1
|
Bai G, Fan Z, Zhao X, Dong Y, Tan C, Bai L, Jiang H, Liu T, Li J, Zhao X, Yu H. Highly sensitive and portable detection of PD-L1 + exosomes using a smartphone-assisted colorimetric sensor. Biosens Bioelectron 2025; 286:117592. [PMID: 40446615 DOI: 10.1016/j.bios.2025.117592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 05/02/2025] [Accepted: 05/16/2025] [Indexed: 06/11/2025]
Abstract
PD-L1+ exosomes act as a useful biomarker in early cancer diagnosis, therapeutic monitoring, prognostic assessment, and immunotherapy for non-invasive liquid biopsy. There exists urgent clinical need for developing rapid, portable, and cost-effective immediate-response assays for PD-L1+ exosomes. Here, we proposed a smartphone-assisted colorimetric sensor using the triple-helix molecular switch (THMS) combined with Y-shaped catalytic hairpin assembly (Y-CHA) reaction. Namely, this strategy initiates the Y-CHA cycle and forms Y-DNA G-quadruplex/Hemin DNAzyme with K+ and Hemin after specific recognition of PD-L1+ exosomes by THMS. Then, PD-L1+ exosomes could be quantified via the oxidation of 3,3',5,5'-tetramethylbenzidine (TMB)-mediated color change. The method demonstrates excellent specificity and sensitivity, with a limit of detection (LOD) of 5.45 × 103 particles/mL and an LOD of 8.56 × 103 particles/mL using smartphone analysis. With this strategy, we found that PD-L1+ exosome levels were significantly elevated in the peripheral blood of patients with colorectal cancer liver metastases (CRLM), indicating great potential for identifying CRLM patients. Additionally, this colorimetric sensor can be used to quantify PD-L1+ exosomes across wide range of cancers, which possess a great prospect in the point-of-care testing filed for cancer companion diagnosis.
Collapse
Affiliation(s)
- Gang Bai
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China; Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Zhichao Fan
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xiaoxin Zhao
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yan Dong
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Congcong Tan
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Liyuan Bai
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Haoran Jiang
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Taorui Liu
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jianjun Li
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Xiang Zhao
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Hua Yu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China; Department of General Surgery, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| |
Collapse
|
2
|
Zhou XY, Jiang YJ, Guo XM, Han DH, Liu Y, Qiao Q. Application of circulating tumor DNA liquid biopsy in nasopharyngeal carcinoma: A case report and review of literature. World J Clin Cases 2025; 13:105066. [DOI: 10.12998/wjcc.v13.i21.105066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/05/2025] [Accepted: 03/26/2025] [Indexed: 04/27/2025] Open
Abstract
BACKGROUND Circulating tumor DNA (ctDNA)-based liquid biopsy has been found to be effective for the detection of minimal residual disease and the evaluation of prognostic risk in various solid tumors, with good sensitivity and specificity for identifying patients at high risk of recurrence. However, use of its results as a biomarker for guiding the treatment and predicting the prognosis of nasopharyngeal carcinoma (NPC) has not been reported.
CASE SUMMARY In this case study of a patient with stage IVb NPC, we utilized ctDNA as an independent biomarker to guide treatment. Chemotherapy was administered in the early stages of the disease, and local intensity-modulated radiation therapy was added when the patient tested positive for ctDNA, while radiation therapy was stopped and the patient was observed when the ctDNA test was negative. During the follow-up period, ctDNA signals became positive before tumor progression and became negative again at the end of treatment. We also explored the potential of ctDNA in combination with Epstein–Barr virus (EBV) DNA status to predict the prognosis of NPC patients, as well as the criteria for selecting genetic mutations and the testing cycle for ctDNA analysis.
CONCLUSION The results of ctDNA-based liquid biopsy can serve as an independent biomarker, either independently or in conjunction with EBV DNA status, to guide the treatment and predict the prognosis of NPC.
Collapse
Affiliation(s)
- Xin-Yao Zhou
- Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Yuan-Jun Jiang
- Department of Urology, The First Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Xiao-Ming Guo
- Department of Artificial Intelligence and Algorithm R and D, Neusoft IntelliRay Technology, Shenyang 110000, Liaoning Province, China
| | - Dong-Hui Han
- Department of Artificial Intelligence and Algorithm R and D, Neusoft IntelliRay Technology, Shenyang 110000, Liaoning Province, China
| | - Yao Liu
- Department of Artificial Intelligence and Algorithm R and D, Neusoft IntelliRay Technology, Shenyang 110000, Liaoning Province, China
| | - Qiao Qiao
- Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| |
Collapse
|
3
|
Chen ZZ, Cheng N, Johnson L, Dufresne J, Marshall JG. Ammonium bicarbonate buffer system for DNA hybridization and quantification by LC-ESI-MS/MS. Anal Biochem 2025; 702:115822. [PMID: 40054549 DOI: 10.1016/j.ab.2025.115822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 02/09/2025] [Accepted: 02/23/2025] [Indexed: 03/18/2025]
Abstract
High salt buffer may be used for the UV/VIS or radiometric based detection of trihybrid DNA but would contaminate the electrospray mass spectrometer. Colorimetric DNA hybridization assays with the substrate BCIP/NBT reacted with the alkaline phosphatase-streptavidin (APSA) enzyme conjugate that showed a linear range for HIV DNA from 1 pM to 100 pM DNA in presence of high salt concentrations. Ammonia bicarbonate (AMBIC) or ethanolamine resulted in strong DNA hybridization similar to NaCl but was compatible with specific and sensitive mass spectrometry. Linear and Gaussian analysis of HIV DNA with 10 % error was achieved across the pico Molar range from APSA amplification that converted the substrate AMP to adenosine for detection by monitoring the precursor ion at m/z 268 and plotting the fragment intensity at m/z 136 (m/z 268→ m/z 136) that was linear to 100 fM after log transformation. The novel observation that specific DNA hybridization in NaCl may be substituted with AMBIC permitted the direct analysis of a target DNA in femto molar to pico molar range by enzyme linked mass spectrometric assay (ELiMSA) using as little as 0.1 μL (100 nL) of sample reaction injected on column.
Collapse
Affiliation(s)
- Zhuo Zhen Chen
- Research Analytical Biochemistry Laboratory, Department of Chemistry and Biology, Ryerson University, Canada
| | - Nan Cheng
- Research Analytical Biochemistry Laboratory, Department of Chemistry and Biology, Ryerson University, Canada
| | - Lloyd Johnson
- Research Analytical Biochemistry Laboratory, Department of Chemistry and Biology, Ryerson University, Canada
| | - Jaimie Dufresne
- Research Analytical Biochemistry Laboratory, Department of Chemistry and Biology, Ryerson University, Canada
| | - John G Marshall
- Research Analytical Biochemistry Laboratory, Department of Chemistry and Biology, Ryerson University, Canada.
| |
Collapse
|
4
|
Glovi A, Kalligosfyri PM, Miglione A, Singh S, Iula G, Giordano A, De Laurentiis M, Cinti S. Towards liquid biopsy on chip for Triple Negative Breast Cancer: preliminary results on monitoring circulating miRNA-21 using portable diagnostics. Discov Oncol 2025; 16:1022. [PMID: 40481371 PMCID: PMC12144016 DOI: 10.1007/s12672-025-02846-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 05/28/2025] [Indexed: 06/11/2025] Open
Abstract
Liquid biopsy has emerged as a promising non-invasive, cost-effective and real-time approach for cancer diagnosis and monitoring, allowing for the detection of biomarkers in bodily fluids. Among these, microRNAs (miRNAs) are a valuable choice due to their stability and ability to reflect the tumor's heterogeneity. Concerning triple negative breast cancer (TNBC), an aggressive subtype, several studies have demonstrated consistent upregulation of miRNA-21. Its elevated levels, linked to poor prognosis, make it valuable for early detection, risk stratification, and targeted therapies. While traditional miRNA quantification methods are accurate, they often require complex procedures and skilled personnel, limiting their accessibility in low-resource environments. These challenges can be addressed by electrochemical point-of-care (POC) platforms, inspired by the glucose strip, offering a good alternative by reducing matrix effects, integrating cost-effective and eco-friendly substrates. In this work, miRNA-21 was selectively detected using a complementary DNA probe modified with methylene blue, as a redox mediator, immobilized onto an AuNPs-functionalized, paper-based screen-printed electrode. Significant experimental parameters and sensor's selectivity were carefully evaluated, allowing miRNA-21 detection in both standard solution and human serum with limit of detection (LOD) 1.2 nM and satisfactory repeatability of about 8%. The platform's performance improved tenfold with an external paper-based origami pre-concentration device, enabling pM-level miRNA detection and advancing its potential for real clinical practice applications. The platform is envisioned as a starting point for developing accessible, rapid, cost-effective POC testing, with significant implications for personalized medicine and early TNBC detection.
Collapse
Affiliation(s)
- Alessandra Glovi
- Scuola Superiore Meridionale, University of Naples "Federico II", Naples, Italy
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | | | | | - Sima Singh
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Gabriella Iula
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, USA
- Department of Medical Biotechnologies, University of Siena, 53100, Siena, Italy
| | - Michelino De Laurentiis
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, USA
- Department of Breast and Thoracic Oncology, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Naples, Italy
| | - Stefano Cinti
- Department of Pharmacy, University of Naples Federico II, Naples, Italy.
