1
|
Chen Y, Cui F, Wu X, Zhao W, Xia Q. The expression and clinical significance of serum exosomal-long non-coding RNA DLEU1 in patients with cervical cancer. Ann Med 2025; 57:2442537. [PMID: 39687982 DOI: 10.1080/07853890.2024.2442537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/25/2024] [Accepted: 11/23/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Accumulating evidence has demonstrated that the long non-coding RNA (lncRNA) lymphocytic leukaemia deletion gene 1 (DLEU1) is abnormally overexpressed in many cancer types, including cervical cancer (CC). However, the potential clinical significance of DLEU1 in serum exosomes of patients with CC remains unclear. METHODS The expression of serum exosomal DLEU1 was detected by quantitative real-time polymerase chain reaction (qRT-PCR). A receiver operating characteristic (ROC) curve was plotted to evaluate the clinical diagnostic efficacy of DLEU1. The Kaplan-Meier survival curve and Cox proportional hazards model were used to assess the effect of DLEU1 on postoperative recurrence, metastasis and prognosis among patients with CC. RESULTS Our research showed that DLEU1 expression in the serum exosomes of patients with CC was significantly upregulated compared to that in patients with cervical intraepithelial neoplasia (CIN) and healthy controls (HCs) (both p < .001). DLEU1 relative expression was significantly correlated with tumour size, cervical invasion depth, pathological grade, International Federation of Gynecology and Obstetrics (FIGO) stage and lymph node metastasis among patients with CC (p < .01 all). The combined detection of DLEU1, carbohydrate antigen 125 (CA-125) and squamous cell carcinoma (SCC) exhibited significantly higher diagnostic efficiency (p < .01). Furthermore, the overall survival (OS) and disease-free survival (DFS) of CC patients in the high DLEU1 expression group were markedly lower than those in the low DLEU1 expression group (both p < .01). Cox univariate and multivariate regression analyses indicated that DLEU1 was an independent risk factor for postoperative recurrence and metastasis in CC patients. CONCLUSIONS Our findings suggest that serum exosome DLEU1 has certain clinical value for diagnosing, monitoring recurrence and metastasis, and evaluating CC prognosis.
Collapse
Affiliation(s)
- Yu Chen
- Department of Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, PR China
| | - Facai Cui
- Department of Clinical Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Xiaoyu Wu
- Department of Clinical Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Weifeng Zhao
- Department of Oncology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Qingxin Xia
- Department of Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, PR China
| |
Collapse
|
2
|
Ruiz-Torres DA, Bryan ME, Hirayama S, Merkin RD, Luciani E, Roberts TJ, Patel M, Park JC, Wirth LJ, Sadow PM, Sade-Feldman M, Stott SL, Faden DL. Spatial characterization of tertiary lymphoid structures as predictive biomarkers for immune checkpoint blockade in head and neck squamous cell carcinoma. Oncoimmunology 2025; 14:2466308. [PMID: 39963988 PMCID: PMC11845054 DOI: 10.1080/2162402x.2025.2466308] [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: 11/13/2024] [Revised: 01/27/2025] [Accepted: 02/08/2025] [Indexed: 02/23/2025] Open
Abstract
Immune checkpoint blockade (ICB) is the standard of care for recurrent/metastatic head and neck squamous cell carcinoma (HNSCC), yet efficacy remains low. The combined positive score (CPS) for PD-L1 is the only biomarker approved to predict response to ICB and has limited performance. Tertiary Lymphoid Structures (TLS) have shown promising potential for predicting response to ICB. However, their exact composition, size, and spatial biology in HNSCC remain understudied. To elucidate the impact of TLS spatial biology in response to ICB, we utilized pre-ICB tumor tissue sections from 9 responders (complete response, partial response, or stable disease) and 11 non-responders (progressive disease) classified via RECISTv1.1. A custom multi-immunofluorescence (mIF) staining assay was applied to characterize tumor cells (pan-cytokeratin), T cells (CD4, CD8), B cells (CD19, CD20), myeloid cells (CD16, CD56, CD163), dendritic cells (LAMP3), fibroblasts (α Smooth Muscle Actin), proliferative status (Ki67) and immunoregulatory molecules (PD1). A machine learning model was employed to measure the effect of spatial metrics on achieving a response to ICB. A higher density of B cells (CD20+) was found in responders compared to non-responders to ICB (p = 0.022). The presence of TLS within 100 µm of the tumor was associated with improved overall (p = 0.04) and progression-free survival (p = 0.03). A multivariate machine learning model identified TLS density as a leading predictor of response to ICB with 80% accuracy. Immune cell densities and TLS spatial location play a critical role in the response to ICB in HNSCC and may potentially outperform CPS as a predictor of response.
Collapse
Affiliation(s)
- Daniel A. Ruiz-Torres
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear, Boston, MA, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michael E. Bryan
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Shun Hirayama
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear, Boston, MA, USA
| | - Ross D. Merkin
- Department of Medicine, Harvard Medical School, Boston MA, USA
- Department of Medicine, Center for Head and Neck Cancers, Massachusetts General Hospital, Boston MA, USA
| | - Evelyn Luciani
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Thomas J. Roberts
- Department of Medicine, Harvard Medical School, Boston MA, USA
- Department of Medicine, Center for Head and Neck Cancers, Massachusetts General Hospital, Boston MA, USA
| | - Manisha Patel
- Department of Medicine, Harvard Medical School, Boston MA, USA
- Department of Medicine, Center for Head and Neck Cancers, Massachusetts General Hospital, Boston MA, USA
| | - Jong C. Park
- Department of Medicine, Harvard Medical School, Boston MA, USA
- Department of Medicine, Center for Head and Neck Cancers, Massachusetts General Hospital, Boston MA, USA
| | - Lori J. Wirth
- Department of Medicine, Harvard Medical School, Boston MA, USA
- Department of Medicine, Center for Head and Neck Cancers, Massachusetts General Hospital, Boston MA, USA
| | - Peter M. Sadow
- Department of Medicine, Harvard Medical School, Boston MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Moshe Sade-Feldman
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Shannon L. Stott
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Engineering in Medicine and BioMEMS Resource Center, Surgical Services, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Daniel L. Faden
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Center for Head and Neck Cancers, Massachusetts General Hospital, Boston MA, USA
| |
Collapse
|
3
|
Song H, Kim D, Jang SJ, Hwang HS, Song JS. Clinicopathologic features of histologic transformation in lung adenocarcinoma after treatment with epidermal growth factor receptor-tyrosine kinase inhibitors. Ann Diagn Pathol 2025; 77:152478. [PMID: 40215564 DOI: 10.1016/j.anndiagpath.2025.152478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/01/2025] [Accepted: 04/04/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) may lead to drug resistance, and the underlying mechanism may involve histologic transformations to small cell carcinoma (SCC), squamous cell carcinoma (SqCC), and sarcomatoid carcinoma (SC). Although there are reports regarding these transformations, comprehensive analyses are limited. METHODS A total of 233 patients with primary lung adenocarcinoma treated with EGFR-TKIs were reviewed. Among them, 26 patients (11.1 %) showed histologic transformation. RESULTS Eleven patients (42.3 %) showed SCC and SqCC transformations respectively, and four patients (15.4 %) showed SC transformation. The median time from TKI initiation to transformation was 19.8 months (6.8-51.4) for SCC, 45.3 months (2.4-101.5) for SqCC, and 11.8 months (6.8-15.7) for SC. The median overall survival (OS) was 41.8 months (12.5-78.9), 72.6 months (18.8-112.7), and 23.7 months (17.4-34.4), respectively. The survival from transformation was 12.3 months (2.1-28.3), 16.9 months (0.7-43.2), and 11.4 months (1.6-23.5), respectively. The most common mutations were TP53, PTEN, and RB1 in SCC; TP53 and RB1 in SqCC; and TP53 and KMT2D in SC. SC transformation had the worst OS, followed by SCC and SqCC (p < 0.001). This prognosis difference was also reflected in the time to transformation after EGFR-TKI treatment (p = 0.005). However, survival after transformation was not associated with tumor subtypes (p = 0.536). CONCLUSIONS The analysis of mutation profiles and survival outcomes revealed that the transformation subtype affects prognosis. Additionally, the time taken to undergo transformation is critical for patient outcomes.
Collapse
Affiliation(s)
- Halim Song
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea; Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05505, South Korea
| | - Deokhoon Kim
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea; Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05505, South Korea
| | - Se Jin Jang
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea; Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05505, South Korea
| | - Hee Sang Hwang
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea; Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05505, South Korea
| | - Joon Seon Song
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea; Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05505, South Korea.
| |
Collapse
|
4
|
Tai YJ, Lee CC, Chen YC, You SL, Chiang YC. Warning factors of metachronous uterine cancer in patients with breast cancer: a real-world nationwide cohort study. Gynecol Oncol Rep 2025; 59:101732. [PMID: 40270980 PMCID: PMC12013394 DOI: 10.1016/j.gore.2025.101732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 03/24/2025] [Accepted: 03/28/2025] [Indexed: 04/25/2025] Open
Abstract
Objective This study aimed to explore the risk factors for metachronous uterine cancer, including age at diagnosis, stage, body mass index (BMI), medical history and tamoxifen treatment in women with breast cancer to enable tailored risk-based gynecological surveillance. Methods A nationwide population-based cohort study was conducted. Data were retrieved from the Taiwan National Health Insurance Research Database and Taiwan Cancer Registry from 2011 to 2019. The risk of metachronous uterine cancer was analyzed by Cox regression model. Results There were 307 patients identified with uterine cancer among 114,906 patients with breast cancer. The incidence was 37.46 cases per 100,000 person-years among tamoxifen non-users (69 cases per 184,207 person-years). Among tamoxifen users (n = 58,227), the incidence rates were 105.52 cases (79 cases per 74,869 person-years) for <1 year of use, 111.62 cases (111 cases per 99,444 person-years) for 1-3 years, 58.19 cases (31 cases per 53,276 person-years) for 3-5 years, and 62.12 cases (17 cases per 27,366 person-years) for ≥5 years. The hazard ratio (HR) for uterine cancer was 3.06 (95 % confidence interval [CI] 2.14-4.39), 3.03 (95 % CI 2.15-4.28), 1.61 (95 % CI 1.01-2.57), and 1.77 (95 % CI 1.00-3.13) for patients on tamoxifen for <1, 1-3, 3-5, and ≥5 years. High BMI (≥25 kg/m2) was associated with an increased risk (HR 2.46, 95 % CI 1.07-5.64). Abnormal uterine bleeding was a significant predictor regardless of the clinical or sonographic diagnosis of endometrial lesions. Conclusion Routine ultrasound is not recommended for the detection of metachronous uterine cancer, and gynecologic investigations should focus on breast cancer patients with high BMI, tamoxifen use, and abnormal uterine bleeding.
Collapse
Affiliation(s)
- Yi-Jou Tai
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei, Taiwan
- Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chung-Chen Lee
- Data Science Center, College of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Yong-Chen Chen
- School of Medicine, College of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - San-Lin You
- School of Medicine, College of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Ying-Cheng Chiang
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei, Taiwan
- Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Hsin-Chu Branch, Hsin-Chu City, Taiwan
| |
Collapse
|
5
|
Ahn JH, Lee SJ, Yang SH, Kim JY, Park HS, Kim SI, Park BW, Park S. Clinical treatment score post-5 years and survival benefit from extended endocrine therapy for breast cancer patients under and over 50 years of age. Breast Cancer Res Treat 2025; 211:657-667. [PMID: 40097770 PMCID: PMC12031770 DOI: 10.1007/s10549-025-07679-6] [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: 06/03/2024] [Accepted: 03/05/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND This study aimed to determine whether clinical treatment score post-5 years (CTS5) could predict the clinical benefits of extended endocrine therapy (ExET) in young and old patients. METHODS We reviewed 2495 hormone receptor-positive breast cancer patients treated between 2001 and 2012 who were free from recurrence or death during the 5 years post-surgery in South Korea. The cohort was analyzed separately based on age (≤ 50 years and > 50 years). Multivariable analysis was conducted, and a cutoff of CTS5 < 3.13 was defined as the low group and CTS5 ≥ 3.13 as the intermediate/high (int/high) group. RESULTS The median follow-up duration was 115 months. Regardless of young and old age at diagnosis, the low group displayed considerably enhanced disease-free survival. Multivariate analysis revealed that the low group emerged as an independent and favorable prognostic factor for disease-free survival after adjusting for ExET use and prognostic parameters. Patients in the low group demonstrated a trend toward improved overall survival compared to those in the int/high group, reaching marginal statistical significance. ExET use demonstrated a significant correlation with improved disease-free survival, particularly in patients aged ≤ 50 years. CONCLUSIONS ExET should be considered in premenopausal and postmenopausal breast cancer patients with high CTS5 levels.
Collapse
Affiliation(s)
- Jee Hyun Ahn
- Division of Breast Surgery, Department of Surgery, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Republic of Korea
| | - Suk Jun Lee
- Division of Breast Surgery, Department of Surgery, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Republic of Korea
| | - Seung Hye Yang
- Division of Breast Surgery, Department of Surgery, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Republic of Korea
| | - Jee Ye Kim
- Division of Breast Surgery, Department of Surgery, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Republic of Korea
| | - Hyung Seok Park
- Division of Breast Surgery, Department of Surgery, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Republic of Korea
| | - Seung Il Kim
- Division of Breast Surgery, Department of Surgery, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Republic of Korea
| | - Byeong-Woo Park
- Division of Breast Surgery, Department of Surgery, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Republic of Korea
| | - Seho Park
- Division of Breast Surgery, Department of Surgery, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|
6
|
Khidr WA, Alfarttoosi KH, Taher WM, Alwan M, Ali Al-Nuaimi AM, Jawad MJ. A review of the role of tumor-derived exosomes in cancers treatment and progression. Int Immunopharmacol 2025; 157:114782. [PMID: 40334624 DOI: 10.1016/j.intimp.2025.114782] [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: 03/11/2025] [Revised: 04/23/2025] [Accepted: 04/29/2025] [Indexed: 05/09/2025]
Abstract
Tumor cells (TCs) produce exosomes (EXOs), nanovesicles formed in endosomes. Tumor-derived exosomes (TDEs) are tiny, bubble-shaped structures formed by TCs that include microRNAs (miRNA), proteins, enzymes, and copies of DNA and RNA. Many different kinds of cancer rely on TDEs. For instance, TDEs play a large role in the tumor microenvironment (TME) and promote tumor spread via many pathways. Furthermore, TDEs impact the efficacy of cancer treatments. Additionally, because of their low immunogenicity, high biocompatibility, and low toxicity, TDEs have been extensively used as drug delivery vehicles for cancer immunotherapy. Consequently, future cancer treatments may benefit from focusing on both the therapeutic function and the tumorigenic pathways of TDEs. Consequently, in this work, we have examined the roles of TDEs in cancer development, such as tumor angiogenesis, immune system evasion, and tumor metastasis. Then, we reviewed TDEs used to transport anticancer medicines, including chemotherapeutic medications, therapeutic compounds (including miRNA), and anticancer nanoparticles. We have concluded by outlining the challenges of clinical translation, including carcinogenicity and medication resistance, and by offering some suggestions for addressing these issues.
Collapse
Affiliation(s)
- Wajida Ataallah Khidr
- Department of Medical Laboratory Technics, College of Health and Medical Technology, Alnoor University, Mosul, Iraq
| | | | - Waam Mohammed Taher
- College of Nursing, National University of Science and Technology, Dhi Qar, Iraq
| | | | | | | |
Collapse
|
7
|
Liu N, Wu T, Han G, Chen M. Exosome-mediated ferroptosis in the tumor microenvironment: from molecular mechanisms to clinical application. Cell Death Discov 2025; 11:221. [PMID: 40328736 PMCID: PMC12056189 DOI: 10.1038/s41420-025-02484-y] [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: 02/15/2025] [Revised: 04/01/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
Ferroptosis in the tumor microenvironment (TME) plays a crucial role in the development, metastasis, immune escape, and drug resistance of various types of cancer. A better understanding of ferroptosis in the TME could illuminate novel aspects of this process and promote the development of targeted therapies. Compelling evidence indicates that exosomes are key mediators in regulating the TME. In this respect, it is now understood that exosomes can deliver biologically functional molecules to recipient cells, influencing cancer progression by reprogramming the metabolism of cancer cells and their surrounding stromal cells through ferroptosis. In this review, we focus on the role of exosomes in the TME and describe how they contribute to tumor reprogramming, immunosuppression, and the formation of pre-metastatic niches through ferroptosis. In addition, we highlight exosome-mediated ferroptosis as a potential target for cancer therapy and discuss strategies employing exosomes in ferroptosis treatment. Finally, we outline the current applications and challenges of targeted exosome-mediated ferroptosis therapy in tumor immunotherapy and chemotherapy. Our aim is to advance research on the link between exosomes and ferroptosis in the TME, and we pose questions to guide future studies in this area.
