term impact on radicality are still needed.
Collapse
Affiliation(s)
- Alessandro D Mazzotta
- Department of Digestive, Oncological and Metabolic Surgery, Institut Mutualiste Montsouris, Paris, France.
| | - Pietro Addeo
- Hepato-Pancreato-Biliary Surgery and Liver Transplantation, Université de Strasbourg, Strasbourg, France
| | - Benedetto Ielpo
- Hepatobiliary and Pancreatic Surgery Unit, Hospital del Mar. Universitat Pompeu Fabra, Barcelona, Spain
| | - Michael Ginesini
- Division of General and Transplant Surgery, University of Pisa, Pisa, Italy
| | - Nicolas Regenet
- Department of Digestive Surgery, Nantes Hospital, Nantes, France
| | - Ugo Boggi
- Division of General and Transplant Surgery, University of Pisa, Pisa, Italy
| | - Philippe Bachellier
- Hepato-Pancreato-Biliary Surgery and Liver Transplantation, Université de Strasbourg, Strasbourg, France
| | - Olivier Soubrane
- Department of Digestive, Oncological and Metabolic Surgery, Institut Mutualiste Montsouris, Paris, France
| |
Collapse
|