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, USA.
| |
Collapse
|
5
|
Gong L, Wu L, Zhao S, Xiao S, Chu X, Zhang Y, Li F, Li S, Yang H, Jiang P. Epigenetic regulation of ferroptosis in gastrointestinal cancers (Review). Int J Mol Med 2025; 55:93. [PMID: 40242977 PMCID: PMC12045471 DOI: 10.3892/ijmm.2025.5534] [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: 11/13/2024] [Accepted: 04/03/2025] [Indexed: 04/18/2025] Open
Abstract
Ferroptosis is a type of iron‑dependent cell death characterized by excessive lipid peroxidation and may serve as a potential therapeutic target in cancer treatment. While the mechanisms governing ferroptosis continue to be explored and elucidated, an increasing body of research highlights the significant impact of epigenetic modifications on the sensitivity of cancer cells to ferroptosis. Epigenetic processes, such as DNA methylation, histone modifications and non‑coding RNAs, have been identified as key regulators that modulate the expression of ferroptosis‑related genes. These alterations can either enhance or inhibit the sensitivity of gastrointestinal cancer (GIC) cells to ferroptosis, thereby affecting the fate of GICs. Drugs that target epigenetic markers for advanced‑stage cancer have shown promising results in enhancing ferroptosis and inhibiting tumor growth. This review explores the intricate relationship between epigenetic regulation and ferroptosis in GICs. Additionally, the potential of leveraging epigenetic modifications to trigger ferroptosis in GICs is investigated. This review highlights the importance of further research to elucidate the specific mechanisms underlying epigenetic control of ferroptosis and to advance the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Linqiang Gong
- Department of Gastroenterology, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Linlin Wu
- Oncology Department, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Shiyuan Zhao
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, Shandong 272000, P.R. China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, Shandong 272000, P.R. China
| | - Shuai Xiao
- Department of Intensive Care Medicine, Tengzhou Central People's Hospital, Jining Medical University, Tengzhou, Shandong 277500, P.R. China
| | - Xue Chu
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, Shandong 272000, P.R. China
| | - Yazhou Zhang
- Department of Foot and Ankle Surgery, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Fengfeng Li
- Neurosurgery Department, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Shuhui Li
- Department of Gastroenterology, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Hui Yang
- Department of Gynecology, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Pei Jiang
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, Shandong 272000, P.R. China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, Shandong 272000, P.R. China
| |
Collapse
|
6
|
Garofoli M, Maiorano BA, Bruno G, Giordano G, Falagario UG, Necchi A, Carrieri G, Landriscina M, Conteduca V. Circulating Tumor DNA: A New Research Frontier in Urological Oncology from Localized to Metastatic Disease. Eur Urol Oncol 2025; 8:805-817. [PMID: 39627072 DOI: 10.1016/j.euo.2024.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/05/2024] [Accepted: 11/18/2024] [Indexed: 06/01/2025]
Abstract
BACKGROUND AND OBJECTIVE Circulating tumor DNA (ctDNA) testing provides valuable prognostic and predictive information for guiding therapeutic choices and monitoring disease progression and drug resistance for urological tumors. Our review focuses on emerging opportunities for ctDNA analysis in urological tumors and the development of potential circulating biomarkers within a multidisciplinary framework to improve personalized treatment. METHODS A nonsystematic literature review was conducted in the PubMed and MEDLINE databases. Prospective and retrospective peer-reviewed studies, review articles, and research abstracts on the use of ctDNA for urological tumors were included. KEY FINDINGS AND LIMITATIONS Several studies have demonstrated that ctDNA analysis is a promising tool that can help clinicians in the diagnosis and clinical management of urological tumors. In prostate and urothelial cancers, the ctDNA fraction increases proportionally from localized to metastatic disease, indicating a higher tumor burden and more aggressive behavior. Thus, ctDNA seems to be a useful tool for improving prognostic risk stratification and treatment selection. Data on the use of liquid biopsy in renal cell carcinoma are still limited, and assessment of prognostic and predictive biomarkers is a critical unmet need. CONCLUSIONS AND CLINICAL IMPLICATIONS ctDNA analysis promises to revolutionize the management of urological tumors in different disease settings. Integration of ctDNA testing in routine clinical practice will require a multidisciplinary approach that involve patients, clinicians, and molecular biologists. PATIENT SUMMARY We reviewed how testing for tumor DNA in blood (circulating tumor DNA, ctDNA) is used in urological cancers. A great deal of evidence supports the usefulness of this noninvasive test. However, further research via a multidisciplinary approach is needed before ctDNA testing becomes part of routine patient care.
Collapse
Affiliation(s)
- Marianna Garofoli
- Unit of Medical Oncology and Biomolecular Therapy and CREATE Center for Research and Innovation Medicine, Department of Medical and Surgical Sciences, University of Foggia, Policlinico Riuniti, Foggia, Italy
| | | | - Giuseppina Bruno
- Unit of Medical Oncology and Biomolecular Therapy and CREATE Center for Research and Innovation Medicine, Department of Medical and Surgical Sciences, University of Foggia, Policlinico Riuniti, Foggia, Italy
| | - Guido Giordano
- Unit of Medical Oncology and Biomolecular Therapy and CREATE Center for Research and Innovation Medicine, Department of Medical and Surgical Sciences, University of Foggia, Policlinico Riuniti, Foggia, Italy
| | - Ugo Giovanni Falagario
- Unit of Urology and Renal Transplantation, Department of Medical and Surgical Sciences, University of Foggia, Policlinico Riuniti, Foggia, Italy; Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Andrea Necchi
- Department of Medical Oncology, IRCCS San Raffaele Hospital, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Giuseppe Carrieri
- Unit of Urology and Renal Transplantation, Department of Medical and Surgical Sciences, University of Foggia, Policlinico Riuniti, Foggia, Italy
| | - Matteo Landriscina
- Unit of Medical Oncology and Biomolecular Therapy and CREATE Center for Research and Innovation Medicine, Department of Medical and Surgical Sciences, University of Foggia, Policlinico Riuniti, Foggia, Italy.
| | - Vincenza Conteduca
- Unit of Medical Oncology and Biomolecular Therapy and CREATE Center for Research and Innovation Medicine, Department of Medical and Surgical Sciences, University of Foggia, Policlinico Riuniti, Foggia, Italy.
| |
Collapse
|
7
|
Shaikh N, Pandey M, Manna S, Gawde J, Noronha V, Joshi A, Patil VM, Prabhash K, Mahimkar MB. Evaluating the prognostic value of tumor-derived circulating endothelial cells and their progenitors in platinum-refractory oral cancer patients for response to triple metronomic therapy. Oral Oncol 2025; 165:107346. [PMID: 40347575 DOI: 10.1016/j.oraloncology.2025.107346] [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: 11/12/2024] [Revised: 04/10/2025] [Accepted: 04/30/2025] [Indexed: 05/14/2025]
Abstract
OBJECTIVES Metronomic therapy is one of the modalities used to treat OSCC. The present study is the first comprehensive analysis evaluating the potential of circulating endothelial cells (CECs) and their progenitors (CEPs) as prognostic biomarkers in OfSCC patients receiving metronomic therapy. MATERIAL AND METHODS 91 patients were included in the study group. ORR was 42.9 %, while the median OS and PFS were 6.67 months and 4.3 months, respectively. The counts of CECs and CEPs were enumerated by flow cytometry at baseline and follow-up (8, 30, 60, and 90 days after metronomic therapy). RESULTS We correlated the baseline and reduction of CECs after treatment with clinical response, PFS, and OS. Patients with high CEP levels at day 60 showed longer PFS, while patients with low CEC levels at day 30 had better PFS and OS. Change in CEC levels after 8 and 30 days was also correlated with PFS and OS, respectively. The change in CECs 8 days after metronomic therapy as a prognostic biomarker was further validated by multivariate Cox regression. A decline in CECs after 8 days had a favourable impact on OS, while none of the CEP measurements at any time point were statistically associated with OS or PFS. CONCLUSION The reduction of CECs after treatment might serve as a prognostic biomarker, whereas the baseline CEC counts as a predictive or prognostic factor in patients treated with metronomic therapy. The correlation between clinical response and survival further supports the biological relevance of these CECs and CEPs as prognostic biomarkers.