Collapse
Affiliation(s)
- Na Liu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Tianqing Wu
- XJTLU Wisdom Lake Academy of Pharmacy, Suzhou, Jiangsu Province, China
| | - Guohu Han
- Department of Oncology, Jingjiang People's Hospital Affiliated with Yangzhou University, Jingjiang, China
| | - Minbin Chen
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China.
| |
Collapse
|
8
|
Ng D, Cyr D, Khan S, Dossa F, Swallow C, Kazazian K. Molecular mechanisms of metastatic peritoneal dissemination in gastric adenocarcinoma. Cancer Metastasis Rev 2025; 44:50. [PMID: 40317360 PMCID: PMC12049340 DOI: 10.1007/s10555-025-10265-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 04/17/2025] [Indexed: 05/07/2025]
Abstract
Peritoneal dissemination portends a dismal prognosis in patients with gastric adenocarcinoma in the context of limited effective treatments. The underlying cellular processes that drive gastric peritoneal carcinomatosis remain unclear, limiting the application of novel targeted therapies. In this comprehensive review, we aimed to identify and summarize all existing context-dependent molecular mechanisms that have been implicated in peritoneal dissemination and peritoneal carcinomatosis establishment from primary gastric adenocarcinoma. We applied a multilevel examination including data from in vivo murine models using human gastric cancer cell lines, in vitro technique-based studies, ex vivo models, and genomic/proteomic and molecular profiling analyses to report on various aspects of gastric cancer peritoneal metastasis biology. Mechanisms promoting peritoneal dissemination were grouped into three main functional categories: (1) intrinsic cancer cell biology, (2) cancer cell-peritoneal surface adhesion, and (3) peritoneal tumor microenvironment. We identified significant overlap among the three categories, indicating a complex interplay between multiple molecular mechanisms. By interrupting these pathways, peritoneal-directed therapies have the potential to improve quality and length of life in patients with high-risk primary gastric cancer.
Collapse
Affiliation(s)
- Deanna Ng
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
- Department of Surgery, University of Toronto, Toronto, Canada
| | - David Cyr
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
- Department of Surgery, University of Toronto, Toronto, Canada
| | - Shawn Khan
- Institute of Medical Science, University of Toronto, Toronto, Canada
- Department of Surgery, University of Toronto, Toronto, Canada
| | - Fahima Dossa
- Complex General Surgical Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Carol Swallow
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
- Department of Surgery, University of Toronto, Toronto, Canada
| | - Karineh Kazazian
- Department of Surgery, University of Toronto, Toronto, Canada.
- Department of Surgical Oncology, Toronto General Hospital, University Health Network, 200 Elizabeth Street, 10 Eaton North, Room 219, Toronto, M5G 2 C4, Canada.
| |
Collapse
|
9
|
Hsiao CY, Hsieh SW, Yang YH, Hsieh HM. Longitudinal trends in dementia prevalence among individuals with type 2 diabetes in Taiwan (2009-2022). J Diabetes Investig 2025. [PMID: 40317911 DOI: 10.1111/jdi.70060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/02/2025] [Accepted: 04/19/2025] [Indexed: 05/07/2025] Open
Abstract
OBJECTIVE This study examined the population-based epidemiological prevalence of early- and late-onset dementia in individuals with type 2 diabetes mellitus (T2DM) compared with the general population in Taiwan from 2009 to 2022. METHODS A 14-year nationwide population-based epidemiology study from 2009 to 2022 in Taiwan using the Taiwan National Health Insurance Research Database was conducted to examine the crude and age-standardized dementia prevalence in individuals aged <65 and ≥65. Multivariable logistic regression was used to assess factors associated with dementia prevalence in the T2DM population. RESULTS Among individuals aged ≥65 with T2DM, age-standardized dementia prevalence increased from 5.83 per 100 in 2009 to 7.11 per 100 in 2022, while the crude prevalence ratio compared to the general population ranged from 1.19 to 1.08. In the 40-64 age group, standardized dementia prevalence ranged from 0.58 to 0.43 per 100, with crude ratios ranging from 1.76 to 1.41. Our study found a higher risk of dementia prevalence among individuals aged 65 years and older, particularly among those with T2DM. Although the overall prevalence of dementia in the 40-64 age group was lower compared to older adults, individuals in this younger group with T2DM exhibited a significantly higher risk of developing dementia. Given these findings, public policy should prioritize early preventive strategies for individuals with T2DM to monitor and mitigate dementia progression in patients with T2DM.
Collapse
Affiliation(s)
- Chen-Yang Hsiao
- Department of Public Health, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Sun-Wung Hsieh
- Department of Neurology, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Neurology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Neuroscience Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Public Health, College of Health Sciences, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yuan-Han Yang
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Neurology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Neuroscience Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Neurology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Master's Program in Neurology, Faculty of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Chinese Mentality Protection Association, Kaohsiung, Taiwan
| | - Hui-Min Hsieh
- Department of Public Health, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Center for Big Data Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
10
|
Kyriacou NM, Gross AS, McLachlan AJ. Inter-Ethnic Differences in the Efficacy and Safety of Tyrosine Kinase Inhibitors Used in Oncology: Insights From Phase 3 Clinical Trials. Clin Transl Sci 2025; 18:e70224. [PMID: 40296413 PMCID: PMC12037692 DOI: 10.1111/cts.70224] [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: 12/30/2024] [Revised: 02/20/2025] [Accepted: 03/31/2025] [Indexed: 04/30/2025] Open
Abstract
Differences in the efficacy and safety of tyrosine kinase inhibitors (TKIs) have been observed across ethnic/ancestry subpopulations (previously reviewed to 2017). With an expanding number of TKIs approved since that time, an updated review of TKI response across ethnic/ancestry subpopulations in Phase 3 TKI clinical trials was conducted. A total of 73 population subgroup analyses (defined by participant race, ethnicity, ancestry or geographic region) of progression-free survival (PFS) and/or overall survival (OS) were identified by a literature search. Twelve (16%) of the analyses investigating the efficacy of afatinib, brigatinib, dacomitinib, gilteritinib, lorlatinib, neratinib, osimertinib, or pazopanib were assessed to report population differences in PFS and/or OS. For 28 (38%) of the analyses that showed suggestions of a potential efficacy difference across subpopulations, limitations in the data available precluded further assessment. There were 17 (23%) analyses assessed to report comparable efficacy outcomes across diverse subpopulations. The majority of clinical trials noted no clinically remarkable differences in safety between subpopulations; however, for brigatinib, crizotinib, pazopanib, and sunitinib, distinct patterns of adverse events were reported in the Asian and non-Asian subgroups. The underrepresentation of specific subpopulations, the grouping together of results of diverse subpopulations, as well as inconsistencies in the definition and reporting of participant ethnicity/ancestry are barriers to the meaningful exploration of inter-ethnic differences in TKI response. Therefore, further insight into the associations between ethnicity/ancestry and TKI response will require an increase in the diversity of clinical trial participants and appropriate analysis and reporting of subpopulation results.
Collapse
Affiliation(s)
- Nicki M. Kyriacou
- Sydney Pharmacy School, Faculty of Medicine and HealthUniversity of SydneySydneyAustralia
| | - Annette S. Gross
- Sydney Pharmacy School, Faculty of Medicine and HealthUniversity of SydneySydneyAustralia
| | - Andrew J. McLachlan
- Sydney Pharmacy School, Faculty of Medicine and HealthUniversity of SydneySydneyAustralia
| |
Collapse
|
11
|
Azumi M, Inubushi S, Yano Y, Obata K, Yamanaka K, Terai Y. MiR-575 in Exosomes of Vaginal Discharge Is Downregulated in Ovarian Cancer Patients. Cancer Genomics Proteomics 2025; 22:382-396. [PMID: 40280723 PMCID: PMC12041870 DOI: 10.21873/cgp.20508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 01/28/2025] [Accepted: 02/07/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND/AIM Ovarian cancer is asymptomatic in its early stages, and often diagnosed at advanced stages, leading to a high recurrence rate. In recent years, exosomes have been shown to be useful for early-detection, prognosis prediction, and treatment of cancer. Although many studies of cancer-related exosomes using other bodily fluids have been reported, there are few studies examining vaginal discharge, but none related to ovarian cancer. In this study, we investigated a method for early-detection of ovarian cancer using vaginal discharge, which are physically close to the fallopian tubes, where ovarian cancer originates, and can be easily collected from outside the body. MATERIALS AND METHODS Vaginal discharge was collected from 30 patients with ovarian cancer and 29 patients with benign gynecological diseases, and exosomal miRNAs were extracted. Samples from each group were submitted to miRNA microarray in order to examine miRNAs with significant differences in expression levels. We further narrowed down the list to four miRNAs based on literature and microarray data and examined the expression levels of miRNAs in the malignant and benign groups by RT-qPCR. RESULTS MiR-575 expression was significantly decreased in the malignant group compared to the benign group (p=0.00861). qPCR results were analyzed for several patient characteristics and no significant differences were found. CONCLUSION This is the first study to investigate exosomal miRNAs in vaginal discharge of ovarian cancer. Exosomal miR-575 in vaginal discharge may be used as a biomarker for ovarian cancer.
Collapse
Affiliation(s)
- Maho Azumi
- Department of Obstetrics and Gynecology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Sachiko Inubushi
- Department of Breast and Endocrine Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yoko Yano
- Department of Obstetrics and Gynecology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kenta Obata
- Department of Obstetrics and Gynecology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Keitaro Yamanaka
- Department of Obstetrics and Gynecology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yoshito Terai
- Department of Obstetrics and Gynecology, Kobe University Graduate School of Medicine, Kobe, Japan;
| |
Collapse
|
12
|
Jo H, Lee S, Choi S, Kishimoto M, Lee K. Gallbladder and Small Intestinal Luminal Opacification by Vicarious Contrast Medium Excretion Can be Observed on Delayed Computed Tomography in Healthy Dogs. Vet Radiol Ultrasound 2025; 66:e70033. [PMID: 40312826 PMCID: PMC12046108 DOI: 10.1111/vru.70033] [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: 11/18/2024] [Revised: 03/24/2025] [Accepted: 04/13/2025] [Indexed: 05/03/2025] Open
Abstract
Vicarious excretion is a nonrenal pathway of excretion for intravenously injected iodinated contrast media, with a limited understanding of its influencing factors and imaging features. In this prospective pilot study, gallbladder opacification (GBO) and small intestinal luminal opacification (SILO) were assessed to identify vicarious excretion patterns following intravenous contrast media administration for CT in clinically healthy dogs. Eight beagles were studied using a crossover method, divided into fed and fasted groups. The fed group was fed at 5 and 13 h after the first CT scan, while the fasted group was fed only at 13 h. Noncontrast and postcontrast CT scans were performed at 90 s, 10 min, 1 h, 4 h, 12 h, and 24 h after iohexol injection. The GBO was subjectively scored from grade 0 to 5 based on the attenuation value and area of opacification. The SILO was evaluated subjectively based on contrast intensity (weak or marked) and distribution. The GBO was observed from 1 h after contrast injection. Significant differences were noted in median GBO scores within the groups at 4, 12, and 24 h on Friedman's test followed by the post hoc Wilcoxon signed-rank test. The scores were significantly higher in the fasted group at 12 h on the Wilcoxon signed-rank test. The SILO occurred 10 min after contrast administration, with various distributions. In conclusion, GBO and SILO can be observed during delayed CT phases, and fasting increases the intensity and duration of GBO in clinically healthy dogs. These findings should not be interpreted as pathological changes.
Collapse
Affiliation(s)
- Hyeeun Jo
- College of Veterinary MedicineKyungpook National UniversityDaeguRepublic of Korea
| | - Sang‐Kwon Lee
- College of Veterinary MedicineKyungpook National UniversityDaeguRepublic of Korea
| | - Sooyoung Choi
- College of Veterinary MedicineKangwon National UniversityChuncheonRepublic of Korea
| | - Miori Kishimoto
- Cooperative Department of Veterinary MedicineTokyo University of Agriculture and TechnologyTokyoJapan
| | - Kija Lee
- College of Veterinary MedicineKyungpook National UniversityDaeguRepublic of Korea
| |
Collapse
|
13
|
Abida, Alhuthali HM, Alshehri JM, Alkathiri A, Almaghrabi ROM, Alsaeed SS, Albebi SAH, Almethn RM, Alfuraydi BA, Alharbi SB, Kamal M, Imran M. Exosomes in infectious diseases: insights into leishmaniasis pathogenesis, immune modulation, and therapeutic potential. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4913-4931. [PMID: 39702600 DOI: 10.1007/s00210-024-03702-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 12/02/2024] [Indexed: 12/21/2024]
Abstract
Leishmaniasis continues to be a critical international health issue due to the scarcity of efficient treatment and the development of drug tolerance. New developments in the research of extracellular vesicles (EVs), especially exosomes, have revealed novel disease management approaches. Exosomes are small vesicles that transport lipids, nucleic acids, and proteins in cell signalling. Its biogenesis depends on several cellular processes, and their functions in immune response, encompassing innate and adaptive immunity, underline their function in the pathogen-host interface. Exosomes play a significant role in the pathogenesis of some parasitic infections, especially Leishmaniasis, by helping parasites escape host immunity and promote disease progression. This article explains that in the framework of parasitic diseases, exosomes can act as master regulators that define the pathogenesis of the disease, as illustrated by the engagement of exosomes in the Leishmaniasis parasite and immune escape processes. Based on many published articles on Leishmaniasis, this review aims to summarize the biogenesis of exosomes, the properties of the cargo in exosomes, and the modulation of immune responses. We delve deeper into the prospect of using exosomes for the therapy of Leishmaniasis based on the possibility of using these extracellular vesicles for drug delivery and as diagnostic and prognostic biomarkers. Lastly, we focus on the recent research perspectives and future developments, underlining the necessity to continue the investigation of exosome-mediated approaches in Leishmaniasis treatment. Thus, this review intends to draw attention to exosomes as a bright new perspective in the battle against this disabling affliction.
Collapse
Affiliation(s)
- Abida
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, 91911, Rafha, Saudi Arabia
| | - Hayaa M Alhuthali
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
| | - Jawaher Mohammad Alshehri
- Optometry Department, Faculty of Applied Medical Sciences, Albaha University, 65431, Albaha, Saudi Arabia
| | - Afnan Alkathiri
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Albaha University, 65431, Albaha, Saudi Arabia
| | - Ruba Omar M Almaghrabi
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Albaha University, 65431, Albaha, Saudi Arabia
| | | | | | | | | | | | - Mehnaz Kamal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, 11942, Al-Kharj, Saudi Arabia
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, 91911, Rafha, Saudi Arabia.
- Center for Health Research, Northern Border University, Arar, Saudi Arabia.
| |
Collapse
|
14
|
Raman R, Debata S, Govindarajan T, Kumar P. Targeting Triple-Negative Breast Cancer: Resistance Mechanisms and Therapeutic Advancements. Cancer Med 2025; 14:e70803. [PMID: 40318146 PMCID: PMC12048392 DOI: 10.1002/cam4.70803] [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: 07/10/2024] [Revised: 02/12/2025] [Accepted: 03/11/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is one of the most heterogeneous and menacing forms of breast cancer, with no sustainable cure available in the current treatment landscape. Its lack of targets makes it highly unresponsive to various treatment modalities, which is why chemotherapy continues to be the primary form of treatment, despite the high rates of patients developing chemoresistance. In recent years, however, there has been significant progress in identifying and understanding the role of several aspects that might contribute to genomic instability and other hallmarks of cancer, including cellular proteins, immune targets, and epigenetic mechanisms, which are desirable as they permit reversibility easier than the often-adamant genetic changes. METHODS A literature review was conducted on the role of various TNBC associated biomarkers, their therapeutic applications, and their role in tumorigenesis and tumor maintenance, with a focus on linking both the driving biological mechanisms and emerging treatment options for TNBC. CONCLUSIONS Shifting the focus of treatment to identify crucial tumor cell subpopulations and associated biomarkers, such as local immune cell populations and cancer stem cells, could potentially solve or simplify decades' worth of problems that are associated with TNBC, bolstering early detection and the evolution of precision medicine and treatment. The techniques that can be used here are epigenetic analysis and RNA sequencing. Biomarkers, such as PD-L1, survivin, and ABC transporters, are implicated in several crucial processes that maintain tumors, such as cell proliferation, metastasis, immunosuppression, and stemness. Complex treatment options such as, immunotherapy, pathway inhibition, PARP inhibition, virotherapy, and RNA targeting have been considered for TNBC. Phytochemicals are also being considered as a treatment modality for TNBC, as a supplement to chemotherapy and radiation therapy, or as sole treatment.