Collapse
Affiliation(s)
- Nishaat Shaikh
- Mahimkar Lab, Cancer Research Institute (CRI), Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC), Kharghar, Navi Mumbai, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
| | - Manish Pandey
- Mahimkar Lab, Cancer Research Institute (CRI), Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC), Kharghar, Navi Mumbai, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
| | - Subhakankha Manna
- Mahimkar Lab, Cancer Research Institute (CRI), Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC), Kharghar, Navi Mumbai, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India
| | - Jitendra Gawde
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India; Biostatistician, Cancer Research Institute (CRI), Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
| | - Vanita Noronha
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India; Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Amit Joshi
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India; Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Vijay M Patil
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India; Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Kumar Prabhash
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India; Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, India.
| | - Manoj B Mahimkar
- Mahimkar Lab, Cancer Research Institute (CRI), Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC), Kharghar, Navi Mumbai, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, India.
| |
Collapse
|
8
|
Chen H, Pang B, Liu Z, Li B, Wang Q, Fan B, Han M, Gong J, Zhou C, Chen Y, Li Y, Jiang J. The Diagnostic Value of Plasma Small Extracellular Vesicle-Derived CAIX Protein in Prostate Cancer and Clinically Significant Prostate Cancer: A Study on Predictive Models. Prostate 2025; 85:723-741. [PMID: 40013658 PMCID: PMC12038087 DOI: 10.1002/pros.24879] [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: 05/18/2024] [Revised: 01/25/2025] [Accepted: 02/14/2025] [Indexed: 02/28/2025]
Abstract
BACKGROUND Current diagnostic tools are inaccurate and not specific to prostate cancer (PCa) diagnosis. Cancer-derived small extracellular vehicles (sEVs) play a key role in intercellular communication. In this study, we examined the diagnostic value of plasma sEV-derived carbonic anhydrase IX (CAIX) protein for PCa and clinically significant prostate cancer (csPCa) diagnosis and avoiding unnecessary biopsies. METHODS Plasma samples (n = 230) were collected from the patients who underwent prostate biopsy with elevated prostate-specific antigen (PSA) levels. sEVs were isolated and characterized, and sEV protein CAIX was measured using an enzyme-linked immunosorbent assay. Independent predictors of csPCa (Gleason score ≥ 7) were identified, and a predictive model was established. A Nomogram for predicting csPCa was developed using data from the training cohort. RESULTS The expression of sEV protein CAIX was significantly higher in both PCa and csPCa compared to benign patients and nonsignificant PCa (nsPCa) (Gleason score < 7, p < 0.001). sEV protein CAIX performed well in distinguishing PCa from benign patients. The predictive model defined by sEV protein CAIX and PSA density (PSAD) demonstrated the highest discriminative ability for csPCa (AUC = 0.895), with diagnostic sensitivity and specificity of 82.5% and 85.8%, respectively. Furthermore, sEV protein CAIX is an effective predictor of 2-year biochemical recurrence (BCR) in PCa patients (p = 0.013), and its high expression is significantly associated with poorer BCR-free survival (p < 0.05). CONCLUSIONS Our findings demonstrate the excellent performance of sEV protein CAIX in PCa and csPCa diagnosis. The Nomogram-based csPCa predictive model incorporating sEV protein CAIX and PSAD exhibits strong predictive value. Additionally, assessing plasma sEV protein CAIX expression levels can further aid in evaluating patient prognosis and provide a basis for making effective treatment decisions.
Collapse
Affiliation(s)
- Haotian Chen
- The First Affiliated Hospital of Ningbo University, Health Science CenterNingbo UniversityNingboZhejiangChina
- Ningbo Clinical Research Center for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Translational Research Laboratory for Urology, Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
| | - Bairen Pang
- The First Affiliated Hospital of Ningbo University, Health Science CenterNingbo UniversityNingboZhejiangChina
- Ningbo Clinical Research Center for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Translational Research Laboratory for Urology, Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Zhejiang Engineering Research Center of Innovative Technologies and Diagnostic and Therapeutic Equipment for Urinary System DiseasesNingboZhejiangChina
| | - Zhihan Liu
- The First Affiliated Hospital of Ningbo University, Health Science CenterNingbo UniversityNingboZhejiangChina
- Ningbo Clinical Research Center for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Translational Research Laboratory for Urology, Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
| | - Benjie Li
- The First Affiliated Hospital of Ningbo University, Health Science CenterNingbo UniversityNingboZhejiangChina
- Ningbo Clinical Research Center for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Translational Research Laboratory for Urology, Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
| | - Qi Wang
- Cancer Care Centre, St George HospitalKogarahNew South WalesAustralia
- St. George and Sutherland Clinical Campuses, School of Clinical Medicine, UNSW SydneyKensingtonNew South WalesAustralia
| | - Baokun Fan
- The First Affiliated Hospital of Ningbo University, Health Science CenterNingbo UniversityNingboZhejiangChina
- Ningbo Clinical Research Center for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Translational Research Laboratory for Urology, Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
| | - Meng Han
- The First Affiliated Hospital of Ningbo University, Health Science CenterNingbo UniversityNingboZhejiangChina
- Ningbo Clinical Research Center for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Translational Research Laboratory for Urology, Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Zhejiang Engineering Research Center of Innovative Technologies and Diagnostic and Therapeutic Equipment for Urinary System DiseasesNingboZhejiangChina
| | - Jie Gong
- The First Affiliated Hospital of Ningbo University, Health Science CenterNingbo UniversityNingboZhejiangChina
- Ningbo Clinical Research Center for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Translational Research Laboratory for Urology, Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
| | - Cheng Zhou
- The First Affiliated Hospital of Ningbo University, Health Science CenterNingbo UniversityNingboZhejiangChina
- Ningbo Clinical Research Center for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Translational Research Laboratory for Urology, Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Zhejiang Engineering Research Center of Innovative Technologies and Diagnostic and Therapeutic Equipment for Urinary System DiseasesNingboZhejiangChina
| | - Yingzhi Chen
- The First Affiliated Hospital of Ningbo University, Health Science CenterNingbo UniversityNingboZhejiangChina
- Ningbo Clinical Research Center for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Translational Research Laboratory for Urology, Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
| | - Yong Li
- Cancer Care Centre, St George HospitalKogarahNew South WalesAustralia
- St. George and Sutherland Clinical Campuses, School of Clinical Medicine, UNSW SydneyKensingtonNew South WalesAustralia
| | - Junhui Jiang
- The First Affiliated Hospital of Ningbo University, Health Science CenterNingbo UniversityNingboZhejiangChina
- Ningbo Clinical Research Center for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Translational Research Laboratory for Urology, Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Zhejiang Engineering Research Center of Innovative Technologies and Diagnostic and Therapeutic Equipment for Urinary System DiseasesNingboZhejiangChina
| |
Collapse
|
9
|
Hussain MS, Moglad E, Goyal A, Rekha MM, Sharma GC, Jayabalan K, Sahoo S, Devi A, Goyal K, Gupta G, Shahwan M, Alzarea SI, Kazmi I. Tumor-educated platelets in lung cancer. Clin Chim Acta 2025; 573:120307. [PMID: 40228574 DOI: 10.1016/j.cca.2025.120307] [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: 03/24/2025] [Revised: 04/10/2025] [Accepted: 04/10/2025] [Indexed: 04/16/2025]
Abstract
Non-invasive diagnostic monitoring techniques have become essential for treating lung cancer (LC), which continues to be the primary cause of cancer-related death worldwide. The new diagnostic biomarkers called tumour-educated platelets (TEPs) show strong prospects for providing vital information about tumor biology, tumor spread pathways, and treatment reaction patterns. Despite lacking a nucleus, platelets exhibit an active RNA profile that develops through interactions with tumor-derived compounds and the tumor microenvironments (TME). This review explains platelet-tumour interaction regulatory mechanisms while focusing on platelet contributions toward cancer development, immune system avoidance, and blood clot formation. The detection and classification of LC show promise through the analysis of RNA molecules extracted from platelets that encompass mRNAs and non-coding RNAs. RNA sequencing technology based on TEP demonstrates excellent diagnostic power by correctly identifying LC patients alongside their oncogenic alterations of EGFR, KRAS, and ALK. Treatment predictions have proven successful using platelet RNA profiles, specifically in immunotherapy and targeted therapy. Integrating next-generation sequencing with machine learning and artificial intelligence enhances TEP-based diagnostic tools, improving detection accuracy. Standardizing platelet extraction methods and vesicle purification from tumor material needs better development for effective and affordable clinical use. Future investigations should combine TEPs with circulating tumor DNA and exosomal RNA markers to enhance both earliest-stage LC diagnosis and patient-specific therapeutic approaches. TEPs introduce a groundbreaking technique in oncology since they can transform non-invasive medical diagnostics and therapeutic monitoring for cancer.