Collapse
Affiliation(s)
- Rachana Raman
- Photoceutics and Regeneration Laboratory, Department of Biotechnology, Centre for Microfluidics, Biomarkers, Photoceutics and Sensors (μBioPS), Manipal Institute of TechnologyManipal Academy of Higher EducationManipalKarnatakaIndia
- Innotech Manipal, Manipal Institute of TechnologyManipal Academy of Higher EducationManipalKarnatakaIndia
| | - Shristi Debata
- Department of Biotechnology, Manipal Institute of TechnologyManipal Academy of Higher EducationManipalKarnatakaIndia
| | | | - Praveen Kumar
- Photoceutics and Regeneration Laboratory, Department of Biotechnology, Centre for Microfluidics, Biomarkers, Photoceutics and Sensors (μBioPS), Manipal Institute of TechnologyManipal Academy of Higher EducationManipalKarnatakaIndia
- Innotech Manipal, Manipal Institute of TechnologyManipal Academy of Higher EducationManipalKarnatakaIndia
| |
Collapse
|
15
|
Tahara M, Lim DWT, Keam B, Ma B, Zhang L, Wang C, Guo Y. Management approaches for recurrent or metastatic head and neck squamous cell carcinoma after anti-PD-1/PD-L1 immunotherapy. Cancer Treat Rev 2025; 136:102938. [PMID: 40252510 DOI: 10.1016/j.ctrv.2025.102938] [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: 02/20/2025] [Revised: 04/03/2025] [Accepted: 04/07/2025] [Indexed: 04/21/2025]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the seventh most common cancer globally. For patients with recurrent or metastatic (R/M) HNSCC, immunotherapy represents an important advance in clinical practice as an effective and widely used first-line treatment. However, drug resistance following immunotherapy is an emerging problem and, despite the success of immunotherapy in R/M HNSCC, a proportion of patients will become immunotherapy resistant. The mechanisms of immunotherapy resistance are not yet fully understood and subsequent treatment options are limited. Therefore, there is an unmet need for effective and well tolerated treatments for patients who develop immunotherapy-resistant HNSCC. In this review, we address these challenges by summarizing the current definitions of immunotherapy resistance (primary and acquired resistance) as well as knowledge of the mechanisms of resistance to immunotherapy in R/M HNSCC. We then review available clinical data on treatment strategies, including rechallenge with immunotherapy, chemotherapy ± cetuximab, other targeted treatments, antibody-drug conjugates, and bispecific antibodies. We also investigate future research directions by reviewing ongoing clinical trials. Our review shows that the optimal therapeutic strategy for patients with R/M HNSCC remains unclear. While many therapies have reported promising preliminary results, prospective clinical trials are required to support their adoption in clinical practice. In particular, it appears that immunotherapy and antibody-drug conjugates have high potential in this setting. Our review also highlights the importance of further investigation of the mechanisms underlying immunotherapy-resistant R/M HNSCC, to inform selection of optimal therapeutic strategies on an individual patient basis and improve patient outcomes.
Collapse
Affiliation(s)
- Makoto Tahara
- Department of Head and Neck Medical Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Darren Wan-Teck Lim
- Division of Medical Oncology, National Cancer Centre Singapore;Center for Clinician Scientist Development, SingHealth Duke-NUS, Singapore
| | - Bhumsuk Keam
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Brigette Ma
- State Key Laboratory of Translational Oncology, Sir YK Pao Cancer Center; Department of Clinical Oncology, The Chinese University of Hong Kong, Hong Kong, China
| | - Li Zhang
- Value & Implementation, Global Medical & Scientific Affairs, MSD China, Shanghai, China
| | - Chaojun Wang
- Value & Implementation, Global Medical & Scientific Affairs, MSD China, Shanghai, China
| | - Ye Guo
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
16
|
Sonwane S, Telrandhe U, Chambhare N, Vaidya S. Unraveling exosome-mediated cancer therapy resistance: pathways and therapeutic challenges. J Egypt Natl Canc Inst 2025; 37:30. [PMID: 40310494 DOI: 10.1186/s43046-025-00289-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 04/14/2025] [Indexed: 05/02/2025] Open
Abstract
Extracellular vesicles (EVs) have emerged as key cell-to-cell communication mediators and play significant roles in both physiological and pathological processes. In EVs, exosomes represent a distinct subpopulation of EVs that have been found to be involved in cancer initiation and therapeutic resistance. Exosomes transfer a diverse spectrum of molecular cargos that have significant effects on the tumor microenvironment (TME), thereby enabling cancer initiation, metastasis, and therapeutic resistance. Exosomes have recently been of interest in cancer therapy due to their role as important mediators of treatment resistance. The exosomal molecular content-proteins, miRNAs, and lncRNAs-allows exosomes to perform functions including drug efflux and detoxification, cell death pathway modulation, induction of epithelial-to-mesenchymal transition (EMT), and suppression of the immune system. In addition to facilitating immune and stromal cell interactions, exosomes cause extracellular matrix remodeling and induce tumor heterogeneity, making it more difficult to respond to therapy. This review covers intricate roles of exosomes in cancer therapy resistance with regard to their biogenesis, molecular content, and functional impact in the TME. Along with this, we also discuss new therapeutic strategies to overcome exosome-mediated resistance including utilizing exosome inhibitors, designed exosome therapy, and combination with conventional therapies. While exosomes hold promise in prediction and diagnosis through their biomarker function, their heterogeneous origins and cryptic functions make it difficult to target interventions. This review emphasizes that research on exosome-mediated pathways is urgently required to develop new therapeutic strategies that can improve cancer treatment outcomes.
Collapse
Affiliation(s)
- Sandip Sonwane
- Datta Meghe College of Pharmacy, DMIHER (DU), Sawangi, Wardha, Wardha, India.
| | - Umesh Telrandhe
- Datta Meghe College of Pharmacy, DMIHER (DU), Sawangi, Wardha, Wardha, India
| | - Nikhita Chambhare
- Datta Meghe College of Pharmacy, DMIHER (DU), Sawangi, Wardha, Wardha, India
| | - Sunita Vaidya
- Datta Meghe College of Pharmacy, DMIHER (DU), Sawangi, Wardha, Wardha, India
| |
Collapse
|
17
|
Negahban H, Heidari N, Heidari A, Ghane Y, Shirkhoda M, Jalaeefar A. Evaluation of Treatment Response and Survival Outcomes in Anaplastic Thyroid Cancer Patients Following Surgery With and Without Other Treatment Modalities: A Systematic Review. Health Sci Rep 2025; 8:e70710. [PMID: 40309640 PMCID: PMC12042218 DOI: 10.1002/hsr2.70710] [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: 02/10/2024] [Revised: 03/28/2025] [Accepted: 04/02/2025] [Indexed: 05/02/2025] Open
Abstract
Background and Aims Anaplastic thyroid carcinoma (ATC) is a rare type of malignancy ranking among the most aggressive diseases globally, with an extremely poor prognosis. No optimal standardized treatment has been established yet to promote ATC's prognosis and increase the patients' median survival. We aim to assess the effectiveness of surgery alone or combined with other treatment approaches for ATC patients. Methods PubMed, Web of Science, and Scopus databases were systematically searched until June 1st, 2023. Study selection was limited to English retrospective studies. A citation search was also performed for the final articles that were included. This study followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines for systematic reviews and meta-analyses. Results During our search, we came to include 56 articles containing 16,246 patients suffering from ATC. We assessed the overall survival by treatment method and stage, emphasizing surgery's role. The most common efficacious treatment option in patients with resectable cancer is a combination of surgery with adjuvant chemoradiotherapy. However, surgery for stage IVC patients is controversial. Additionally, surgery and multimodality treatment can be affected by patients' characteristics, such as tumor size. Conclusions Stage IVA and IVB resectable cancers may benefit from the combination of surgery and adjuvant therapies. However, the effectiveness of invasive treatments and the selection of appropriate adjuvant therapy options for IVC-stage patients are still controversial.
Collapse
Affiliation(s)
- Hossein Negahban
- Cancer Research Center of Cancer instituteTehran University of Medical SciencesTehranIran
| | - Nazila Heidari
- School of MedicineIran University of Medical SciencesTehranIran
- Research Center for Advanced Technologies in Cardiovascular Medicine, Cardiovascular Diseases, Research InstituteTehran University of Medical SciencesTehranIran
| | - Amirhossein Heidari
- Research Center for Advanced Technologies in Cardiovascular Medicine, Cardiovascular Diseases, Research InstituteTehran University of Medical SciencesTehranIran
- Faculty of Medicine, Tehran Medical SciencesIslamic Azad UniversityTehranIran
| | - Yekta Ghane
- Research Center for Advanced Technologies in Cardiovascular Medicine, Cardiovascular Diseases, Research InstituteTehran University of Medical SciencesTehranIran
- School of MedicineTehran University of Medical SciencesTehranIran
| | - Mohammad Shirkhoda
- Department of Surgical Oncology, Cancer InstituteTehran University of Medical SciencesTehranIran
| | - Amirmohsen Jalaeefar
- Department of Surgical Oncology, Cancer InstituteTehran University of Medical SciencesTehranIran
| |
Collapse
|
18
|
Lobo-Martins S, Arecco L, Cabral TP, Agostinetto E, Dauccia C, Franzoi MA, Del Mastro L, Lambertini M, Piccart M, de Azambuja E. Extended adjuvant endocrine therapy in early breast cancer: finding the individual balance. ESMO Open 2025; 10:105057. [PMID: 40279882 DOI: 10.1016/j.esmoop.2025.105057] [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: 11/23/2024] [Revised: 02/13/2025] [Accepted: 03/20/2025] [Indexed: 04/29/2025] Open
Abstract
Endocrine therapy (ET) is a cornerstone in the management of patients with hormone receptor-positive early breast cancer, which accounts for over 70% of cases worldwide. The efficacy of adjuvant ET for 5 years in reducing the risk of recurrence and improving survival outcomes is well documented. However, the risk for late relapses, occurring >5 years after initial treatment, has prompted exploration of longer treatment durations. Extending ET beyond the traditional 5-year period offers additional benefit in reducing the risk of recurrence and improving long-term outcomes. Nevertheless, determining the optimal duration and identifying suitable candidates for extended therapy is often nuanced. This review aims to comprehensively evaluate the current landscape of extended ET in breast cancer management. It provides an overview of the rationale behind extending endocrine treatment in both premenopausal and postmenopausal women, with a focus on clinical trials and observational studies supporting extended therapy. Furthermore, it emphasizes the significance of considering associated toxicities in patient management. It also explores novel strategies involving the combination of ET with new drugs, leading to an evolution of treatment paradigms that may make the need for extended therapy obsolete.
Collapse
Affiliation(s)
- S Lobo-Martins
- Université libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (H.U.B.), Institut Jules Bordet, Academic Trials Promoting Team, Brussels, Belgium.
| | - L Arecco
- Université libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (H.U.B.), Institut Jules Bordet, Academic Trials Promoting Team, Brussels, Belgium; Department of Internal Medicine and Medical Sciences (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - T P Cabral
- Medical Oncology Department, Hospital de São Francisco Xavier, Unidade Local de Saúde Lisboa Ocidental, Lisbon, Portugal
| | - E Agostinetto
- Université libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (H.U.B.), Institut Jules Bordet, Academic Trials Promoting Team, Brussels, Belgium
| | - C Dauccia
- Université libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (H.U.B.), Institut Jules Bordet, Academic Trials Promoting Team, Brussels, Belgium; Department of Medical Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; University of Pavia, Pavia, Italy
| | - M A Franzoi
- Cancer Survivorship Group, Inserm Unit 981, Gustave Roussy, Villejuif, France
| | - L Del Mastro
- Department of Internal Medicine and Medical Sciences (DiMI), School of Medicine, University of Genova, Genova, Italy; Medical Oncology Department, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - M Lambertini
- Department of Internal Medicine and Medical Sciences (DiMI), School of Medicine, University of Genova, Genova, Italy; Medical Oncology Department, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - M Piccart
- Université libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (H.U.B.), Institut Jules Bordet, Brussels, Belgium
| | - E de Azambuja
- Université libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (H.U.B.), Institut Jules Bordet, Academic Trials Promoting Team, Brussels, Belgium; Université libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (H.U.B.), Institut Jules Bordet, Brussels, Belgium.
| |
Collapse
|
19
|
Abdo N, Thom B, Chang C, Feldman M, Benedict C, Semler R, Kelvin J, Gemignani M, Goldfarb S. Decisions of young women with breast cancer regarding fertility preservation before cancer treatment and family building after treatment. J Cancer Surviv 2025:10.1007/s11764-025-01809-1. [PMID: 40266447 DOI: 10.1007/s11764-025-01809-1] [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: 11/20/2024] [Accepted: 04/07/2025] [Indexed: 04/24/2025]
Abstract
PURPOSE Approximately 19% of breast cancer cases in 2020 were diagnosed in women under age 50. These premenopausal women with early-stage breast cancer face unique challenges related to family building. This study examines the decision-making and outcomes of patients who pursue fertility preservation before treatment and family building afterwards. METHODS Breast cancer survivors seen by a fertility nurse specialist before initiating cancer treatment at Memorial Sloan Kettering Cancer Center (MSKCC) between 2009 and 2017 were contacted to participate. Participants completed a web-based cross-sectional survey examining decisions made about fertility preservation, factors influencing them, and post-treatment reflections and outcomes. RESULTS Prior to treatment, 168 women met with a reproductive endocrinologist and 116 underwent fertility preservation. After completing cancer treatment, 36 women had children and 15 of these women used eggs/embryos frozen prior to treatment. The majority of women deemed these factors "very important" when deciding whether to freeze eggs/embryos: a desire to have a biologic child, the ability to feel hopeful about their future, the chance of success, feeling overwhelmed by their diagnosis, and concerns about having regret if they didn't freeze their eggs/embryo. CONCLUSIONS Over half of patients who attempted family building after treatment were able to have children. Many of these women used eggs/embryos frozen before treatment either by getting pregnant themselves or via surrogacy. IMPLICATIONS FOR CANCER SURVIVORS This data stresses the importance of referring cancer patients early to a reproductive endocrinologist before initiation of systemic therapy and has high translational promise in the clinical care of all young female cancer survivors interested in fertility preservation.
Collapse
Affiliation(s)
| | - Bridgette Thom
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
| | | | | | | | | | - Joanne Kelvin
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Shari Goldfarb
- Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Weill Cornell Medical Center, New York, NY, USA.
| |
Collapse
|
20
|
Bai J, Gao Y, Zhang G. The treatment of breast cancer in the era of precision medicine. Cancer Biol Med 2025; 22:j.issn.2095-3941.2024.0510. [PMID: 40269562 PMCID: PMC12032834 DOI: 10.20892/j.issn.2095-3941.2024.0510] [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] [Accepted: 03/05/2025] [Indexed: 04/25/2025] Open
Abstract
The management of breast cancer, one of the most common and heterogeneous malignancies, has transformed with the advent of precision medicine. This review explores current developments in genetic profiling, molecular diagnostics, and targeted therapies that have revolutionized breast cancer treatment. Key innovations, such as cyclin-dependent kinases 4/6 (CDK4/6) inhibitors, antibody-drug conjugates (ADCs), and immune checkpoint inhibitors (ICIs), have improved outcomes for hormone receptor-positive (HR+), HER2-positive (HER2+), and triple-negative breast cancer (TNBC) subtypes remarkably. Additionally, emerging treatments, such as PI3K inhibitors, poly (ADP-ribose) polymerase (PARP) inhibitors, and mRNA-based therapies, offer new avenues for targeting specific genetic mutations and improving treatment response, particularly in difficult-to-treat breast cancer subtypes. The integration of liquid biopsy technologies provides a non-invasive approach for real-time monitoring of tumor evolution and treatment response, thus enabling dynamic adjustments to therapy. Molecular imaging and artificial intelligence (AI) are increasingly crucial in enhancing diagnostic precision, personalizing treatment plans, and predicting therapeutic outcomes. As precision medicine continues to evolve, it has the potential to significantly improve survival rates, decrease recurrence, and enhance quality of life for patients with breast cancer. By combining cutting-edge diagnostics, personalized therapies, and emerging treatments, precision medicine can transform breast cancer care by offering more effective, individualized, and less invasive treatment options.