Collapse
Affiliation(s)
- Md Sadique Hussain
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Alkharj 11942, Saudi Arabia
| | - Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - M M Rekha
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Girish Chandra Sharma
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Karthikeyan Jayabalan
- Department of Chemistry, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Samir Sahoo
- Department of General Medicine IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha 751003, India
| | - Anita Devi
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307 Punjab, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India; Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Moyad Shahwan
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al-Jouf 72341, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| |
Collapse
|
10
|
Dhar R, Kumarasamy V, Subramaniyan V. Signature of exosomes in cancer translational medicine. Int J Surg 2025; 111:4138-4139. [PMID: 40265479 PMCID: PMC12165583 DOI: 10.1097/js9.0000000000002413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 04/24/2025]
Affiliation(s)
- Rajib Dhar
- Division of Pharmacology, Faculty of Medical and Life Sciences, Sunway University, Bandar Sunway, Selangor Darul Ehsan, Malaysia
| | - Vinoth Kumarasamy
- Department of Parasitology and Medical Entomology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Vetriselvan Subramaniyan
- Division of Pharmacology, Faculty of Medical and Life Sciences, Sunway University, Bandar Sunway, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
11
|
Lu J, Chen Y, Liu X, Wang J, He Y, Shi T, Chen W, Yan W. Artificial intelligence-driven microRNA signature for early detection of gastric cancer: discovery and clinical functional exploration. Br J Cancer 2025; 132:957-972. [PMID: 40234666 DOI: 10.1038/s41416-025-02984-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 03/01/2025] [Accepted: 03/12/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) is a leading cause of cancer-related deaths worldwide, with late-stage diagnoses frequently leading to poor outcomes. This underscores the need for effective early-stage gastric cancer (ESGC) diagnostics. METHODS We introduce ESGCmiRD, an innovative artificial intelligence-driven strategy that identifies a miRNA signature for ESGC detection by integrating robust expression patterns, ESGC relevance, and regulatory capabilities of microRNA (miRNA) based on multiple networks. Expression and biological roles of miRNAs in GC were validated and explored via bioinformatics analysis and in vitro studies. miRNA-target interaction was confirmed by dual-luciferase reporter assay. Molecular docking predicted miRNA-drug binding affinities, assessing the miRNA signature's therapeutic potential. RESULTS ESGCmiRD identified a blood miRNA signature (miR-320b, miR-222-3p, miR-181a-5p, miR-103a-3p, miR-107) for ESGC detection, demonstrated high diagnostic accuracy with AUC values of 0.986, 0.977, 0.815, and 0.811 in the test and three validation sets (GSE211692, TCGA-STAD, and our cohort), respectively. The five miRNAs were overexpressed in ESGC plasma and directly target PTEN, promoting GC cell proliferation, migration, and invasion. Molecular docking suggested Paclitaxel had the strongest potential interaction with these miRNAs. CONCLUSION This method identifies a robust miRNA signature for ESGC detection and sheds light on gastric carcinogenesis mechanisms, opening doors for potential therapeutic strategies.
Collapse
Affiliation(s)
- Jiachun Lu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuqi Chen
- Department of Gastroenterology, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
| | - Xin Liu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiayu Wang
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuxin He
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Tongguo Shi
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Weichang Chen
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China.
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China.
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China.
| | - Wenying Yan
- School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China.
- Suzhou Key Lab of Multi-modal Data Fusion and Intelligent Healthcare, Suzhou, China.
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Suzhou, China.
| |
Collapse
|
12
|
Malik S, Zaheer S. The impact of liquid biopsy in breast cancer: Redefining the landscape of non-invasive precision oncology. THE JOURNAL OF LIQUID BIOPSY 2025; 8:100299. [PMID: 40521566 PMCID: PMC12166835 DOI: 10.1016/j.jlb.2025.100299] [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: 04/15/2025] [Revised: 05/06/2025] [Accepted: 05/19/2025] [Indexed: 06/18/2025]
Abstract
Breast cancer (BC) remains a leading cause of morbidity and mortality among women worldwide, necessitating the development of innovative diagnostic and monitoring strategies. Liquid biopsy (LB), a minimally invasive approach that analyzes circulating tumor cells (CTCs), cell-free DNA (cfDNA), circulating tumor DNA (ctDNA), extracellular vesicles (EVs), and other tumor-derived biomarkers in body fluids, has emerged as a transformative tool in BC management. This review comprehensively explores the role of LB in early detection, disease monitoring, treatment stratification, and resistance surveillance in BC. We discuss the latest advancements in LB technologies, including next-generation sequencing (NGS), digital PCR, and single-cell analysis, highlighting their sensitivity and specificity. Additionally, we examine the clinical utility of LB in guiding personalized therapy, particularly in the context of hormone receptor-positive, HER2positive, and triple-negative BC subtypes. Despite its promise, several challenges, including standardization, validation, and integration into clinical practice, remain to be addressed. By summarizing current evidence and future directions, this review underscores the potential of LB to revolutionize BC diagnosis and treatment, paving the way for a more precise and dynamic approach to disease management.
Collapse
Affiliation(s)
- Shaivy Malik
- Department and Institution - Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Sufian Zaheer
- Department and Institution - Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| |
Collapse
|
13
|
Papayanis PN, Au C, Jelinek M, Tan A. Using liquid biopsies to guide treatment and monitor response in BRAF V600E positive adenocarcinoma of unknown primary. BMJ Case Rep 2025; 18:e264469. [PMID: 40425210 PMCID: PMC12107294 DOI: 10.1136/bcr-2024-264469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 04/29/2025] [Indexed: 05/29/2025] Open
Abstract
Liquid biopsies using circulating tumour DNA (ctDNA) have emerged as an alternative to conventional biopsies. They can be used to aid in diagnosing and selecting an agent for treatment and can possibly be used to monitor disease response to treatment. In this report, we present a patient who initially presented with lower abdominal pain. Imaging showed extensive retroperitoneal lymphadenopathy and lymph node biopsy demonstrated poorly differentiated carcinoma. Further workup did not reveal a primary lesion, but his genetic analysis revealed a BRAF V600E mutation and CD274 amplification which was used to guide treatment of the adenocarcinoma as a melanoma of unknown primary. He was initiated on ipilimumab and nivolumab and his ctDNA levels showed rapid improvement. After treatment was stopped due to adverse events, he was monitored via ctDNA, with an increase prompting repeat imaging that demonstrated enlargement of his lesions prompting a resumption of treatment.
Collapse
Affiliation(s)
| | - Cherry Au
- Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Michael Jelinek
- Hematology, Oncology and Cellular Therapy, Rush University Medical Center, Chicago, IL, USA
| | - Alan Tan
- Hematology Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
14
|
Naghdloo A, Tessone D, Nagaraju RM, Zhang B, Kang J, Li S, Oberai A, Hicks JB, Kuhn P. Contrastive Representation Learning for Single Cell Phenotyping in Whole Slide Imaging of Enrichment-free Liquid Biopsy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.05.21.655334. [PMID: 40475442 PMCID: PMC12139965 DOI: 10.1101/2025.05.21.655334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/16/2025]
Abstract
Tumor-associated cells derived from a liquid biopsy are promising biomarkers for cancer detection, diagnosis, prognosis, and monitoring. However, their rarity, heterogeneity and plasticity make precise identification and biological characterization challenging for clinical utility. Enrichment-free approaches using whole slide imaging of all circulating cells offer a comprehensive and unbiased strategy for capturing the full spectrum of tumor-associated cell phenotypes. However, current analysis methods often depend on engineered features and manual expert review, making them sensitive to technical variations and subjective biases. These limitations highlight the need for a better feature representation to improve performance and reproducibility of applications in large-scale patient cohort analyses. In this study, we present a deep contrastive learning framework for learning features of all circulating cells, enabling robust identification and stratification of single cells in whole slide immunofluorescence microscopy images. We demonstrate performance of learned features in classification of diverse cell phenotypes in the liquid biopsy, achieving an accuracy of 92.64%. We further demonstrate that learned features improve performance in downstream applications such as outlier detection and clustering. Lastly, our feature representation enables automated identification and enumeration of distinct rare cell phenotypes, achieving average F1-score of 0.93 across cell lines mimicking circulating tumor cells and endothelial cells in contrived samples and average F1-score of 0.858 across CTC phenotypes in clinical samples. This workflow has significant implications for scalable analysis of tumor-associated cellular biomarkers in clinical prognosis and personalized treatment strategies.