Collapse
Affiliation(s)
- Jingwen Bai
- The Breast Center of Yunnan Cancer Hospital & The Third Affiliated Hospital of Kunming Medical University & Peking University Cancer Hospital Yunnan, Kunming 650118, China
| | - Yiyang Gao
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, School of Medicine, Xiamen University, Xiamen 361100, China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361100, China
| | - Guojun Zhang
- The Breast Center of Yunnan Cancer Hospital & The Third Affiliated Hospital of Kunming Medical University & Peking University Cancer Hospital Yunnan, Kunming 650118, China
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, School of Medicine, Xiamen University, Xiamen 361100, China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361100, China
| |
Collapse
|
21
|
Tauber N, Amann N, Dannehl D, Deutsch TM, Dimpfl M, Fasching P, Hartkopf A, Heublein S, Hilmer L, Hörner M, Krawczyk N, Krückel A, Krug D, Marmé F, Michel LL, Reinisch M, Rody A, Schäffler H, Schneeweiss A, Utz D, Veselinovic K, Banys-Paluchowski M. Therapy of early breast cancer: current status and perspectives. Arch Gynecol Obstet 2025:10.1007/s00404-025-08028-0. [PMID: 40261372 DOI: 10.1007/s00404-025-08028-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 04/01/2025] [Indexed: 04/24/2025]
Abstract
Medical advancements in breast cancer are truly remarkable. Especially in recent years, numerous new therapeutics have been approved and surgical strategies have been de-escalated for specific patient groups. In the therapeutic setting, CDK4/6 inhibitors as oral maintenance therapy in early breast cancer and immune checkpoint inhibitors (Pembrolizumab) for triple-negative breast cancer (BC) are noteworthy. In the surgical field, prospective randomized controlled trials have currently explored the possibility to deescalate axillary surgery by omitting sentinel lymph node excision (INSEMA, SOUND). As a result, there have been significant improvements in prognosis and a reduction in surgical morbidity for patients. Many exciting trials are underway, and it remains to be seen whether antibody-drug conjugates beyond trastuzumab emtansine, will find their way into the treatment lines for early-stage BC. Furthermore, the integration of artificial intelligence in both diagnostics and treatment recommendation evaluation is a promising area with great potential.
Collapse
Affiliation(s)
- Nikolas Tauber
- Department of Obstetrics and Gynecology, University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany.
| | - Niklas Amann
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich Alexander University of Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Dominik Dannehl
- Department of Obstetrics and Gynecology, University Hospital Tuebingen, 72016, Tuebingen, Germany
| | - Thomas M Deutsch
- Department of Obstetrics and Gynecology, University Hospital Heidelberg, Heidelberg, Germany
| | - Moritz Dimpfl
- Department of Obstetrics and Gynecology, Medical Faculty Mannheim, University Medical Center Mannheim, University of Heidelberg, Mannheim, Deutschland
| | - Peter Fasching
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich Alexander University of Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Andreas Hartkopf
- Department of Obstetrics and Gynecology, University Hospital Tuebingen, 72016, Tuebingen, Germany
| | - Sabine Heublein
- Department of Obstetrics and Gynecology, University Hospital Ulm, 89075, Ulm, Germany
| | - Lisbeth Hilmer
- Department of Obstetrics and Gynecology, University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Manuel Hörner
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich Alexander University of Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Natalia Krawczyk
- Department of Obstetrics and Gynecology, University Hospital Duesseldorf, 40225, Duesseldorf, Germany
| | - Annika Krückel
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich Alexander University of Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - David Krug
- Department of Radiotherapy and Radiation Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Frederik Marmé
- Department of Obstetrics and Gynecology, Medical Faculty Mannheim, University Medical Center Mannheim, University of Heidelberg, Mannheim, Deutschland
| | - Laura L Michel
- National Center for Tumor Diseases, University Hospital and German Cancer Research Center Heidelberg, 69120, Heidelberg, Germany
| | - Mattea Reinisch
- Department of Obstetrics and Gynecology, Medical Faculty Mannheim, University Medical Center Mannheim, University of Heidelberg, Mannheim, Deutschland
| | - Achim Rody
- Department of Obstetrics and Gynecology, University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Henning Schäffler
- Department of Obstetrics and Gynecology, University Hospital Ulm, 89075, Ulm, Germany
| | - Andreas Schneeweiss
- National Center for Tumor Diseases, University Hospital and German Cancer Research Center Heidelberg, 69120, Heidelberg, Germany
- German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - David Utz
- Department of Internal Medicine VIII, Medical Oncology and Pneumology, University Hospital Tuebingen, 72016, Tuebingen, Germany
| | - Kristina Veselinovic
- Department of Obstetrics and Gynecology, University Hospital Ulm, 89075, Ulm, Germany
| | - Maggie Banys-Paluchowski
- Department of Obstetrics and Gynecology, University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| |
Collapse
|
22
|
Sahin G, Bakirci IT, Sozen I, Kilic SO, Afsar S, Kocadal NC, Geyikoglu I. Adenomyosis as a prognostic factor in ovarian cancer: a retrospective study. Arch Gynecol Obstet 2025:10.1007/s00404-025-08023-5. [PMID: 40240534 DOI: 10.1007/s00404-025-08023-5] [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: 12/16/2024] [Accepted: 03/25/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND Adenomyosis is a gynecological condition that frequently coexists with gynecological malignancies and has been shown to influence disease outcomes. However, its impact on ovarian cancer prognosis remains unclear. This study aimed to investigate the relationship between adenomyosis and clinicopathological and prognostic features in ovarian cancer patients. METHODS We retrospectively analyzed 226 patients with ovarian cancer who underwent surgery between 2020 and 2023. The patients were divided into two groups based on the presence (n = 114) or absence (n = 112) of adenomyosis, confirmed by histopathological examination. Clinicopathological characteristics, including histological subtypes, disease-free survival (DFS), and overall survival (OS) were compared between the groups with a median follow-up of 36 months. RESULTS Patients with adenomyosis demonstrated more favorable characteristics, including early stage disease (54.3% vs 39.2%, p = 0.048), lower-grade tumors (55.2% vs 31.2%, p = 0.049), and smaller tumor sizes (39.4% vs 26.7%, p = 0.043). Adenomyosis was significantly associated with endometrioid subtype (OR = 2.89, p = 0.043) and negatively associated with serous carcinoma (OR = 0.39, p = 0.034). Three-year DFS was significantly better in the adenomyosis group (79.2% vs 73.9%, p = 0.01), particularly in high-grade tumors (80% vs 58%, p < 0.05). No significant difference was observed in overall OS (73.3% vs 73.1%, p = 0.14), although high-grade tumors with adenomyosis showed improved OS (71% vs 57%, p < 0.05). CONCLUSION The presence of adenomyosis in patients with ovarian cancer was associated with favorable clinicopathological features, particularly endometrioid histology and low-grade tumors, and improved survival in high-grade tumors. These findings suggest a potential biological interaction between adenomyosis and ovarian cancer that warrants further investigation for personalized treatment approaches.
Collapse
Affiliation(s)
- Gozde Sahin
- Division of Gynaecologic Oncology, Department of Obstetrics and Gynecology, Basaksehir Cam and Sakura City Hospital, Istanbul, Turkey
| | - Isil Turan Bakirci
- Division of Perinatology, Department of Obstetrics and Gynecology, Basaksehir Cam and Sakura City Hospital, Istanbul, Turkey.
| | - Isik Sozen
- Division of Gynaecologic Oncology, Department of Obstetrics and Gynecology, Basaksehir Cam and Sakura City Hospital, Istanbul, Turkey
| | - Sinem Ozsahin Kilic
- Division of Gynaecologic Oncology, Department of Obstetrics and Gynecology, Basaksehir Cam and Sakura City Hospital, Istanbul, Turkey
| | - Selim Afsar
- Division of Gynaecologic Oncology, Department of Obstetrics and Gynecology, Balikesir University, School of Medicine, Balikesir, Turkey
| | - Nilufer Cetinkaya Kocadal
- Division of Gynaecologic Oncology, Department of Obstetrics and Gynecology, Basaksehir Cam and Sakura City Hospital, Istanbul, Turkey
| | - Ipek Geyikoglu
- Department of Obstetrics and Gynecology, Basaksehir Cam and Sakura City Hospital, Istanbul, Turkey
| |
Collapse
|
23
|
Li X, Lu X, Liu M, Chen J, Lu X. Extracellular vesicles: messengers of cross-talk between gastric cancer cells and the tumor microenvironment. Front Cell Dev Biol 2025; 13:1561856. [PMID: 40309240 PMCID: PMC12040901 DOI: 10.3389/fcell.2025.1561856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
Gastric cancer is a common malignancy characterized by an insidious onset and high mortality rate. Exosomes, a special type of extracellular vesicle, contain various bioactive molecules and have been found to play crucial roles in maintaining normal physiological functions and homeostasis in the body. Recent research has shown that the contents of exosome play a significant role in the progression and metastasis of gastric cancer through communication and regulatory functions. These mechanisms involve promoting gastric cancer cell proliferation and drug resistance. Additionally, other cells in the gastric cancer microenvironment can regulate the progression of gastric cancer through exosomes. These include exosomes derived from fibroblasts and immune cells, which modulate gastric cancer cells. Therefore, in this review, we provide a brief overview of recent advances in the contents and occurrence mechanisms of exosome. This review specifically focused on the regulatory mechanisms of exosomes derived from gastric cancer and other cellular subtypes in the tumor microenvironment. Subsequently, we summarize the latest research progress on the use of exosomes in liquid biopsy, discussing the potential of gastric cancer exosomes in clinical applications.
Collapse
Affiliation(s)
- Xiwen Li
- Kunshan Hospital of Chinese Medicine, Affiliated Hospital of Yangzhou University, Kunshan, China
| | - Xian Lu
- Kunshan Hospital of Chinese Medicine, Affiliated Hospital of Yangzhou University, Kunshan, China
| | - Mi Liu
- Kunshan Hospital of Chinese Medicine, Affiliated Hospital of Yangzhou University, Kunshan, China
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou, China
| | - Junjie Chen
- Department of Clinical Medical Research Center, Affiliated Hospital of Nantong University, Nantong, China
| | - Xirong Lu
- Kunshan Hospital of Chinese Medicine, Affiliated Hospital of Yangzhou University, Kunshan, China
| |
Collapse
|
24
|
Yousaf H, Javed A, Sultana N, Farouk K, Usman M, Khan GR, Gomez MK, Asghar S, Justin S. Unlocking the secrets: Exploring the connection between HPV and bladder cancer in Pakistan. Urol Oncol 2025:S1078-1439(25)00118-8. [PMID: 40240277 DOI: 10.1016/j.urolonc.2025.03.014] [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: 11/16/2024] [Revised: 02/06/2025] [Accepted: 03/14/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND Human Papillomavirus' (HPV) clear association with anogenital carcinomas raises concerns about its role in urologic carcinomas due to anatomical proximity. This etiological link is under-researched in Pakistan, creating a critical gap in the literature. OBJECTIVES To decipher any probable pathological association of high-risk HPV genotypes in bladder cancer etiology. METHODS Bladder biopsies from 63 bladder cancer patients, confirmed by H&E and IHC staining were collected with histopathological and clinical data, after informed consent. DNA was extracted and processed for HPV detection using L1 consensus primers. HPV genotyping was performed using E6-E7 specific primers for HPV16 and 18, via conventional PCR. RESULT About 15.9% (10/63) of bladder cancer cases were HPV-positive. 30% (3/10) of which were HPV16-positive, 60% (6/10) were HPV18-positive, while the remaining 1 sample (1/10) was neither positive for HPV16 nor 18 indicating a probable involvement of other high-risk genotypes. Histologically, most of the HPV-positive bladder cancers were low-grade invasive urothelial carcinoma, having a male-to-female incident ratio of 9:1 with an average age of 65 years. No significant association was found between HPV prevalence and tobacco consumption, physical activity, co-morbidities (diabetes and hypertension), and treatment effectiveness. CONCLUSION The study provides the latest epidemiological data on the prevalence and genotype distribution of HPV in bladder cancer in Pakistan. Although the frequency of HPV was not so high, nonetheless an association with bladder cancer incidence was identified as an important risk factor. Consequently, HPV remains a serious issue in low- and middle-income countries (LMICs), significantly contributing to otherwise preventable cancers.
Collapse
Affiliation(s)
- Hafsa Yousaf
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12 Sector, Islamabad - 44000, Pakistan
| | - Aneela Javed
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12 Sector, Islamabad - 44000, Pakistan
| | - Nuzhat Sultana
- Department of Pathology, Northwest School of Medicine, Peshawar, Pakistan
| | - Khalid Farouk
- Urology and Renal Transplantation, Foundation University Medical College/ Fauji Foundation Hospital, Rawalpindi, Pakistan
| | - Muhammad Usman
- Department of Virology, National Institutes of Health (NIH), Islamabad, Pakistan
| | - Gul Rehman Khan
- Urology and Renal Transplantation, Foundation University Medical College/ Fauji Foundation Hospital, Rawalpindi, Pakistan
| | - Miriam Kathleen Gomez
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12 Sector, Islamabad - 44000, Pakistan
| | - Sobia Asghar
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12 Sector, Islamabad - 44000, Pakistan
| | - Saira Justin
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12 Sector, Islamabad - 44000, Pakistan.
| |
Collapse
|
25
|
Gong YXY, Huang XF, Liang J, Xie J, Qing LS. Aptamer-based microfluidics for the detection of cancer biomarkers. Anal Bioanal Chem 2025:10.1007/s00216-025-05863-7. [PMID: 40227354 DOI: 10.1007/s00216-025-05863-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/18/2025] [Accepted: 03/25/2025] [Indexed: 04/15/2025]
Abstract
Early diagnosis of cancer is a major concern in clinical medicine. Recently, aptamer-based microfluidics have offered promising platforms for the sensitive detection of cancer biomarkers. This review summarizes the application of aptamer sensors in the microfluidic platform for cancer biomarker analysis. The materials for microfluidic aptamer biosensor fabrication, unique design based on microposts, materials for enhanced detection capability, and the application principle of combining with other detection methods are introduced in detail, so as to demonstrate its development potential in cancer diagnosis and personalized therapy. Finally, the challenges and opportunities for developing miniaturized diagnostic platforms are discussed.
Collapse
Affiliation(s)
- Yi-Xin-Yue Gong
- School of Pharmacy, Chengdu Medical College, Chengdu, 610500, China
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610041, China
| | - Xiao-Feng Huang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610041, China
| | - Jian Liang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610041, China
| | - Jing Xie
- School of Pharmacy, Chengdu Medical College, Chengdu, 610500, China.
| | - Lin-Sen Qing
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610041, China.
| |
Collapse
|
26
|
Chen M, Chen J, Wang X, Long T. Serum Klotho levels and epilepsy among U.S. adults in the NHANES 2013-2016: a cross-sectional study. Sci Rep 2025; 15:12218. [PMID: 40211052 PMCID: PMC11986094 DOI: 10.1038/s41598-025-97112-1] [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: 09/26/2024] [Accepted: 04/02/2025] [Indexed: 04/12/2025] Open
Abstract
The relationship between Klotho and epilepsy is poorly understood. This study aimed to investigate the association between serum Klotho levels and epilepsy. A total of 99,966 individuals who participated in the NHANES from the 2013 to 2016 were initially included. After excluding participants with missing data on serum Klotho concentration (57,286), epilepsy (6) and relevant covariates (6,096), the final sample consisted of 36,578 participants, with a mean age of 59 years. Weighted multivariable logistic regression analyses showed that high serum Klotho levels were negatively associated with the prevalence of epilepsy, with an odds ratio (OR) of 0.74 (95% confidence interval [CI]: 0.64 to 0.86; P < 0.001. The Restricted Cubic Spline analyses (RCS) model showed a non-linear relationship between the serum Klotho levels and epilepsy. Subgroup analysis showed that serum Klotho levels were negatively correlated with epilepsy prevalence in individuals under 65 years of age and in males. Our study suggests that serum Klotho levels were associated with the prevalence of epilepsy. Further large-scale prospective studies and randomized trials are warranted to confirm our findings.
Collapse
Affiliation(s)
- Minghong Chen
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Junyu Chen
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xuerui Wang
- Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Tianyi Long
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
27
|
Huang C, Qiu Z, Huang H, Xiao X, Du F, Ji J, Xu X, Jiang X, Wang Y, Gao C. Alterations in genomic features and the tumour immune microenvironment predict immunotherapy outcomes in advanced biliary tract cancer patients. Br J Cancer 2025:10.1038/s41416-025-03011-7. [PMID: 40211026 DOI: 10.1038/s41416-025-03011-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 03/19/2025] [Accepted: 03/31/2025] [Indexed: 04/12/2025] Open
Abstract
BACKGROUND The response to immunotherapy is limited in advanced biliary tract cancer (BTC). Response prediction is a serious challenge in the clinic. METHODS This study included 60 patients with advanced BTC who received anti-PD-1 treatment. Among these patients, 30 were subjected to 520 gene panel sequencing, and 50 were subjected to multiplex circulating cytokine testing. The entropy and mutation features were analysed via the optimized pipeline based on our previous work. The repeated LASSO algorithm was used to identify the optimal features. The associations between sequence features and cell communications were explored by analysing single-cell transcriptome data from BTC (GSE125449). Cox regression was used to develop the integrated model. Time-dependent C-index, Kaplan‒Meier, and receiver operating characteristic (ROC) curves were used to assess the prediction performance. RESULTS TP53, NRAS, FBXW7, and APC were identified as prognosis-related genes. The average C-indices of sequence entropy (0.819) and mutation (0.817) for overall survival (OS) were significantly greater than those of tumour mutation burden (TMB, 0.392) and mutation score (0.638). Single-cell transcriptome data revealed that TP53, KRAS, and NRAS were enriched in plasmacytoid dendritic cells (pDCs) and that the communication between pDCs and macrophages was mediated through the CXCL signalling pathway. The integrated model (EM-CXCL10) showed powerful predictive performance for survival status (AUC: 0.863, 95% CI: 0.643-0.972) and objective response rate (AUC: 0.990, 95% CI: 0.822-1.000). CONCLUSIONS This study constructed a multidimensional strategy that might indicate the prognosis of BTC immunotherapy, enabling the recognition of BTC patients who would benefit from immunotherapy, thereby guiding personalized clinical decision-making.
Collapse
Affiliation(s)
- Chenjun Huang
- Department of Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Zhiquan Qiu
- Department of Biliary Tract Surgery I, Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China
| | - Honglian Huang
- Department of Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
- School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Xiao Xiao
- Department of Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Fei Du
- Department of Laboratory Medicine, Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China
| | - Jun Ji
- Department of Laboratory Medicine, Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China
| | - Xuewen Xu
- Department of Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Xiaoqing Jiang
- Department of Biliary Tract Surgery I, Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China
| | - Ying Wang
- Department of Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China.
- Department of Laboratory Medicine, Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Chunfang Gao
- Department of Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China.
- Department of Laboratory Medicine, Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| |
Collapse
|
28
|
S BR, Dhar R, Devi A. Exosomes-mediated CRISPR/Cas delivery: A cutting-edge frontier in cancer gene therapy. Gene 2025; 944:149296. [PMID: 39884405 DOI: 10.1016/j.gene.2025.149296] [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: 12/10/2024] [Revised: 01/09/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
Cancer is considered the second most common disease globally. In the past few decades, many approaches have been proposed for cancer treatment. One among those is targeted therapy using CRISPR/Cas system which plays a significant role in translational research through gene editing. However, due to its inability to cope with specific targeting, off-target effects, and limited tumor penetration, it is very challenging to use this approach in cancer studies. To increase its efficacy, CRISPR components are engineered into the extracellular vesicles (EVs), especially exosomes (a subpopulation of EVs). Exosomes have a significant role in cellular communication. Exosomes-based CRISPR/Cas system transport for gene editing enhances specificity, reduces off-target effects, and improves the therapeutic potential. This review highlights the role of exosomes and the CRISPR/Cas system in cancer research, exosomes-based CRISPR delivery for cancer treatment, and its future orientation.
Collapse
Affiliation(s)
- Bhavanisha Rithiga S
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, TamilNadu 603203, India
| | - Rajib Dhar
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, TamilNadu 603203, India
| | - Arikketh Devi
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, TamilNadu 603203, India.
| |
Collapse
|
29
|
Liang Z, Huang X, Mao J, Xie J, Li X, Qin L. The impact of KRAS mutations on risk of venous thromboembolism recurrence in patients with metastatic colorectal cancer. BMC Gastroenterol 2025; 25:240. [PMID: 40211193 PMCID: PMC11987216 DOI: 10.1186/s12876-025-03843-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 04/02/2025] [Indexed: 04/12/2025] Open
Abstract
BACKGROUND The relationship between KRAS mutations and the risk of venous thromboembolism (VTE) recurrence in metastatic colorectal cancer (mCRC) patients with established cancer-associated thrombosis (CAT) remains uncertain. This study aims to (1) evaluate the predictive value of seven KRAS mutation subtypes for VTE recurrence and (2) assess the impact of incorporating these mutations into two existing VTE risk scores: the Khorana score and the Ottawa score. METHODS Between 2019 and 2023, we identified patients with histologically confirmed mCRC who had symptomatic or incidental index VTE and received anticoagulation therapy. Regression analyses were conducted to calculate hazard ratios (HRs) for recurrent VTE associated with the seven KRAS mutation subtypes. We used receiver operating characteristic (ROC) curves to assess the performance of both the original scores and the modified scores that included KRAS mutations. To quantify the improvements of the modified scores, we calculated the net reclassification improvement (NRI). RESULTS A total of 2,195 patients were enrolled. KRAS G12C, KRAS G12A, and KRAS G13D mutations were significantly associated with a higher risk of recurrent VTE compared to other subtypes, with HRs of 1.84 (95% CI: 1.09-2.97), 2.02 (95% CI: 1.07-3.79), and 1.55 (95% CI: 1.02-2.27), respectively. The original Khorana and Ottawa scores demonstrated moderate predictive ability for VTE recurrence, each with an area under the ROC curve (ROC-AUC) of 0.56 (95% CI: 0.52-0.60). Incorporating the KRAS G12C, KRAS G12A, and KRAS G13D mutations improved the AUCs to 0.70 (95% CI: 0.67-0.74) for the modified Khorana score and 0.71 (95% CI: 0.67-0.74) for the modified Ottawa score. After dichotomizing risk using thresholds from ROC analysis, the NRI values were 0.54 (95% CI: 0.43-0.65) for the modified Khorana score and 0.48 (95% CI: 0.37-0.60) for the modified Ottawa score. CONCLUSIONS The KRAS G12C, KRAS G12A, and KRAS G13D mutations are significantly associated with an increased risk of recurrent VTE. Incorporating these specific KRAS mutations into existing risk scores may enhance their predictive accuracy for recurrent VTE in patients with mCRC.
Collapse
Affiliation(s)
- Zhikun Liang
- Department of Pharmacy, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoyan Huang
- Department of Pharmacy, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jieling Mao
- Department of Pharmacy, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, China
| | - Jingwen Xie
- Department of Pharmacy, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoyan Li
- Department of Pharmacy, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
- Department of Pharmacy, The Sixth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, 26 Erheng Road of Yuan Village, Tianhe District, Guangzhou, 510655, China.
| | - Li Qin
- Department of Pharmacy, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
- Department of Pharmacy, The Sixth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, 26 Erheng Road of Yuan Village, Tianhe District, Guangzhou, 510655, China.
| |
Collapse
|
30
|
Liu T, Cui Y, Ouyang Y, Wang M, Yue S. Exosomal CCT3 as a biomarker for diagnosis and immune therapy response in patients diagnosed with hepatocellular carcinoma. Dig Liver Dis 2025:S1590-8658(25)00301-9. [PMID: 40221386 DOI: 10.1016/j.dld.2025.03.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 11/19/2024] [Accepted: 03/21/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the dominant type of liver cancer and is associated with a high mortality rate. However, HCC lacks biomarkers for diagnosis and immune therapy response. Tumor-derived exosomes (TDEs) carcinogen-specific molecules have been used for screening multiple biomarkers. This study aimed to identify new biomarkers for the diagnosis of HCC and response to immune checkpoint blockade (ICB) therapy. METHODS Analysis of differentially expressed genes (DEGs) in HCC and normal tissues was integrated using The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO), and ExoCarta datasets. The expression of CCT3 was validated in samples from patients with HCC using quantitative polymerase chain reaction (qPCR), Western blotting, and immunohistochemistry (IHC) techniques. RESULTS Exosomal CCT3 was identified as a potential biomarker with significant impact. The expression of CCT3 in different tumor stages and normal tissues adjacent to the tumors (NATs) was validated using qPCR, western blotting, and IHC. CCT3 expression significantly increased the number of activated natural killer cells in HCC, as confirmed by qPCR and IHC. CCT3 expression significantly increases the expression of immune checkpoints in HCC. HCC-derived exosomes significantly increase the enrichment of CCT3. CONCLUSION Exosomal CCT3 is a biomarker for diagnosis and ICB therapy of HCC via MYC pathway activation and immune infiltration.
Collapse
Affiliation(s)
- Tiange Liu
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China; Nankai University Affiliated Eye Hospital, Nankai University, Tianjin, China; Tianjin Eye Hospital, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin, China
| | - Yanyan Cui
- The Affiliated Hospital of Chifeng University, Chifeng, Inner Mongolia, China
| | - Yiben Ouyang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Meilin Wang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Shijing Yue
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China.
| |
Collapse
|
31
|
Kwas K, Szubert M, Wilczyński JR. Latest Update on lncRNA in Epithelial Ovarian Cancer-A Scoping Review. Cells 2025; 14:555. [PMID: 40214508 PMCID: PMC11988607 DOI: 10.3390/cells14070555] [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: 02/10/2025] [Revised: 03/22/2025] [Accepted: 03/24/2025] [Indexed: 04/14/2025] Open
Abstract
Long noncoding RNAs (lncRNAs) are RNA molecules exceeding 200 nucleotides that do not encode proteins yet play critical roles in regulating gene expression at multiple levels, such as chromatin modification and transcription. These molecules are significantly engaged in cancer progression, development, metastasis, and chemoresistance. However, the function of lncRNAs in epithelial ovarian cancer (EOC) has not yet been thoroughly studied. EOC remains challenging due to its complex molecular pathogenesis, characterized by genetic and epigenetic alterations. Emerging evidence suggests that lncRNAs, such as XIST, H19, NEAT1, and MALAT1, are involved in EOC by modulating gene expression and signaling pathways, influencing processes like cell proliferation, invasion, migration, and chemoresistance. Despite extensive research, the precise mechanism of acting of lncRNAs in EOC pathogenesis and treatment resistance still needs to be fully understood, highlighting the need for further studies. This review aims to provide an updated overview of the current understanding of lncRNAs in EOC, emphasizing their potential as biomarkers and therapeutic targets. We point out the gaps in the knowledge regarding lncRNAs' influence on epithelial ovarian cancer (EOC), deliberating on new possible research areas.
Collapse
Affiliation(s)
- Katarzyna Kwas
- Department of Surgical and Oncologic Gynaecology, 1st Department of Gynaecology and Obstetrics, Medical University of Lodz, 90-136 Łódź, Poland; (M.S.); (J.R.W.)
| | | | | |
Collapse
|
32
|
Zhang C, Wang H, Li X, Jiang Y, Sun G, Yu H. Enhancing antitumor immunity: the role of immune checkpoint inhibitors, anti-angiogenic therapy, and macrophage reprogramming. Front Oncol 2025; 15:1526407. [PMID: 40260303 PMCID: PMC12009726 DOI: 10.3389/fonc.2025.1526407] [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: 11/11/2024] [Accepted: 03/19/2025] [Indexed: 04/23/2025] Open
Abstract
Cancer treatment has long been hindered by the complexity of the tumor microenvironment (TME) and the mechanisms that tumors employ to evade immune detection. Recently, the combination of immune checkpoint inhibitors (ICIs) and anti-angiogenic therapies has emerged as a promising approach to improve cancer treatment outcomes. This review delves into the role of immunostimulatory molecules and ICIs in enhancing anti-tumor immunity, while also discussing the therapeutic potential of anti-angiogenic strategies in cancer. In particular, we highlight the critical role of endoplasmic reticulum (ER) stress in angiogenesis. Moreover, we explore the potential of macrophage reprogramming to bolster anti-tumor immunity, with a focus on restoring macrophage phagocytic function, modulating hypoxic tumor environments, and targeting cytokines and chemokines that shape immune responses. By examining the underlying mechanisms of combining ICIs with anti-angiogenic therapies, we also review recent clinical trials and discuss the potential of biomarkers to guide and predict treatment efficacy.
Collapse
Affiliation(s)
- Chong Zhang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Xinying Li
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuxin Jiang
- Department of Nephrology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Guoping Sun
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hanqing Yu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
33
|
Long K, Kui X, Zeng Q, Dong W. Cancer cell-derived exosomal miR-34a inhibits the malignant progression of pancreatic adenocarcinoma cells by restraining the M2 polarization of macrophages. Eur J Histochem 2025; 69:4176. [PMID: 40244037 PMCID: PMC12051414 DOI: 10.4081/ejh.2025.4176] [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: 12/24/2024] [Accepted: 04/02/2025] [Indexed: 04/18/2025] Open
Abstract
This study aimed to investigate the crosstalk mechanism between pancreatic cancer (PAC) cells and M2 tumor-associated macrophages induced by tumor-derived exosomal miR-34a. MicroRNA and mRNA expression levels were detected using RT-qPCR. Cell Counting Kit-8, wound-healing, transwell assays and flow cytometry were respectively employed to assess cell proliferation, migration, invasion and apoptosis. Enzyme-linked immunosorbent assay was utilized to determine cytokine secretion. Transmission electron microscopy and nanoparticle tracking analyses were performed to detect the exosome morphology and particle size. Phagocytosis of exosomes by macrophages was verified by PKH26 labeling. The effects of exosome-treated macrophages on the epithelial-mesenchymal transition, invasion, and migration of PANC-1 cells were investigated using coculture experiments. The identification of miR-34a's potential targets were determined with TargetScan and validated by a dual-luciferase reporter assay. miR-34a was expressed at low levels in PAC tissues, cells, and exosomes. The overexpression of miR-34a restrains the malignant progression of PANC-1 cells. After miR-34a-overexpressed PANC-1-derived exosomes were phagocytosed by macrophages, the process of M2 polarization in macrophages was obstructed, leading to the suppression of epithelial-mesenchymal transition, migration, and invasion of the cocultured PANC-1 cells. Suppressor of cytokine signaling 3 is a direct target of miR-34a. MiR-34a negatively modulates the suppressor of cytokine signaling 3 to prevent the M2 polarization of macrophages by engaging the Janus kinase/signal transducers and activators of the transcription pathway and influencing the malignancy of PAC cells. miR-34a in cancer cell-derived exosomes inhibits the malignant progression of pancreatic cancer cells by restraining M2 polarization of macrophages.