Collapse
Affiliation(s)
- Amin Naghdloo
- Convergent Science Institute in Cancer, University of Southern California, Los Angeles, 90089, CA, USA
- Department of Aerospace and Mechanical Engineering, University of Southern California, Los Angeles, 90089, CA, USA
| | - Dean Tessone
- Convergent Science Institute in Cancer, University of Southern California, Los Angeles, 90089, CA, USA
- Department of Biological Sciences, University of Southern California, Los Angeles, 90089, CA, USA
| | - Rajiv M Nagaraju
- Convergent Science Institute in Cancer, University of Southern California, Los Angeles, 90089, CA, USA
| | - Brian Zhang
- Convergent Science Institute in Cancer, University of Southern California, Los Angeles, 90089, CA, USA
| | - Jeffrey Kang
- Convergent Science Institute in Cancer, University of Southern California, Los Angeles, 90089, CA, USA
| | - Shouyi Li
- Convergent Science Institute in Cancer, University of Southern California, Los Angeles, 90089, CA, USA
| | - Assad Oberai
- Convergent Science Institute in Cancer, University of Southern California, Los Angeles, 90089, CA, USA
- Department of Aerospace and Mechanical Engineering, University of Southern California, Los Angeles, 90089, CA, USA
| | - James B Hicks
- Convergent Science Institute in Cancer, University of Southern California, Los Angeles, 90089, CA, USA
- Department of Aerospace and Mechanical Engineering, University of Southern California, Los Angeles, 90089, CA, USA
- USC Norris Comprehensive Cancer, Keck School of Medicine, University of Southern California, Los Angeles, 90033, CA, USA
| | - Peter Kuhn
- Convergent Science Institute in Cancer, University of Southern California, Los Angeles, 90089, CA, USA
- Department of Aerospace and Mechanical Engineering, University of Southern California, Los Angeles, 90089, CA, USA
- Department of Biological Sciences, University of Southern California, Los Angeles, 90089, CA, USA
- USC Norris Comprehensive Cancer, Keck School of Medicine, University of Southern California, Los Angeles, 90033, CA, USA
- Department of Biomedical Engineering, University of Southern California, Los Angeles, 90089, CA, USA
- Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, 90033, CA, USA
| |
Collapse
|
15
|
Englisz A, Smycz-Kubańska M, Królewska-Daszczyńska P, Błaut M, Duszyc A, Mielczarek-Palacz A. The Application of Circulating Tumour DNA (ctDNA) in the Diagnosis, Prognosis, and Treatment Monitoring of Gynaecological and Breast Cancers (Review). Diagnostics (Basel) 2025; 15:1289. [PMID: 40428282 PMCID: PMC12109643 DOI: 10.3390/diagnostics15101289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2025] [Revised: 05/16/2025] [Accepted: 05/19/2025] [Indexed: 05/29/2025] Open
Abstract
Gynaecological cancers, including endometrial, ovarian, and cervical cancers as well as breast cancer, despite numerous studies, still constitute a challenge for modern oncology. For this reason, research aimed at the application of modern diagnostic methods that are useful in early detection, prognosis, and treatment monitoring deserves special attention, Great hopes are currently being placed on the use of liquid biopsy (LB), which examines various tumour components, including cell-free RNA (cfRNA), circulating tumour cells (CTCs), circulating tumour DNA (ctDNA), exosomes, and tumour-educated platelets (TEPs). LB has shown promise as a minimally invasive means of early diagnosis of cancers, detection of recurrence, prediction of therapy response, treatment monitoring, and drug selection. The integration of this test into clinical practice in modern oncology is challenging, but offers many benefits, including reducing the risks associated with invasive procedures, improving diagnostic and therapeutic efficacy, and improving the quality of life of oncology patients. The aim of this review is to present recent reports on the use of ctDNA in diagnosing, predicting the outcome of, and monitoring the treatment of gynaecological and breast cancers.
Collapse
Affiliation(s)
- Aleksandra Englisz
- The Doctoral School, Medical University of Silesia, 40-055 Katowice, Poland;
| | - Marta Smycz-Kubańska
- Department of Immunology and Serology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland; (M.S.-K.); (P.K.-D.); (M.B.); (A.D.)
| | - Patrycja Królewska-Daszczyńska
- Department of Immunology and Serology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland; (M.S.-K.); (P.K.-D.); (M.B.); (A.D.)
| | - Magdalena Błaut
- Department of Immunology and Serology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland; (M.S.-K.); (P.K.-D.); (M.B.); (A.D.)
| | - Agnieszka Duszyc
- Department of Immunology and Serology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland; (M.S.-K.); (P.K.-D.); (M.B.); (A.D.)
| | - Aleksandra Mielczarek-Palacz
- Department of Immunology and Serology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland; (M.S.-K.); (P.K.-D.); (M.B.); (A.D.)
| |
Collapse
|
16
|
Raicevic Toungouz G, Alessandrello R, Giacomini P, Kamal M, Gausachs M, Mazzarella L, Sujobert P, Frigè G, Alay A, Planchon JM, Blau O, Mias Carballal MR, Antoine-Poirel H, Nadal E, Bullinger L, Hebrant A, Servant N, D'Haene N, Aftimos P, Silkenstedt E, Michalík A, Dupain C, Delcourt T, Huet S, Van Campenhout C, Cordero D, Castellano Garcia JM, Codony C, Krol A, Gonzalo J, Van Damme N, Delnord M, Carbone R, Trapani V, Van Garsse S, Van Den Bulcke M. Development and implementation of novel liquid biopsy NGS panels via the OncNGS precommercial procurement (PCP) initiative. ESMO Open 2025; 10:105127. [PMID: 40393376 DOI: 10.1016/j.esmoop.2025.105127] [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: 01/27/2025] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 05/22/2025] Open
Abstract
BACKGROUND Circulating tumor DNA (ctDNA) analysis is transforming oncology, but challenges such as insufficient analytical sensitivity, difficult variant interpretation, suboptimal turnaround time, limited deployment flexibility, and high costs hinder its broader adoption and raise concerns about reimbursement sustainability across European health care systems. MATERIALS AND METHODS To address these challenges, we created the OncNGS consortium, comprising academic, public, and private hospitals (buyers' group) and several supporting entities, to run a European precommercial procurement (PCP) initiative. The consortium defined ctDNA diagnostic testing requirements, conducted an open market consultation, and launched a call for tender. Suppliers were invited to develop an end-to-end, Conformité Européenne In Vitro Diagnostic (CE-IVD)-compliant solution integrating wet laboratory, dry laboratory, and reporting workflow in a single procedure, offering short turnaround time and reasonable cost. RESULTS The OncNGS consortium defined criteria for a versatile, modular, cost-effective solution, deployable centrally or on-site, and adaptable to advancements in precision oncology. Launched in July 2022, the tender attracted seven companies, with four selected for phase I-OncNGS solution(s) design. From these, three advanced to phase II-prototyping. Ultimately, two contractors were awarded contracts for phase III to assess the clinical performance of their prototypes. CONCLUSIONS By leveraging the PCP approach, OncNGS aims to deliver innovative, affordable solutions to standardize ctDNA testing and reporting across European Union countries, improving diagnostic and therapeutic strategies for oncology patients.
Collapse
Affiliation(s)
| | - R Alessandrello
- Innovation Unit, Agency for Health Quality and Assessment of Catalonia (AQuAS), Barcelona, Spain
| | - P Giacomini
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, UOSD Medicina di Precisione in Senologia, Rome, Italy; Alleanza Contro Il Cancro (ACC), Rome, Italy
| | - M Kamal
- Department of Drug Development & Innovation, Institut Curie (IC), Paris, France; IHU PRISM National PRecISion Medicine Center in Oncology, Gustave Roussy, France
| | - M Gausachs
- Institut Català d'Oncologia (ICO), L'Hospitalet de Llobregat, Barcelona, Spain
| | - L Mazzarella
- Alleanza Contro Il Cancro (ACC), Rome, Italy; Department of Experimental Oncology, European Institute of Oncology (IEO)-IRCCS, Milan, Italy
| | - P Sujobert
- Laboratory of Haematology, Hospices Civils de Lyon (HCL), Hôpital Lyon Sud, Lyon, France
| | - G Frigè
- Alleanza Contro Il Cancro (ACC), Rome, Italy; Department of Experimental Oncology, European Institute of Oncology (IEO)-IRCCS, Milan, Italy
| | - A Alay
- Institut Català d'Oncologia (ICO), L'Hospitalet de Llobregat, Barcelona, Spain
| | - J M Planchon
- Department of Genetics, Institut Curie (IC), Paris, France
| | - O Blau
- Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin (HC), Berlin, Germany
| | - M R Mias Carballal
- Innovation Unit, Agency for Health Quality and Assessment of Catalonia (AQuAS), Barcelona, Spain
| | | | - E Nadal
- Institut Català d'Oncologia (ICO), L'Hospitalet de Llobregat, Barcelona, Spain
| | - L Bullinger
- Department of Hematology, Oncology and Tumor Immunology, Charité-Universitätsmedizin Berlin (HC), Berlin, Germany
| | - A Hebrant
- Cancer Center, Sciensano (SC), Brussels, Belgium
| | - N Servant
- Bioinformatics Core Facility, INSERM U1331, Mines Paris Tech, Institut Curie (IC), Paris, France
| | - N D'Haene
- Medical Oncology Department, Institut Jules Bordet (IJB), Hôpital Universitaire de Bruxelles (H.U.B), Brussels, Belgium
| | - P Aftimos
- Medical Oncology Department, Institut Jules Bordet (IJB), Hôpital Universitaire de Bruxelles (H.U.B), Brussels, Belgium
| | - E Silkenstedt
- Department of Medicine III, University Hospital Groβhadern, Ludwig Maximilians University (LMU), Munich, Germany
| | - A Michalík
- De Clercq & Partners (DCP), Sint-Martens-Latem, Belgium
| | - C Dupain
- Department of Drug Development & Innovation, Institut Curie (IC), Paris, France
| | - T Delcourt
- Cancer Center, Sciensano (SC), Brussels, Belgium
| | - S Huet
- Laboratory of Haematology, Hospices Civils de Lyon (HCL), Hôpital Lyon Sud, Lyon, France
| | - C Van Campenhout
- Medical Oncology Department, Institut Jules Bordet (IJB), Hôpital Universitaire de Bruxelles (H.U.B), Brussels, Belgium
| | - D Cordero
- Institut Català d'Oncologia (ICO), L'Hospitalet de Llobregat, Barcelona, Spain
| | - J M Castellano Garcia
- Innovation Unit, Agency for Health Quality and Assessment of Catalonia (AQuAS), Barcelona, Spain
| | - C Codony
- Institut Català d'Oncologia (ICO), L'Hospitalet de Llobregat, Barcelona, Spain
| | - A Krol
- Institut National du Cancer (INCa), Boulogne-Billancourt, France
| | - J Gonzalo
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - N Van Damme
- Belgian Cancer Registry (BCR), Brussels, Belgium
| | - M Delnord
- Cancer Center, Sciensano (SC), Brussels, Belgium
| | - R Carbone
- Alleanza Contro Il Cancro (ACC), Rome, Italy; Department of Experimental Oncology, European Institute of Oncology (IEO)-IRCCS, Milan, Italy
| | - V Trapani
- Alleanza Contro Il Cancro (ACC), Rome, Italy; Saint Camillus International University of Health and Medical Sciences, Rome, Italy
| | | | | |
Collapse
|
17
|
Zhang L, Zhang Y, Chen L, Wang X, Liu Y, Huang Y, Song Y, Zhang Y, Tai J. Research trends and hotspots of circulating tumor DNA in colorectal cancer: a bibliometric study. Front Oncol 2025; 15:1492880. [PMID: 40438683 PMCID: PMC12116327 DOI: 10.3389/fonc.2025.1492880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 04/23/2025] [Indexed: 06/01/2025] Open
Abstract
Introduction Colorectal cancer (CRC) is the third most common cancer worldwide and the second leading cause of cancer-related deaths. The current standard of care for patients with early-stage CRC includes surgical resection and, in selected patients, adjuvant chemotherapy. Circulating tumor DNA (ctDNA) testing is an important component of liquid biopsy, and with the development of testing technology, its value for clinical application has attracted widespread attention. The aim of this study was to help researchers review what has been achieved and better understand the direction of future research through bibliometric analysis. Methods We used the Web of Science Core Collection database to search for ctDNA in CRC-related articles published between 2014 - 2023. Bibliometric analyses of major keywords, authors, countries, institutions, literature and journals in the field were performed using CiteSpace and VOSviewer. Results The number of publications in the field has continued to increase over the last decade. The United States has the highest number of publications, and Italian research scholars have made outstanding contributions. Cancers is the journal with the highest number of publications. Conclusion This study systematically summarizes the research findings in the field of ctDNA in CRC from 2014 to 2023 and describes the research hotspots and trends worldwide that can guide future research.
Collapse
Affiliation(s)
- Lele Zhang
- Department of Colorectal & Anal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Yuzhe Zhang
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, China
| | - Lei Chen
- Department of Colorectal & Anal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Xu Wang
- Department of Colorectal & Anal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Yulian Liu
- Department of Traditional Chinese Medicine, Chongqing Hospital of Jiangsu Province Hospital, Chongqing, China
| | - Yishan Huang
- Department of Colorectal & Anal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Yu Song
- Department of Colorectal & Anal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Ye Zhang
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, China
| | - Jiandong Tai
- Department of Colorectal & Anal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
18
|
Galant N, Grenda A, Krawczyk P, Pięt M, Milanowski J. Liquid biopsy in diagnosis and monitoring of treatment efficacy in patients with small cell lung cancer. Mol Biol Rep 2025; 52:455. [PMID: 40358752 PMCID: PMC12075280 DOI: 10.1007/s11033-025-10569-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 05/02/2025] [Indexed: 05/15/2025]
Abstract
Small-cell lung cancer (SCLC) remains one of the deadliest cancers worldwide. Patients' survival remains poor due to its rapid growth, high metastatic rate and limited possibilities of treatment. For many years, SCLC management has been based mostly on chemo and radiotherapy. However, new therapeutic approaches have been proposed in the past few years, including immunotherapy, which is currently implemented in clinical practice. Unfortunately, in many cases, response to therapy, especially chemotherapy, remains poor, or the patient becomes resistant to initially effective treatment. One of the crucial problems during SCLC patient care is a lack of appropriate predictive biomarkers for various therapeutic approaches. Another critical issue is the scarcity of collected tissue during biopsy, which may be insufficient or of too poor quality for analysis. A liquid biopsy might be the key to solving both of those problems as it is collected in a non-invasive way and enables the measurement of various biomarkers, including circulating tumor DNA (ctDNA) and circulating tumor cells (CTCs). In this review, we discuss various approaches to potentially incorporating liquid biopsy into clinical application - as a companion to imaging during SCLC diagnostics, a new approach to molecular subtyping, and a material enabling predictive or prognostic biomarkers assessment. We also summarize ongoing clinical trials encompassing SCLC patients in which liquid biopsy is collected and examined.
Collapse
Affiliation(s)
- Natalia Galant
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Lublin, Poland.
| | - Anna Grenda
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Lublin, Poland
| | - Paweł Krawczyk
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Lublin, Poland
| | - Mateusz Pięt
- Department of Virology and Immunology, Maria Curie-Skłodowska University, Lublin, Poland
| | - Janusz Milanowski
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
19
|
Lee Y, Park HJ, Lee JS, Jeong HJ, Kim SM, Cho M, Woo W, Dho SH, Kim SM, Kim LK. Epigenomic profiling of papillary thyroid carcinoma reveals distinct subtypes with clinical implications. NPJ Precis Oncol 2025; 9:136. [PMID: 40348904 PMCID: PMC12065858 DOI: 10.1038/s41698-025-00932-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 04/30/2025] [Indexed: 05/14/2025] Open
Abstract
Papillary thyroid carcinoma (PTC) is the most prevalent form of thyroid cancer with generally favorable outcomes. However, surgeons often face challenges regarding optimal surgical timing, extent of surgery, and identifying patients at risk for metastasis or progression to more aggressive subtypes. The ongoing debate over immediate surgery versus active surveillance emphasizes the need for reliable, minimally invasive diagnostic tools to inform surgical decision-making. This study aims to develop an epigenetic biomarker-based prediction system using fine-needle aspiration biopsy (FNAB) samples to assess PTC aggressiveness preoperatively. We conducted a comprehensive analysis of methylome data to identify approximately 7200 CpG islands with altered methylation levels in thyroid cancer tissues. These candidate regions were further examined in our cohort of 55 PTC patients to develop methylation-specific primers suitable for FNAB samples. Methylation patterns allowed us to stratify patients into two distinct prognostic groups, one of which exhibited a poorer survival rate. Our methylation-specific primers effectively classified FNAB samples into these groups, demonstrating their potential as a preoperative tool for assessing tumor aggressiveness. This stratification aids in informing surgical planning and personalizing treatment strategies. DNA methylation profiling of PTC identifies key epigenetic biomarkers associated with tumor aggressiveness. Utilizing these biomarkers in FNAB samples provides a minimally invasive method for preoperative risk assessment, assisting surgeons in tailoring surgical interventions and potentially improving patient outcomes.
Collapse
Affiliation(s)
- Yeongun Lee
- Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyo Jin Park
- Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jin Seok Lee
- Department of Surgery, Thyroid Cancer Center, Gangnam Severance Hospital, Institute of Refractory Thyroid Cancer, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ho Jung Jeong
- Department of Surgery, Thyroid Cancer Center, Gangnam Severance Hospital, Institute of Refractory Thyroid Cancer, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Su Min Kim
- Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Minjeong Cho
- Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Wonjin Woo
- Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - So Hee Dho
- Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seok-Mo Kim
- Department of Surgery, Thyroid Cancer Center, Gangnam Severance Hospital, Institute of Refractory Thyroid Cancer, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Lark Kyun Kim
- Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Division of Biology, Pohang University of Science and Technology, Pohang, Republic of Korea.
| |
Collapse
|
20
|
Li M, Zhang R, Huang X, Jian M, Qi H, Chen X, Deng J, Chen W, Feng C, Dong Q, Su B. Orthogonal DNA self-assembly technology enables rapid and accurate analysis of circulating tumor cells in breast cancer. Anal Chim Acta 2025; 1350:343871. [PMID: 40155170 DOI: 10.1016/j.aca.2025.343871] [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: 01/01/2025] [Revised: 02/22/2025] [Accepted: 02/24/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND As a non-invasive liquid biopsy technology, the detection of circulating tumor cells (CTCs) overcomes the limitations of traditional tissue biopsy methods, enabling continuous sample collection and long-term dynamic monitoring. However, current CTCs analysis methods typically rely on cell size to separate and identify tumor cells, which fails to effectively distinguish tumor cells from different sources. In addition, existing methods are often constrained by limited antibody species, typically detecting only 2-3 molecular phenotypes. This narrow detection scope does not fully capture the heterogeneity of CTCs at the single-cell level, thus limiting its utility in precision diagnosis and personalized treatment. To address these challenges, it is urgent to develop CTCs detection methods that can simultaneously integrate comprehensive target and cell morphology information. RESULTS Using breast cancer as a research model, we developed a computer-aided design-based hybridization chain reaction (CAD-HCR) by combining DNA encoding and antibody coupling technologies with orthogonal DNA self-assembly to achieve multiple detection and heterogeneity analysis of breast cancer mimic samples. This technology overcomes the limitation of antibody species in traditional CTCs detection and utilizes antibody-trigger strand coupling to convert target protein signals into DNA signals, thereby circumventing throughput limitation of existing detection methods. By utilizing the signal amplification effect of DNA self-assembly, this technology enhances sensitivity significantly, allowing for accurate single-cell level detection of CTCs. SIGNIFICANCE This technology provides spatial positioning and cell morphological characteristics information for CTCs analysis of breast cancer, which is expected to provide a more accurate basis for diagnosis and treatment decision-making for in-depth understanding of tumor heterogeneity and clinical applications.