Collapse
Affiliation(s)
- Kui Long
- Department of Three Wards of Hepatobiliary Surgery
| | - Xiang Kui
- Department of Pathology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Qingbin Zeng
- Department of Three Wards of Hepatobiliary Surgery
| | - Wenzhi Dong
- Department of Three Wards of Hepatobiliary Surgery
| |
Collapse
|
34
|
Ying Z, Linxun L, Kechang Z, Xiaowu W, Huazhen G, Zhijun M. Optimal extension time after initial endocrine therapy for postmenopausal hormone receptor-positive early-stage breast cancer: a systematic review and meta-analysis. BMC Womens Health 2025; 25:156. [PMID: 40181354 PMCID: PMC11969833 DOI: 10.1186/s12905-025-03610-9] [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: 12/19/2023] [Accepted: 02/16/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND The optimal duration of extended endocrine therapy (ET) for women with hormone receptor-positive (HR-positive) early-stage postmenopausal breast cancer remains uncertain. This meta-analysis systematically evaluated the optimal time to prolong aromatase inhibitors ( AIs) therapy for postmenopausal early stage breast cancer who received initial endocrine therapy. METHODS PubMed, Web of Science, Ovid, Scopus, EmBase, and Cochrane Library were searched for randomized controlled trials (RCTs) using keywords related to breast cancer, HR-positive, AIs, and tamoxifen (TAM). Disease-free survival (DFS) was used as the primary endpoint. Meta-analysis was performed using STATA 16.0 and Revman 5.4 statistical software. Hazard ratio (HR) with its corresponding 95% confidence intervals (CI) was used as an effective indicator to assess DFS, OS, and subgroups of extended ET. Relative ratio (RR) was used to assess adverse events. RESULTS The study included four RCTs involving 8,748 patients with HR-positive breast cancer. Pooled data showed an improvement in DFS when extending endocrine therapy from 5 to 7-8 years (HR = 0.82, 95% CI: 0.73 ~ 0.93), especially in patients with tumor size ≥ 2 cm (HR = 0.69, 95% CI: 0.49 ~ 0.98), estrogen receptor (ER) and progesterone receptor (PR) positive (HR = 0.77, 95% CI: 0.67 ~ 0.89), human epidermal growth factor receptor 2 (HER-2) positive or negative (HR = 0.85, 95% CI: 0.74 ~ 0.97; HR = 0.44, 95% CI: 0.22 ~ 0.89) and previous chemotherapy (HR = 0.80, 95% CI: 0.68 ~ 0.95). However, DFS has not improved with the extension from 7-8 to 10 years (HR = 0.97, 95% CI: 0.85 ~ 1.10). Furthermore, we found no significant difference in overall survival (OS), adverse events (AEs) analysis revealed a significant increase in the incidence of arthralgia, osteoporosis, bone fractures and asthenia after extended AIs. CONCLUSIONS The proportion of patients with breast cancer receiving ET extended beyond 5 years has increased, while the extension of AIs treatment from 5 to 7-8 years may be an option for high-risk patients with well-tolerated tumor size ≥ 2 cm, HR-positive, and previous chemotherapy. However, a variety of adverse events may accompany ET therapy, the identification of factors that may benefit breast cancer patients requires further randomized controlled studies. PROSPERO REGISTRATION NUMBER CRD42022335497.
Collapse
Affiliation(s)
- Zhang Ying
- Department of General Surgery, Qinghai Provincial People's Hospital, Xining, 810000, China.
| | - Liu Linxun
- Department of General Surgery, Qinghai Provincial People's Hospital, Xining, 810000, China
| | - Zhao Kechang
- Department of General Surgery, Qinghai Provincial People's Hospital, Xining, 810000, China
| | - Wang Xiaowu
- Department of Tumor Surgery, Affiliated Hospital of Qinghai University, Xining, 810000, China
| | - Gengzhi Huazhen
- Department of Tumor Surgery, Affiliated Hospital of Qinghai University, Xining, 810000, China
| | - Ma Zhijun
- Department of Tumor Surgery, Affiliated Hospital of Qinghai University, Xining, 810000, China.
| |
Collapse
|
35
|
Hernández CF, Villaman C, Leu C, Lal D, Mata I, Klein AD, Pérez-Palma E. Polygenic score analysis identifies distinct genetic risk profiles in Alzheimer's disease comorbidities. Sci Rep 2025; 15:11407. [PMID: 40181078 PMCID: PMC11968852 DOI: 10.1038/s41598-025-95755-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 03/24/2025] [Indexed: 04/05/2025] Open
Abstract
Alzheimer's disease (AD) is usually accompanied by comorbidities such as type 2 diabetes (T2D), epilepsy, major depressive disorder (MDD), and migraine headaches (MH) that can significantly affect patient management and progression. As AD, these comorbidities have their own cumulative common genetic risk component that can be explored in a single individual through polygenic scores. Utilizing data from the UK Biobank, we investigated the correlation between polygenic scores (PGS) for these comorbidities and their actual presentation in AD patients. We show that individuals with higher PGS values showed an elevated risk of developing T2D (OR 2.1, p = 1.07 × 10-11) and epilepsy (OR 1.5, p = 0.0176). High T2D-PGS is also associated with an earlier AD onset in individuals at high genetic risk for AD (AD-PGS). In contrast, no significant genetic associations were found for MDD and MH. Our findings show distinct common genetic risk factors for T2D and epilepsy carried by AD patients that are associated with increased prevalence and earlier disease onset. These results highlight the contribution of common genetic variation to the broader clinical landscape of AD and will contribute to future tailored patient management strategies for individuals at high genetic risk.
Collapse
Affiliation(s)
- Carlos F Hernández
- Universidad del Desarrollo, Centro de Genética y Genómica, Facultad de Medicina Clínica Alemana, 7610658, Santiago, Chile
| | - Camilo Villaman
- Universidad del Desarrollo, Centro de Genética y Genómica, Facultad de Medicina Clínica Alemana, 7610658, Santiago, Chile
| | - Costin Leu
- Center for Neurogenetics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Dennis Lal
- Center for Neurogenetics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
- Department of Neurology, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Cologne Center for Genomics (CCG), Medical Faculty of the University of Cologne, 50923, Köln, Germany
| | - Ignacio Mata
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Andrés D Klein
- Universidad del Desarrollo, Centro de Genética y Genómica, Facultad de Medicina Clínica Alemana, 7610658, Santiago, Chile
| | - Eduardo Pérez-Palma
- Universidad del Desarrollo, Centro de Genética y Genómica, Facultad de Medicina Clínica Alemana, 7610658, Santiago, Chile.
| |
Collapse
|
36
|
Boselli D, Clemente F, Di Terlizzi S, Pagiatakis C, Papa L, Del Zotto G, Villa C, Ramirez GA, Maugeri N, Manfredi AA, Anselmo A. Unravelling Plasma Extracellular Vesicle Diversity With Optimised Spectral Flow Cytometry. JOURNAL OF EXTRACELLULAR BIOLOGY 2025; 4:e70045. [PMID: 40292386 PMCID: PMC12025886 DOI: 10.1002/jex2.70045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 02/27/2025] [Accepted: 03/13/2025] [Indexed: 04/30/2025]
Abstract
Extracellular vesicles (EVs) are crucial for intercellular communication and are found in various biological fluids. The identification and immunophenotyping of such small particles continue to pose significant challenges. Here, we have developed a workflow for the optimisation of a next-generation panel for in-depth immunophenotyping of circulating plasma EVs using spectral flow cytometry. Our data collection followed a multistep optimisation phase for both instrument setup and 21-colour panel design, thus maximising fluorescent signal recovery. This spectral approach enabled the identification of novel EV subpopulations. Indeed, besides common EVs released by erythrocytes, platelets, leukocytes and endothelial cells, we observed rare and poorly known EV subsets carrying antigens related to cell activation or exhaustion. Notably, the unsupervised data analysis of major EV subsets revealed subpopulations expressing up to five surface antigens simultaneously. However, the majority of EVs expressed only a single surface antigen, suggesting they may not fully represent the phenotype of their parent cells. This is likely due to the small surface area or the biogenesis of EVs rather than antibody steric hindrance. Finally, we tested our workflow by analysing the plasma EV landscape in a cohort of systemic lupus erythematosus (SLE) patients. Interestingly, we observed a significant increase in CD54+ EVs, supporting the notion of elevated circulating ICAM under SLE conditions. To our knowledge, these are the first data highlighting the importance of a spectral flow cytometry approach in deciphering the heterogeneity of plasma EVs paving the way for the routine use of a high-dimensional immunophenotyping in EV research.
Collapse
Affiliation(s)
- Daniela Boselli
- Experimental Imaging Center, FRACTAL, Flow cytometry Resource, Advanced Cytometry Technical Applications LaboratoryIRCCS Ospedale San RaffaeleMilanItaly
| | - Francesca Clemente
- Experimental Imaging Center, FRACTAL, Flow cytometry Resource, Advanced Cytometry Technical Applications LaboratoryIRCCS Ospedale San RaffaeleMilanItaly
| | - Simona Di Terlizzi
- Experimental Imaging Center, FRACTAL, Flow cytometry Resource, Advanced Cytometry Technical Applications LaboratoryIRCCS Ospedale San RaffaeleMilanItaly
| | - Christina Pagiatakis
- Department of Cardiovascular MedicineIRCCS Humanitas Research HospitalRozzanoMilanItaly
- Department of Biotechnology and Life SciencesUniversity of InsubriaVareseItaly
| | - Laura Papa
- Department of Cardiovascular MedicineIRCCS Humanitas Research HospitalRozzanoMilanItaly
| | - Genny Del Zotto
- Department of Research and DiagnosticsIRCCS Istituto Giannina GasliniGenoaItaly
| | - Chiara Villa
- Experimental Imaging Center, FRACTAL, Flow cytometry Resource, Advanced Cytometry Technical Applications LaboratoryIRCCS Ospedale San RaffaeleMilanItaly
- Università Vita‐Salute San RaffaeleMilanItaly
| | - Giuseppe Alvise Ramirez
- Unit of Immunology, Rheumatology, Allergy and Rare DiseasesIRCCS Ospedale San RaffaeleMilanItaly
- Division of Immunology, Transplantation and Infectious DiseasesIRCCS Ospedale San RaffaeleMilanItaly
| | - Norma Maugeri
- Università Vita‐Salute San RaffaeleMilanItaly
- Division of Immunology, Transplantation and Infectious DiseasesIRCCS Ospedale San RaffaeleMilanItaly
| | - Angelo A. Manfredi
- Università Vita‐Salute San RaffaeleMilanItaly
- Unit of Immunology, Rheumatology, Allergy and Rare DiseasesIRCCS Ospedale San RaffaeleMilanItaly
- Division of Immunology, Transplantation and Infectious DiseasesIRCCS Ospedale San RaffaeleMilanItaly
| | - Achille Anselmo
- Experimental Imaging Center, FRACTAL, Flow cytometry Resource, Advanced Cytometry Technical Applications LaboratoryIRCCS Ospedale San RaffaeleMilanItaly
- Università Vita‐Salute San RaffaeleMilanItaly
| |
Collapse
|
37
|
Raghavendra AS, Bassett R, Damodaran S, Barcenas CH, Mouabbi JA, Layman R, Tripathy D. Clinical Characteristics and Survival Outcomes of Metastatic Invasive Lobular and Ductal Carcinoma. JAMA Netw Open 2025; 8:e251888. [PMID: 40293750 PMCID: PMC12038507 DOI: 10.1001/jamanetworkopen.2025.1888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 01/24/2025] [Indexed: 04/30/2025] Open
Abstract
Importance Comparing the clinical and molecular features of metastatic invasive lobular carcinoma (mILC) and metastatic invasive ductal carcinoma (mIDC) is essential to enhance understanding of breast cancer biology and improve personalized treatment approaches. Objective To compare mILC and mIDC in terms of survival outcomes and to investigate the association of clinicopathologic characteristics with those outcomes. Design, Setting, and Participants This cohort study included adult patients at the University of Texas MD Anderson Cancer Center with their first metastatic diagnosis occurring between January 1997 and December 2020. Patient records were obtained from an institutional database. The study follow-up concluded in July 2023, and data were analyzed from July to December 2024. Exposure Diagnosis of mIDC and mILC. Main Outcomes and Measures Progression-free survival (PFS), overall survival (OS), and disease-free interval (DFI) were estimated using the Kaplan-Meier method. Survival distributions were compared using the log-rank test. Multivariable Cox proportional hazards regression was used to assess the association of metastasis onset, estrogen receptor (ER) expression level, and tumor grade with OS and PFS. Results The analysis included a total of 9714 patients (9628 women [99%]), 8535 with mIDC and 1179 with mILC. The median age at metastasis was 53.3 years (range, 17.6-62.0 years). Generally, patients with mILC were older and had lower nuclear grade tumors and fewer metastasis sites than patients with mIDC. Patients with mILC had longer PFS (median, 0.65 years; 95% CI, 0.58-0.74) than patients with mIDC (median, 0.46 years; 95% CI, 0.45-0.48) (hazard ratio [HR], 0.78; 95% CI, 0.73-0.84; P < .001). For OS, patients with mILC had longer OS (median, 3.06 years; 95% CI, 2.87-3.29) than patients with mIDC (median, 2.60 years; 95% CI, 2.52-2.67 years) (HR, 0.91; 95% CI, 0.84-0.98; P = .01). Overall, patients with mILC had longer DFI than patients with mIDC (HR, 0.69; 95% CI, 0.64-0.75; P < .001). At initial diagnosis, patients with mILC were less likely to present with visceral metastasis (522 patients [44.3%]) than patients with mIDC (4909 patients [57.5%]). A higher proportion of patients with mILC (931 patients [79.0%]) than patients with mIDC (5431 patients [63.6%]) had bone-only metastasis. Conclusions and Relevance In this cohort study, patients with mILC had longer OS and PFS than those with mIDC. Metastasis onset, ER positivity, and tumor grade were associated with survival outcomes, and distinct metastatic patterns of mIDC and mILC were also associated with survival for mIDC and mILC, which may help guide more personalized treatment strategies for each subtype.
Collapse
Affiliation(s)
- Akshara S. Raghavendra
- Division of Cancer Medicine, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Roland Bassett
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston
| | - Senthil Damodaran
- Division of Cancer Medicine, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Carlos H. Barcenas
- Division of Cancer Medicine, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Jason A. Mouabbi
- Division of Cancer Medicine, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Rachel Layman
- Division of Cancer Medicine, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Debu Tripathy
- Division of Cancer Medicine, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| |
Collapse
|
38
|
Yeung CLS, Ng TH, Lai CJ, Xue T, Mao X, Tey SK, Lo RCL, Sin C, Ng KM, Wong DKH, Mak L, Yuen M, Ng IO, Cao P, Gao Y, Yun JP, Yam JWP. Small Extracellular Vesicle-Derived Nicotinamide Phosphoribosyltransferase (NAMPT) Induces Acyl-Coenzyme A Synthetase SLC27A4-Mediated Glycolysis to Promote Hepatocellular Carcinoma. J Extracell Vesicles 2025; 14:e70071. [PMID: 40237223 PMCID: PMC12000932 DOI: 10.1002/jev2.70071] [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: 08/19/2024] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
Tumour-derived small extracellular vesicles (sEV) are critical mediators within the tumour microenvironment (TME) and are known to regulate various metabolic pathways. In metastatic hepatocellular carcinoma (HCC), mass spectrometry protein analysis of HCC-derived sEV (HCC-sEV) identified an upregulation of nicotinamide phosphoribosyltransferase (NAMPT), a key enzyme in maintaining cellular nicotinamide adenine dinucleotide (NAD+) levels. Our study demonstrates that sEV-NAMPT enhances glycolysis, tumorigenesis, and metastasis in HCC. Specifically, sEV-NAMPT activates the NF-κB transcription factor through toll-like receptor 4 (TLR4), leading to elevated SLC27A4 expression. SLC27A4 functions primarily as a long-chain fatty acid transporter and acyl-CoA synthetase. Lipidomic and metabolomic analyses revealed a positive correlation between SLC27A4 and intracellular levels of triacylglycerol (TG) and dihydroxyacetone phosphate (DHAP). Increased TG levels enhance lipolysis via hepatic lipase and facilitate the conversion of glycerol-3-P to DHAP, an intermediate that bridges lipid metabolism and glycolysis. This study uncovers a novel regulatory axis involving sEV-NAMPT and SLC27A4 in glycolysis, independent of traditional fatty acid metabolism pathways. Clinically, targeting sEV-NAMPT with the inhibitor FK866 significantly inhibited tumour growth in various HCC in vivo models, highlighting the potential of sEV-NAMPT as both a biomarker and therapeutic target in HCC.