Collapse
Affiliation(s)
- Min Li
- School of Medicine, Anhui University of Science & Technology, Huainan, 232001, China; Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Runchi Zhang
- Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Xiaozhi Huang
- Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Meng Jian
- School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Haipeng Qi
- School of Medicine, Anhui University of Science & Technology, Huainan, 232001, China; Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Xiaolong Chen
- Shanghai Chrona Life Science Technology Co., Ltd., Shanghai, 201306, China
| | - Jie Deng
- Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Wen Chen
- Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Chang Feng
- Shanghai Key Laboratory of Pathogenic Fungi Medical Testing, Shanghai Pudong New Area People's Hospital, Shanghai, 201299, China.
| | - Qiongzhu Dong
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer, Shanghai Municipal Health Commission (SMHC), Minhang Hospital, Fudan University, Shanghai, 200040, China.
| | - Bin Su
- Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China.
| |
Collapse
|
21
|
Martinelli C, Ercoli A, Vizzielli G, Burk SR, Cuomo M, Satasiya V, Kacem H, Braccia S, Mazzarotti G, Miriello I, Tchamou MN, Restaino S, Arcieri M, Poli A, Tius V, Parisi S, Pergolizzi S, Iatì G, Nibali CC, Pizzimenti C, Pepe L, Ieni A, Cortellino S, Giordano A. Liquid biopsy in gynecological cancers: a translational framework from molecular insights to precision oncology and clinical practice. J Exp Clin Cancer Res 2025; 44:140. [PMID: 40340939 PMCID: PMC12060497 DOI: 10.1186/s13046-025-03371-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 03/17/2025] [Indexed: 05/10/2025] Open
Abstract
Liquid biopsy offers a noninvasive method to identify and monitor tumor-derived biomarkers, including circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), exosomes, microRNAs, and tumor-educated platelets, that provide real-time insights into the biological behavior of gynecological cancers. The detection of these markers has the potential to revolutionize cancer management by enabling earlier detection, providing novel data to personalize treatments, and predicting disease recurrence before clinical imaging and predicting disease recurrence before clinical imaging can confirm progression, thereby also guiding complex clinical decision-making. However, because this new "omics" layer introduces additional complexity, it must be fully understood, from its biological rationale to technical development and clinical integration, to prevent confusion or misapplication. That is why, focusing on 14 critical fields of inquiry, our goal is to map the current state of liquid biopsy from bench to bedside while highlighting practical considerations for clinical integration. Each topic integrates recent advances in assay sensitivity, biomarker variability, and data interpretation, underscoring how standardized protocols and robust analytical methods are pivotal for reliable results. We then translate these findings into disease-specific insights, examining how liquid biopsy could refine early detection, minimal residual disease assessment, and therapy guidance in endometrial, cervical, and ovarian cancers. Although several FDA-approved assays and promising commercial tests illustrate the field's rapid evolution, many translational hurdles remain, including the need for harmonized protocols, larger prospective clinical trials, and cost-effectiveness analyses. Crucially, our synthesis clarifies the pivotal role of interdisciplinary collaboration. Oncologists, laboratory scientists, and industry partners must align on standardized procedures and clinically relevant endpoints. Without such coordination, promising biomarkers may remain confined to research settings, limiting their practical benefit. Taken together, our review offers a translational view designed to contextualize liquid biopsy in gynecological oncology.
Collapse
Affiliation(s)
- Canio Martinelli
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, 1900 N 12 St, Philadelphia, PA, 19122, USA
- Department of Human Pathology of Adult and Childhood "Gaetano Barresi", Unit of Obstetrics and Gynecology, University of Messina, Via Consolare Valeria 1, Messina, 98124, Italy
| | - Alfredo Ercoli
- Department of Human Pathology of Adult and Childhood "Gaetano Barresi", Unit of Obstetrics and Gynecology, University of Messina, Via Consolare Valeria 1, Messina, 98124, Italy
| | - Giuseppe Vizzielli
- Clinic of Obstetrics and Gynecology, Santa Maria Della Misericordia" University Hospital, Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy
| | - Sharon Raffaella Burk
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, 1900 N 12 St, Philadelphia, PA, 19122, USA
- Department of Medical Biotechnology, University of Siena, Via Aldo Moro 2, Siena, 53100, Italy
| | - Maria Cuomo
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, 1900 N 12 St, Philadelphia, PA, 19122, USA
- Department of Medical Biotechnology, University of Siena, Via Aldo Moro 2, Siena, 53100, Italy
| | - Vrunda Satasiya
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, 1900 N 12 St, Philadelphia, PA, 19122, USA
- Department of Medical Biotechnology, University of Siena, Via Aldo Moro 2, Siena, 53100, Italy
| | - Housem Kacem
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, 1900 N 12 St, Philadelphia, PA, 19122, USA
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Simone Braccia
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, 1900 N 12 St, Philadelphia, PA, 19122, USA
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano 49, Naples, 80131, Italy
| | - Giulio Mazzarotti
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, 1900 N 12 St, Philadelphia, PA, 19122, USA
- Department of Medical Biotechnology, University of Siena, Via Aldo Moro 2, Siena, 53100, Italy
| | - Irene Miriello
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, 1900 N 12 St, Philadelphia, PA, 19122, USA
| | - Manuela Nana Tchamou
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, 1900 N 12 St, Philadelphia, PA, 19122, USA
- Department of Medical Biotechnology, University of Siena, Via Aldo Moro 2, Siena, 53100, Italy
| | - Stefano Restaino
- Clinic of Obstetrics and Gynecology, Santa Maria Della Misericordia" University Hospital, Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy
| | - Martina Arcieri
- Clinic of Obstetrics and Gynecology, Santa Maria Della Misericordia" University Hospital, Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy
| | - Alice Poli
- Clinic of Obstetrics and Gynecology, Santa Maria Della Misericordia" University Hospital, Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy
| | - Veronica Tius
- Clinic of Obstetrics and Gynecology, Santa Maria Della Misericordia" University Hospital, Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy
| | - Silvana Parisi
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, Messina, 98125, Italy
| | - Stefano Pergolizzi
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, Messina, 98125, Italy
| | - Giuseppe Iatì
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, Messina, 98125, Italy
| | - Chiara Conti Nibali
- Department of Human Pathology of Adult and Childhood "Gaetano Barresi", Unit of Obstetrics and Gynecology, University of Messina, Via Consolare Valeria 1, Messina, 98124, Italy
| | - Cristina Pizzimenti
- Section of Pathological Anatomy, Department of Human Pathology of Adult and Evolutive Age "Gaetano Barresi", G. Martino Hospital, Messina, 98125, Italy
| | - Ludovica Pepe
- Section of Pathological Anatomy, Department of Human Pathology of Adult and Evolutive Age "Gaetano Barresi", G. Martino Hospital, Messina, 98125, Italy
| | - Antonio Ieni
- Section of Pathological Anatomy, Department of Human Pathology of Adult and Evolutive Age "Gaetano Barresi", G. Martino Hospital, Messina, 98125, Italy
| | - Salvatore Cortellino
- Clinical and Translational Oncology, Scuola Superiore Meridionale (SSM), Naples, Italy.
- Laboratory of Molecular Oncology, Research Hospital, Campobasso, 86100, Italy.
- SHRO Italia Foundation ETS, Candiolo, Turin, Italy.
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, 1900 N 12 St, Philadelphia, PA, 19122, USA.
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy.
| |
Collapse
|
22
|
Zhong X, He H, Xiong Y, Sun J, Zeng N, Wang S, Xia Q. A bibliometric analysis of nucleic acid probe and its applications in oncology: towards more precise molecular medicine. Discov Oncol 2025; 16:702. [PMID: 40341658 PMCID: PMC12061834 DOI: 10.1007/s12672-025-02478-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 04/23/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND Nucleic acid probes, which are short sequences of nucleic acids designed to complement specific DNA or RNA targets, have broad applications in biosensing, genetic studies, and various other fields. In tumor diagnosis and treatment, nucleic acid probes offer a precise and accessible approach that is essential for improving patient care and quality of life. Despite substantial research on nucleic acid probes over the past three decades, few comprehensive reviews have retrospectively examined the field. METHODS This study extracted 30 years of nucleic acid probe-related research articles from the Web of Science Core Collection database. We used CiteSpace, VOSviewer, and R tools to systematically analyze the field's current status and developmental trends, with an emphasis on applications in oncology. RESULTS Our findings indicate a continuous growth trend in nucleic acid probe research, with the United States and China, along with their leading institutions and authors, making the most significant contributions. In oncology specifically, nucleic acid probe research has focused primarily on signal amplification, liquid biopsy, and drug delivery. The emergence of novel biomarkers and assay techniques has been a pivotal factor driving advancements in this field. CONCLUSION Nucleic acid probes show strong potential for applications in tumor precise diagnosis and treatment. Continued innovation and closer interdisciplinary collaboration will be vital for further advancements, while large-scale clinical studies are needed to validate their clinical utility.