Collapse
Affiliation(s)
- Cherlie Lot Sum Yeung
- Department of Pathology, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong
| | - Tung Him Ng
- Department of Pathology, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong
| | - Charlotte Jiaqi Lai
- Department of Pathology, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong
| | - Tingmao Xue
- Department of Pathology, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong
- Department of Hepatobiliary Surgery IIZhujiang Hospital, Southern Medical UniversityGuangzhouGuangdongChina
| | - Xiaowen Mao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical SciencesUniversity of MacauMacau
| | - Sze Keong Tey
- Department of Surgery, School of Clinical Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong
| | - Regina Cheuk Lam Lo
- Department of Pathology, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong
| | - Chun‐Fung Sin
- Department of Pathology, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong
| | - Kwan Ming Ng
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Hong Kong Science ParkHong Kong
| | - Danny Ka Ho Wong
- Department of Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong
| | - Lung‐Yi Mak
- Department of Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong
- State Key Laboratory of Liver ResearchThe University of Hong KongHong Kong
| | - Man‐Fung Yuen
- Department of Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong
- State Key Laboratory of Liver ResearchThe University of Hong KongHong Kong
| | - Irene Oi‐Lin Ng
- Department of Pathology, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong
- State Key Laboratory of Liver ResearchThe University of Hong KongHong Kong
| | - Peihua Cao
- Clinical Research Center, Zhujiang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Yi Gao
- Department of Hepatobiliary Surgery IIZhujiang Hospital, Southern Medical UniversityGuangzhouGuangdongChina
| | - Jing Ping Yun
- Department of PathologySun Yat‐sen University Cancer CenterGuangzhouGuangdongChina
| | - Judy Wai Ping Yam
- Department of Pathology, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong
- State Key Laboratory of Liver ResearchThe University of Hong KongHong Kong
- Materials Innovation Institute for Life Sciences and Energy (MILES), HKU‐SIRIShenzhenChina
| |
Collapse
|
39
|
Mahajan M, Dhabalia S, Dash T, Sarkar A, Mondal S. A comprehensive multi-omics study reveals potential prognostic and diagnostic biomarkers for colorectal cancer. Int J Biol Macromol 2025; 303:140443. [PMID: 39909246 DOI: 10.1016/j.ijbiomac.2025.140443] [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: 07/18/2024] [Revised: 12/11/2024] [Accepted: 01/27/2025] [Indexed: 02/07/2025]
Abstract
BACKGROUND AND OBJECTIVE Colorectal cancer (CRC) is a complex disease with diverse genetic alterations and causes 10 % of cancer-related deaths worldwide. Understanding its molecular mechanisms is essential for identifying potential biomarkers and therapeutic targets for its effective management. METHODS We integrated copy number alterations (CNA) and mutation data via their differentially expressed genes termed as candidate genes (CGs) computed using bioinformatics approaches. Then, using the CGs, we perform Weighted correlation network analysis (WGCNA) and utilise several hazard models such as Univariate Cox, Least Absolute Shrinkage and Selection Operator (LASSO) Cox and multivariate Cox to identify the key genes involved in CRC progression. We used different machine-learning models to demonstrate the discriminative power of selected hub genes among normal and CRC (early and late-stage) samples. RESULTS The integration of CNA with mRNA expression identified over 3000 CGs, including CRC-specific driver genes like MYC and APC. In addition, pathway analysis revealed that the CGs are mainly enriched in endocytosis, cell cycle, wnt signalling and mTOR signalling pathways. Hazard models identified four key genes, CASP2, HCN4, LRRC69 and SRD5A1, that were significantly associated with CRC progression and predicted the 1-year, 3-years, and 5-years survival times. WGCNA identified seven hub genes: DSCC1, ETV4, KIAA1549, NOP56, RRS1, TEAD4 and ANKRD13B, which exhibited strong predictive performance in distinguishing normal from CRC (early and late-stage) samples. CONCLUSIONS Integrating regulatory information with gene expression improved early versus late-stage prediction. The identified potential prognostic and diagnostic biomarkers in this study may guide us in developing effective therapeutic strategies for CRC management.
Collapse
Affiliation(s)
- Mohita Mahajan
- Department of Biological Sciences, Birla Institute of Technology and Science Pilani, K.K. Birla Goa campus, Zuarinagar, Goa 403726, India.
| | - Subodh Dhabalia
- Department of Mathematics, Amrita Vishwa Vidyapeetham, Amritanagar, Coimbatore 64112, India.
| | - Tirtharaj Dash
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK.
| | - Angshuman Sarkar
- Department of Biological Sciences, Birla Institute of Technology and Science Pilani, K.K. Birla Goa campus, Zuarinagar, Goa 403726, India.
| | - Sukanta Mondal
- Department of Biological Sciences, Birla Institute of Technology and Science Pilani, K.K. Birla Goa campus, Zuarinagar, Goa 403726, India.
| |
Collapse
|
40
|
Wimmer K, Hlauschek D, Balic M, Pfeiler G, Greil R, Singer CF, Halper S, Steger G, Suppan C, Gampenrieder SP, Helfgott R, Egle D, Filipits M, Jakesz R, Sölkner L, Fesl C, Gnant M, Fitzal F. Validation of the CTS5 in four prospective, multicenter, randomized ABCSG trials. Breast 2025; 80:104415. [PMID: 39985843 PMCID: PMC11904565 DOI: 10.1016/j.breast.2025.104415] [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: 11/14/2024] [Revised: 01/09/2025] [Accepted: 02/06/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND The Clinical Treatment Score post-5 years (CTS5) is a clinicopathological tool designed to estimate late distant recurrence (LDR) in hormone receptor-positive (HR+) breast cancer patients after 5 years of adjuvant endocrine therapy (ET). While intended as a prognostic algorithm, its predictive value for ET extension remains uncertain. METHODS The score was calculated in 4931 patients from four prospective randomized ABCSG trials (ABCSG-6, -6a, -8, and -16) with 250 LDR events. We assessed its prognostic power, calibration accuracy, and predictive value. Time to LDR was analyzed using Cox regression models. RESULTS In our cohorts, the CTS5 provided prognostic information whether used as a continuous or categorical score. In the ABCSG-8 cohort (n = 2054) and the combined ABCSG-6+8 cohort (n = 3308), a higher continuous score was significantly associated with increased LDR risk. The categorical CTS5 showed that high-risk patients had significantly higher LDR rates compared to low- or intermediate-risk patients. The score slightly overestimated LDR risk, regardless of predicted risk. Although no significant predictive value was found on the relative scale, an absolute LDR risk reduction of 23.4 % was found in patients with a high CTS5 of 5 when extended ET was administered additional five than two years. In patients with a CTS5 of 2, no benefit was found when ET was extended to 10 instead of 7 years. CONCLUSION The CTS5 is a valid tool for LDR risk stratification in HR + breast cancer, but should be used cautiously for determining benefits from ET extension, as no significant predictive value was found.
Collapse
Affiliation(s)
- Kerstin Wimmer
- Division of General Surgery, Department of Surgery, Medical University of Vienna, Vienna, Austria; Austrian Breast & Colorectal Cancer Study Group, Vienna, Austria; Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| | | | - Marija Balic
- Department of Oncology, Medical University of Graz, Austria
| | - Georg Pfeiler
- Department of Gynecology and Obstetrics, Medical University of Vienna, Vienna, Austria; Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Richard Greil
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Paracelsus Medical University Salzburg, Salzburg, Austria; Salzburg Cancer Research Institute-CCCIT, Salzburg, Austria; Cancer Cluster Salzburg, Salzburg, Austria
| | - Christian F Singer
- Department of Gynecology and Obstetrics, Medical University of Vienna, Vienna, Austria; Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | | | - Günther Steger
- Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | | | - Simon Peter Gampenrieder
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Paracelsus Medical University Salzburg, Salzburg, Austria; Salzburg Cancer Research Institute-CCCIT, Salzburg, Austria; Cancer Cluster Salzburg, Salzburg, Austria
| | - Ruth Helfgott
- Department of Surgery, Ordensklinikum Linz - Sisters of Charity, Linz, Austria
| | - Daniel Egle
- Department of Gynecology, Medical University Innsbruck, Innsbruck, Austria
| | - Martin Filipits
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Raimund Jakesz
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Lidija Sölkner
- Austrian Breast & Colorectal Cancer Study Group, Vienna, Austria
| | - Christian Fesl
- Austrian Breast & Colorectal Cancer Study Group, Vienna, Austria
| | - Michael Gnant
- Austrian Breast & Colorectal Cancer Study Group, Vienna, Austria; Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Florian Fitzal
- Division of General Surgery, Department of Surgery, Medical University of Vienna, Vienna, Austria; Department of Gynecology, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
41
|
Bi W, Cao X, Li J, Gao Y, Song Y, He B. Ultrasensitive Detection of Extracellular Vesicles Based on Metal-Organic Framework DNA Biobarcodes Triggered G-Quadruplex Coupled with Rolling Circle Amplification Assay. ACS Sens 2025; 10:2136-2146. [PMID: 40048560 DOI: 10.1021/acssensors.4c03384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Extracellular vesicles (EVs), as liquid biopsy markers for accurate tumor diagnosis, are considered to hold great promise. However, effectively isolating and sensitively detecting EVs with convenience still face challenges. Herein, we propose a highly sensitive and specific platform for EV detection by integrating a metal-organic framework (MOF)-based DNA biobarcodes strategy with a rolling circle amplification (RCA)/G-quadruplex system. In this study, first, Zr-MOFs act as signal converters by comodification with DNA barcodes and antibodies, converting and amplifying the abundance of EVs into DNA barcodes. Second, the released DNA can trigger RCA, followed by G-quadruplex formation to further amplify the signal. Consequently, this approach significantly enhances the sensitivity for EV biomarker detection, achieving a low limit of detection of 100 EVs mL-1. Furthermore, the strategy offers high sensitivity, specificity, accuracy, and simplicity, highlighting its potential for clinical applications in noninvasive EV detection.
Collapse
Affiliation(s)
- Wen Bi
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Xiaoqing Cao
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Jingjing Li
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Yanfeng Gao
- School of Medical Imaging, Wannan Medical College, Wuhu 241002, China
| | - Yujun Song
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210023, China
| | - Bangshun He
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| |
Collapse
|
42
|
Żuber Z, Podwójcic K, Szeląg M, Krajewska-Włodarczyk M, Batko K, Orleański M, Sowiński J, Świderek M, Śmiglewska A, Maluchnik M, Brzosko M, Kwiatkowska B, Stajszczyk M, Batko B. Epidemiology and comorbidity of juvenile idiopathic arthritis in Poland- a nationwide study. Pediatr Rheumatol Online J 2025; 23:33. [PMID: 40156029 PMCID: PMC11951515 DOI: 10.1186/s12969-025-01065-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 01/28/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Diagnostic pathways for patients with juvenile idiopathic arthritis (JIA) have gradually improved over time. Provider practice has also shifted towards goal-oriented treatment with disease-modifying drugs (DMARDs) that together may have changed the epidemiologic landscape of JIA. METHODS Public healthcare utilization records from the National Health Fund (NHF) were screened between 2010 and 2022. For individuals aged < 16 years, we utilized a narrow JIA case definition combining repeat ICD-10 encoding with DMARDs prescription based on ATC codes. RESULTS In 2022, we identified 1,625 incident and 29,758 prevalent JIA cases (< 16 years), which corresponds to incidence (IRs) and prevalence rates of 4.30 and 78.80 per 100,000 persons of the general population. For the pediatric population, annual IRs for JIA (< 16 years) ranged between 24.0 (95% CI 22.8, 25.2) and 38.7 (95% CI 37.2-40.3) per 100,000. Greater susceptibility among females was also consistently observed with the annual IR ratio ranging between 1.16 and 1.53. The most common concurrent disorders based on medical care services were allergic rhinitis (N = 5,200, 17.5%), bronchial asthma (N = 3,661, 12.3%) and chronic tonsillitis/pharyngitis (N = 3641, 12.2%). Analysis of 214,285 outpatient care visits revealed a median (IQR, range) annual healthcare cost of 37.8€ (35.8-47.4€, 30.3-86.1€) per JIA patient. CONCLUSIONS This comprehensive, nationwide study provides a contemporary estimate of JIA burden in Poland. Our findings indicate that both the occurrence of new cases and overall burden of JIA in the past ten years align with the lower end of projected figures for our geographical area, especially when compared with Scandinavian nations.
Collapse
Affiliation(s)
- Zbigniew Żuber
- Department of Pediatrics, Faculty of Medicine and Health Sciences, Andrzej Frycz Modrzewski University, Krakow, Poland.
| | | | - Mateusz Szeląg
- Department of Analysis and Strategy, Ministry of Health, Warsaw, Poland
| | | | - Krzysztof Batko
- Department of Dermatology, University Hospital, Krakow, Poland
| | - Michał Orleański
- Department of Analysis and Strategy, Ministry of Health, Warsaw, Poland
| | - Jakub Sowiński
- Department of Analysis and Strategy, Ministry of Health, Warsaw, Poland
| | - Maria Świderek
- Department of Analysis and Strategy, Ministry of Health, Warsaw, Poland
| | - Agata Śmiglewska
- Department of Analysis and Strategy, Ministry of Health, Warsaw, Poland
| | - Michał Maluchnik
- Department of Adult Neurology, Medical University of Gdansk, Gdansk, Poland
| | - Marek Brzosko
- Department of Rheumatology, Internal Diseases, Geriatrics and Clinical Immunology, Faculty of Medicine and Dentistry, Pomeranian Medical University, Szczecin, Poland
| | - Brygida Kwiatkowska
- Clinic of Early Arthritis, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Marcin Stajszczyk
- Department of Rheumatology and Autoimmune Diseases, Silesian Center for Rheumatology, Orthopedics and Rehabilitation, Ustroń, Poland
| | - Bogdan Batko
- Department of Rheumatology and Immunology, Faculty of Medicine and Health Sciences, Andrzej Frycz Modrzewski University, Krakow, Poland
| |
Collapse
|
43
|
Sarkar S, Schechter C, Kurian AW, Caswell-Jin JL, Jayasekera J, Mandelblatt JS. Impact of endocrine therapy regimens for early-stage ER+/HER2-breast cancer on contralateral breast cancer risk. NPJ Breast Cancer 2025; 11:30. [PMID: 40140385 PMCID: PMC11947086 DOI: 10.1038/s41523-025-00746-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 03/15/2025] [Indexed: 03/28/2025] Open
Abstract
Endocrine therapy for breast cancer may reduce the risk of contralateral breast cancer (CBC). However, there are no published estimates quantifying the lifetime outcomes by age at primary diagnosis, regimen, or duration. Here, we adapted an established Cancer Intervention and Surveillance Network (CISNET) model to simulate life histories of multiple US female birth-cohorts diagnosed with stage 0-III ER+/HER2- breast cancer receiving different durations (none, 2.5, 5, 10 years) of two endocrine therapy regimens (aromatase inhibitors or tamoxifen; including ovarian-function suppression for premenopausal women). As expected, greater duration of endocrine therapy led to more avoided CBC cases, as did aromatase inhibitors over tamoxifen, but the numbers varied greatly by the age of diagnosis. The maximum number of CBC were avoided using 10-year aromatase inhibitor regimens (6.0 vs. 11.2 for no adjuvant therapy, per 100 women with ER+/HER2- breast cancer). For the 5-year aromatase inhibitors therapy, women <45 years had the largest reduction in CBC cases (5.0/100), which dropped to 2.7/100 for women at 75+ years. Quantification of the lifetime risk of CBC for specific endocrine therapy types and duration is helpful for weighing therapeutic options. The risk of breast cancer death has a larger weight, but inclusion of the risk of CBC increases the separation between different therapy options.
Collapse
Affiliation(s)
- Swarnavo Sarkar
- Department of Oncology, Georgetown University Medical Center and Cancer Prevention and Control Program, Lombardi Comprehensive Cancer Center, Washington, DC, USA.
| | - Clyde Schechter
- Department of Family and Social Medicine, Albert Einstein College of Medicine, New York City, NY, USA
| | - Allison W Kurian
- Department of Medicine (Oncology) and Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA, USA
| | - Jennifer L Caswell-Jin
- Department of Medicine (Oncology), Stanford University School of Medicine, Stanford, CA, USA
| | - Jinani Jayasekera
- Health Equity and Decision Sciences Laboratory, National Institute on Minority Health and Health Disparities, National Institutes of Health, Rockville, MD, USA
| | - Jeanne S Mandelblatt
- Department of Oncology, Georgetown University Medical Center and Cancer Prevention and Control Program, Lombardi Comprehensive Cancer Center, Washington, DC, USA
- Georgetown Lombardi Institute for Cancer and Aging REsearch (I-CARE), Lombardi Comprehensive Cancer Center, Washington, DC, USA
| |
Collapse
|
44
|
Messina M, Nechuta S. A Review of the Clinical and Epidemiologic Evidence Relevant to the Impact of Postdiagnosis Isoflavone Intake on Breast Cancer Outcomes. Curr Nutr Rep 2025; 14:50. [PMID: 40131602 PMCID: PMC11937148 DOI: 10.1007/s13668-025-00640-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2025] [Indexed: 03/27/2025]
Abstract
PURPOSE OF REVIEW This narrative review aims to determine the impact of postdiagnosis isoflavone intake, via supplements and foods, on breast cancer outcomes. Foods derived from soybeans are uniquely rich sources of isoflavones, naturally occurring compounds that can bind to estrogen receptors although the extent to which they exert estrogen-like effects in humans is unclear. Isoflavones have been rigorously investigated for a wide range of health benefits including breast cancer prevention. However, their classification as phytoestrogens has led to concern that isoflavones and hence, soy food consumption, could worsen the prognosis of women with breast cancer and interfere with the efficacy of endocrine therapy for this disease. RECENT FINDINGS Research in athymic ovariectomized mice shows isoflavones stimulate the growth of existing estrogen-sensitive mammary tumors. However, extensive clinical research indicates that neither soy foods nor isolated isoflavones affect markers of breast cancer risk including mammographic density and breast cell proliferation. No effects are observed even when isoflavone exposure greatly exceeds typical intake in Asian countries. Furthermore, the results from epidemiologic studies indicate postdiagnosis isoflavone intake from soy foods reduces recurrence and possibly mortality from breast cancer. Additionally, the limited observational data do not show that isoflavones interfere with the efficacy of tamoxifen or aromatase inhibitors. Regardless of their treatment status, evidence indicates that women with breast cancer can safely consume soy foods. Limiting intake to no more than two servings of traditional Asian soy foods daily, an amount that provides approximately 50 mg isoflavones, is recommended, not because data indicate exceeding this amount is harmful, but because few population-based studies involved participants consuming more than this intake recommendation.
Collapse
Affiliation(s)
- Mark Messina
- Soy Nutrition Institute Global, Jefferson, MO, USA.
| | | |
Collapse
|
45
|
Alves SS, Rossi L, de Oliveira JAC, Servilha-Menezes G, Grigorio-de-Sant'Ana M, Mazzei RF, Almeida SS, Sebollela A, da Silva Junior RMP, Garcia-Cairasco N. Metformin Improves Spatial Memory and Reduces Seizure Severity in a Rat Model of Epilepsy and Alzheimer's Disease comorbidity via PI3K/Akt Signaling Pathway. Mol Neurobiol 2025:10.1007/s12035-025-04844-2. [PMID: 40126600 DOI: 10.1007/s12035-025-04844-2] [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: 10/15/2024] [Accepted: 03/12/2025] [Indexed: 03/25/2025]
Abstract
Emerging evidence suggests a bidirectional relationship between Alzheimer's disease (AD) and epilepsy. In our previous studies, we identified a partial AD-like phenotype associated with central insulin resistance in the Wistar audiogenic rat (WAR), a genetic model of epilepsy. We also found that intracerebroventricular administration of streptozotocin, a compound used to model diabetes and AD, exacerbates seizure susceptibility. Given the role of insulin signaling in both AD and epilepsy, we hypothesized that metformin (MET), an anti-diabetic drug known for enhancing insulin sensitivity, could be a potential therapeutic agent for both conditions. Our objective was to investigate MET's effects on brain insulin signaling, seizure activity, and AD-like pathology in WARs. Adult male WARs received oral MET (250 mg/kg) for 21 days. Audiogenic seizures were assessed using the Categorized Severity Index and Racine's scale. Spatial memory was tested with the Morris water maze (MWM), followed by Western blot analysis of hippocampal proteins. MET significantly reduced seizure severity and improved MWM performance. Although MET did not affect insulin receptor levels or activation, it increased phosphoinositide 3-kinase (PI3K), activated Akt, and increased glycogen synthase kinase-3α/β (GSK-3α/β) levels. MET also decreased amyloid β precursor protein (AβPP) levels but did not affect Tau phosphorylation. These results suggest that chronic MET treatment alleviates behaviors related to both AD and epilepsy in WARs and modulates insulin signaling independently of insulin receptor activation. Our findings highlight MET's potential as a therapeutic agent for managing comorbid AD and epilepsy, warranting further investigation into its mechanisms of action.
Collapse
Affiliation(s)
- Suélen Santos Alves
- Department of Neurosciences and Behavioral Sciences, Ribeirão Preto Medical School University of São Paulo (FMRP-USP), Ribeirão Preto, Brazil
| | - Letícia Rossi
- Department of Physiology, Neurophysiology and Experimental Neuroethology Laboratory, Ribeirão Preto Medical School University of São Paulo (FMRP-USP), Ribeirão Preto, Brazil
| | - Jose Antonio Cortes de Oliveira
- Department of Physiology, Neurophysiology and Experimental Neuroethology Laboratory, Ribeirão Preto Medical School University of São Paulo (FMRP-USP), Ribeirão Preto, Brazil
| | - Gabriel Servilha-Menezes
- Department of Physiology, Neurophysiology and Experimental Neuroethology Laboratory, Ribeirão Preto Medical School University of São Paulo (FMRP-USP), Ribeirão Preto, Brazil
| | - Mariana Grigorio-de-Sant'Ana
- Department of Physiology, Neurophysiology and Experimental Neuroethology Laboratory, Ribeirão Preto Medical School University of São Paulo (FMRP-USP), Ribeirão Preto, Brazil
| | - Rodrigo Focosi Mazzei
- Department of Psychology, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto University of São Paulo (FFCLRP-USP), Ribeirão Preto, Brazil
| | - Sebastião Sousa Almeida
- Department of Psychology, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto University of São Paulo (FFCLRP-USP), Ribeirão Preto, Brazil
| | - Adriano Sebollela
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School University of São Paulo (FMRP-USP), Ribeirão Preto, Brazil
| | | | - Norberto Garcia-Cairasco
- Department of Neurosciences and Behavioral Sciences, Ribeirão Preto Medical School University of São Paulo (FMRP-USP), Ribeirão Preto, Brazil.
- Department of Physiology, Neurophysiology and Experimental Neuroethology Laboratory, Ribeirão Preto Medical School University of São Paulo (FMRP-USP), Ribeirão Preto, Brazil.
| |
Collapse
|
46
|
Hong Y, Liu Y, Shen H, Li B, Li Q. A strategy for synergistic enhancement of immune circulation in head and neck squamous cell carcinoma by novel nucleic acid drug therapy and immunotherapy. J Transl Med 2025; 23:354. [PMID: 40114181 PMCID: PMC11927285 DOI: 10.1186/s12967-025-06344-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 03/01/2025] [Indexed: 03/22/2025] Open
Abstract
Studies have shown that in the pathogenesis of head and neck squamous cell carcinoma, immune circulation obstruction caused by various factors including metabolic abnormalities, gene mutations, and matrix barrier, is a critical factor for the induction of tumor development and progression. Therefore, the immunotherapy strategy of killing head and neck squamous cell carcinoma cells by an enhanced immune circulation mechanism has attracted much attention. In addition, the rapid development of new nucleic acid drug therapy, such as mRNA, oligonucleotide and small guide RNA (sgRNA), has taken immunotherapy of head and neck squamous cell carcinoma (immune checkpoint inhibitors, tumor vaccines, cellular immunotherapy, cytokines and adjuvants, etc.) to a new level. The combination of nucleic acid therapy with immunotherapy developed for its therapeutic properties has brought a new direction for the diagnosis and treatment of head and neck squamous cell carcinoma, and the combination of the two has had considerable curative effect to patients with refractory/recurrent head and neck squamous cell carcinoma. In this review, we summarized the latest progress of nucleic acid therapy applied to conventional immunotherapy for head and neck squamous cell carcinoma, discussed its mechanism of action and efficacy, and looked into the future development trend.
Collapse
Affiliation(s)
- Yangjian Hong
- Key Laboratory of Head & Neck Cancer Translation Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Yanyang Liu
- Key Laboratory of Head & Neck Cancer Translation Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Huize Shen
- Key Laboratory of Head & Neck Cancer Translation Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Bowen Li
- Key Laboratory of Head & Neck Cancer Translation Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China.
- Hangzhou Institute of Medicine(HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China.
| | - Qinglin Li
- Key Laboratory of Head & Neck Cancer Translation Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China.
| |
Collapse
|
47
|
Su Y, Bai Q, Zhang W, Xu B, Hu T. The Role of Long Non-Coding RNAs in Modulating the Immune Microenvironment of Triple-Negative Breast Cancer: Mechanistic Insights and Therapeutic Potential. Biomolecules 2025; 15:454. [PMID: 40149989 PMCID: PMC11939868 DOI: 10.3390/biom15030454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 03/18/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly heterogeneous and aggressive subtype of breast cancer that faces therapeutic challenges due to a shortage of effective targeted therapies. The complex biology of TNBC renders its clinical management fraught with difficulties, especially regarding the immune microenvironment of the tumor. In recent years, long non-coding RNAs (lncRNAs) have been recognized as important gene regulators with key roles in tumor development and microenvironmental regulation. Previous studies have shown that lncRNAs play important roles in the immune microenvironment of TNBC, including the regulation of tumor immune escape and the function of tumor-infiltrating immune cells. However, despite the increasing research on lncRNAs, there are still many unanswered questions, such as their specific mechanism of action and how to effectively utilize them as therapeutic targets. Therefore, the aim of this study was to review the mechanisms of lncRNAs in the TNBC immune microenvironment, explore their regulatory roles in tumor immune escape and immune cell infiltration, and explore their prospects as potential therapeutic targets. By integrating the latest research results, this study aims to provide new ideas and directions for future TNBC treatment.
Collapse
Affiliation(s)
- Yongcheng Su
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (Y.S.); (Q.B.); (W.Z.)
| | - Qingquan Bai
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (Y.S.); (Q.B.); (W.Z.)
| | - Wenqing Zhang
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (Y.S.); (Q.B.); (W.Z.)
| | - Beibei Xu
- Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Tianhui Hu
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, China; (Y.S.); (Q.B.); (W.Z.)
- Shenzhen Research Institute, Xiamen University, Shenzhen 518057, China
| |
Collapse
|
48
|
Liu R, Zhang F, He X, Huang K. Plant Derived Exosome-Like Nanoparticles and Their Therapeutic Applications in Glucolipid Metabolism Diseases. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:6385-6399. [PMID: 40048449 DOI: 10.1021/acs.jafc.4c12480] [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: 03/20/2025]
Abstract
Plant derived exosome-like nanoparticles (PELNs) are membrane structures isolated from different plants, which encapsulate many active substances such as proteins, lipids, and nucleic acids, which exert a substantial influence on many physiological processes such as plant growth and development, self-defense, and tissue repair. Compared with synthetic nanoparticles and mammalian cell derived exosomes (MDEs), PELNs have lower toxicity and immunogenicity and possess excellent biocompatibility. The intrinsic properties of PELNs establish a robust basis for their applications in the therapeutic management of a diverse array of pathologies. It is worth mentioning that PELNs have good biological targeting, which promotes them to load and deliver drugs to specific tissues, offering a superior development pathway for the construction of a new drug delivery system (DDS). Glucose and lipid metabolism is a vital life process for the body's energy and material supply. The maintenance of homeostatic balance provides a fundamental basis for the body's ability to adjust to modifications in both its internal and external environment. Conversely, homeostatic imbalance can lead to a range of severe metabolic disorders. This work provides a comprehensive overview of the extraction and representation methods of PELNs, their transportation and storage characteristics, and their applications as therapeutic agents for direct treatment and as delivery vehicles to enhance nutrition and health. Additionally, it examines the therapeutic efficacy and practical applications of PELNs in addressing abnormalities in glucose and lipid metabolism. Finally, combined with the above contents, the paper summarizes and provides a conceptual framework for the better application of PELNs in clinical disease treatment.
Collapse
Affiliation(s)
- Ruolan Liu
- Key Laboratory of Precision Nutrition and Food Quality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Feng Zhang
- Key Laboratory of Precision Nutrition and Food Quality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Xiaoyun He
- Key Laboratory of Precision Nutrition and Food Quality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture and Rural Affairs of the P.R. China, Beijing 100083, China
| | - Kunlun Huang
- Key Laboratory of Precision Nutrition and Food Quality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture and Rural Affairs of the P.R. China, Beijing 100083, China
| |
Collapse
|
49
|
Chang O, Cheon S, Semenova N, Azad N, Iyer AK, Yakisich JS. Prolonged Low-Dose Administration of FDA-Approved Drugs for Non-Cancer Conditions: A Review of Potential Targets in Cancer Cells. Int J Mol Sci 2025; 26:2720. [PMID: 40141362 PMCID: PMC11942989 DOI: 10.3390/ijms26062720] [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] [Revised: 03/09/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
Though not specifically designed for cancer therapy, several FDA-approved drugs such as metformin, aspirin, and simvastatin have an effect in lowering the incidence of cancer. However, there is a great discrepancy between in vitro concentrations needed to eliminate cancer cells and the plasma concentration normally tolerated within the body. At present, there is no universal explanation for this discrepancy and several mechanisms have been proposed including targeting cancer stem cells (CSCs) or cellular senescence. CSCs are cells with the ability of self-renewal and differentiation known to be resistant to chemotherapy. Senescence is a response to damage and stress, characterized by permanent cell-cycle arrest and apoptotic resistance. Although, for both situations, there are few examples where low concentrations of the FDA-approved drugs were the most effective, there is no satisfactory data to support that either CSCs or cellular senescence are the target of these drugs. In this review, we concisely summarize the most used FDA-approved drugs for non-cancer conditions as well as their potential mechanisms of action in lowering cancer incidence. In addition, we propose that prolonged low-dose administration (PLDA) of specific FDA-approved drugs can be useful for effectively preventing metastasis formation in selected patients.
Collapse
Affiliation(s)
- Olivia Chang
- Governor’s School for Science and Technology, Hampton, VA 23666, USA; (O.C.); (S.C.)
| | - Sarah Cheon
- Governor’s School for Science and Technology, Hampton, VA 23666, USA; (O.C.); (S.C.)
| | - Nina Semenova
- Department of Pharmaceutical Sciences, School of Pharmacy, Hampton University, Hampton, VA 23668, USA; (N.S.); (A.K.I.)
| | - Neelam Azad
- The Office of the Vice President for Research, Hampton University, Hampton, VA 23668, USA;
| | - Anand Krishnan Iyer
- Department of Pharmaceutical Sciences, School of Pharmacy, Hampton University, Hampton, VA 23668, USA; (N.S.); (A.K.I.)
| | - Juan Sebastian Yakisich
- Department of Pharmaceutical Sciences, School of Pharmacy, Hampton University, Hampton, VA 23668, USA; (N.S.); (A.K.I.)
| |
Collapse
|
50
|
Mao YW, Zeng HD, Fang Y, Wu XY, Zhang MH, Hu CD, Zhao YX. Effect of subcutaneous fat on long-term survival after surgery for stage I-III breast cancer as determined by computed tomography. Front Oncol 2025; 15:1506629. [PMID: 40165893 PMCID: PMC11955644 DOI: 10.3389/fonc.2025.1506629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 02/27/2025] [Indexed: 04/02/2025] Open
Abstract
Introduction Monitoring body composition through Computed Tomography (CT) scans, including muscle and adipose tissue, plays a key role in the prognosis of various cancers. However, abdominal CT is impractical for patients with breast cancer (BC), making chest CT an essential tool for postoperative surveillance. This study aims to evaluate the effect of subcutaneous fat tissue at the 11th thoracic vertebral plane on the postoperative prognosis of BC patients by analyzing chest CT images, providing evidence for postoperative nutritional and rehabilitation guidance. Methods We conducted a retrospective analysis of the medical records of 188 BC patients treated and discharged from the Second Affiliated Hospital of Wenzhou Medical University between January 1, 2013, and December 31, 2013. The subcutaneous fat area (SFA) at the 11th thoracic vertebra (T11) was measured using chest CT images, and the subcutaneous fat index (SFI, area/height2) was calculated. Using multivariate Cox proportional hazards models and propensity score matching (PSM), the relationships between the SFI and overall survival (OS), as well as recurrence-free survival (RFS), were assessed. Additionally, Kaplan-Meier survival curves were applied to compare prognostic differences between the groups. Results The median follow-up duration was 128 months (range: 27-188 months). Of the 188 patients included in the study, the optimal cutoff value for the SFI was determined to be 49.31 cm²/m². Multivariate analysis indicated that SFI was an independent prognostic factor for both OS (HR 2.50, 95% CI 1.07-5.83, P = 0.034) and RFS (HR 2.04, 95% CI 1.10-3.78, P = 0.024). After PSM, Kaplan-Meier survival curve analysis revealed significant differences in both RFS and OS between the two groups (P = 0.025 and P = 0.018, respectively). All the results showed that the prognosis of BC with more subcutaneous fat was poor. Discussion The findings demonstrated that the SFI at T11 was negatively correlated with patient survival. This offers a new perspective on personalized management for BC patients, suggesting that future research should validate these results and investigate combining imaging assessments with lifestyle interventions, such as exercise, nutrition, and diet, to optimize patient outcomes.
Collapse
Affiliation(s)
- Yi-Wen Mao
- Department of Breast Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hong-Dou Zeng
- Department of Breast Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ye Fang
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xin-Yao Wu
- Department of Breast Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ming-Hao Zhang
- Department of Breast Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Cheng-Da Hu
- Department of Breast Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ya-Xin Zhao
- Department of Breast Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|