Collapse
Affiliation(s)
- Xingyu Zhong
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haodong He
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yifan Xiong
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianxuan Sun
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Na Zeng
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shaogang Wang
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Qidong Xia
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
23
|
Poggiana C, Piazza AF, Catoni C, Gallingani I, Piccin L, Pellegrini S, Aneloni V, Salizzato V, Pigozzo J, Fabozzi A, Facchinetti A, Menin C, Del Fiore P, Mocellin S, Chiarion-Sileni V, Rosato A, Scaini MC. A model workflow for microfluidic enrichment and genetic analysis of circulating melanoma cells. Sci Rep 2025; 15:15329. [PMID: 40316673 PMCID: PMC12048555 DOI: 10.1038/s41598-025-99153-y] [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: 11/07/2024] [Accepted: 04/17/2025] [Indexed: 05/04/2025] Open
Abstract
Circulating melanoma cells (CMCs) are responsible for the hematogenous spread of melanoma and, ultimately, metastasis. However, their study has been limited by the low abundance in patient blood and the heterogeneous expression of surface markers. The FDA-approved CellSearch platform enriches CD146-positive CMCs, whose number correlates with progression-free survival and overall survival. However, a single marker may not be sufficient to identify them all. The Parsortix system allows enrichment of CMCs based on their size and deformability, keeping them viable and suitable for downstream molecular analyses. In this study, we tested the strengths, weaknesses and potential convergences of both platforms to integrate the counting of CMCs with a protocol for their genetic analysis. Samples run on Parsortix were labeled with a customized melanoma antibody cocktail, which efficiently labeled and distinguished CMCs from endothelial cells/leukocytes. The capture rate of CellSearch and Parsortix was comparable for cell lines, but Parsortix had a higher capture rate in real-life samples. Moreover, double enrichment with both CellSearch and Parsortix succeeded in removing most of the leukocyte contamination, resulting in an almost pure CMC sample suitable for genetic analysis. In this regard, a proof-of-concept analysis of CMCs from a paradigmatic case of a metastatic uveal melanoma patient led to the identification of multiple genetic alterations. In particular, the GNAQ p.Q209L was identified as homozygous, while a deletion in BAP1 exon 9 was found hemizygous. Moreover, an isochromosome 8 and a homozygous deletion of the CDKN2A gene were detected. In conclusion, we have optimized an approach to successfully enrich and retrieve viable CMCs from metastatic melanoma patients. Moreover, this study provides proof-of-principle for the feasibility of a marker-agnostic CMC enrichment followed by CMC phenotypic identification and genetic analysis.Kindly check and confirm the processed contributed equally is correctly identify We confirm.
Collapse
Affiliation(s)
- Cristina Poggiana
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | | | - Cristina Catoni
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy.
| | - Ilaria Gallingani
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy.
| | - Luisa Piccin
- Medical Oncology 2, Veneto Institute of Oncology, IOV-IRCCS, Padova, Italy
| | - Stefania Pellegrini
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Vittorio Aneloni
- UOC Immunotrasfusionale, University-Hospital of Padova, Padova, Italy
| | | | - Jacopo Pigozzo
- Medical Oncology 2, Veneto Institute of Oncology, IOV-IRCCS, Padova, Italy
| | - Alessio Fabozzi
- Oncology Unit 3, Veneto Institute of Oncology IOV-IRCCS, Padova, 35128, Italy
| | - Antonella Facchinetti
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Chiara Menin
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Paolo Del Fiore
- Soft-Tissue, Peritoneum and Melanoma Surgical Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Simone Mocellin
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- Soft-Tissue, Peritoneum and Melanoma Surgical Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | | | - Antonio Rosato
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Maria Chiara Scaini
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| |
Collapse
|
24
|
de Lima BP, Ferraz LS, Devalle S, Borges HL. Liquid Biopsy-Derived Tumor Biomarkers for Clinical Applications in Glioblastoma. Biomolecules 2025; 15:658. [PMID: 40427551 PMCID: PMC12108610 DOI: 10.3390/biom15050658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 04/25/2025] [Accepted: 04/30/2025] [Indexed: 05/29/2025] Open
Abstract
Glioblastoma (GBM) is the most aggressive primary brain tumor in adults, characterized by rapid growth and resistance to chemotherapy. Conventional treatments remain largely ineffective, with patient survival averaging around 18 months after diagnosis. Current diagnostic methods rely on invasive tissue biopsies and imaging tests. While traditional biopsies involve extracting tissue samples, their routine use is often limited by surgical risks and the challenge of accessing sensitive brain regions. Liquid biopsy has emerged as a promising noninvasive alternative, analyzing circulating tumor components-such as DNA, RNA, extracellular vesicles, and microRNAs-found in body fluids. This approach enables initial diagnosis and continuous disease monitoring, offering a significant advantage over traditional biopsies, which are impractical for frequent repetition during treatment follow-up. This review highlights recent advances in liquid biopsy-derived biomarkers for the clinical management of GBM. The discussion includes the advantages, limitations, and potential of these biomarkers as tools for early diagnosis and disease monitoring. A narrative review of the literature published over the last decade (2014-2024) was conducted using major health-focused scientific databases. The analysis focuses on evaluating the clinical relevance and applicability of liquid biopsy in GBM, offering insights into its potential as a minimally invasive and effective tool for improving glioblastoma management.
Collapse
Affiliation(s)
- Bruna Pereira de Lima
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), Av. Carlos Chagas Filho, 373, Rio de Janeiro 21941–902, RJ, Brazil;
| | - Leticia Silva Ferraz
- Centro de Ciências Naturais e Humanas (CCNH), Universidade Federal do ABC (UFABC), Av. dos Estados, 5001, Santo André 09280-560, SP, Brazil;
| | - Sylvie Devalle
- Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Rua do Rezende, 156-Centro, Rio de Janeiro 20231-092, RJ, Brazil;
| | - Helena Lobo Borges
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), Av. Carlos Chagas Filho, 373, Rio de Janeiro 21941–902, RJ, Brazil;
| |
Collapse
|
25
|
HOU G, YUAN H, LIANG Z, ZHANG L, ZHANG Y. [Exosome separation and enrichment technologies and their applications in disease diagnosis and treatment]. Se Pu 2025; 43:434-445. [PMID: 40331608 PMCID: PMC12059991 DOI: 10.3724/sp.j.1123.2024.09007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Indexed: 05/08/2025] Open
Abstract
Exosomes are nanoscale vesicles wrapped in lipid bilayers that are secreted by cells and carry a variety of proteins, lipids, nucleic acids, and metabolites. Exosomes are widely present in various bodily fluids and mediate intercellular communication. They participate in a variety of physiological and pathological processes, including immune regulation, angiogenesis, tumorigenesis, and metastasis, and have significant clinical diagnosis and treatment potential. Exosomes are source-rich, structurally stable, and reflect the states of their parental cells. Therefore, they are expected to serve as novel diagnostic markers for various diseases. In addition, stem-cell-derived exosomes show therapeutic potential and have the advantages of low immunogenicity, high safety and easy storage, and exhibit therapeutic potential for neurodegenerative disorder, cardiovascular disease, and cancer. Furthermore, exosomes are highly biocompatible, have natural homing properties, and are capable of easily penetrating biological barriers, making them excellent drug-delivery carriers. Isolation and enrichment of exosomes is a prerequisite for downstream analysis and application. High-purity, high-yield, and high-throughput exosome-isolation methods are expected to be used in clinical diagnosis and treatment applications. Based on the physicochemical properties of exosomes, including density, size, charge, and surface composition, exosome-isolation methods are mainly divided into density-based (e.g., differential ultracentrifugation, density-gradient ultracentrifugation), size-based (e.g., ultrafiltration, size-exclusion chromatography, field-flow fractionation), polymer-precipitation (e.g., polyethylene-glycol-based precipitation), and chemical affinity (e.g., antibody-based, aptamer-based, and surface-lipid-based lipid probes) methods. Currently, basic research into exosomes and their clinical applications face a number of challenges. Firstly, the complexity and heterogeneity of exosomes and the lack of standardized isolation methods has led to highly variable research results that hinder comparing and reproducing results between different laboratories and clinical settings. Current isolation methods are generally hindered by insufficient purity, low yield, low throughput, and difficulties separating specific subpopulations, which seriously restrict the development of the exosome field. Secondly, exosome-isolation methods that are easy to use in the clinic, have few technical requirements, and are highly efficient and inexpensive are lacking. Commonly used classical methods, such as ultracentrifugation, are time-consuming, labor-intensive, require large sample volumes, and are inappropriate for clinical settings. Methods such as immunoaffinity can be used to isolate exosomes from precious trace samples in clinical practice; however, high costs, low recoveries, and high operating requirements are shortcomings that restrict sample analysis in the clinic. In addition, robust large-scale methods for preparing exosomes are lacking. There is an urgent need to develop repeatable and scalable methods for preparing batches of high-quality exosomes owing to the rapid development of exosomes for the treatment of clinical diseases. Generally, exosome research progress is expected to greatly improve our understanding of the biological functions and components of exosomes, which will help transform the exosome research into effective diagnostic and therapeutic strategies and lead to new precision-medicine and personalized-treatment applications. This article summarizes the latest progress in exosome-isolation and -enrichment technologies and introduces the application of exosomes as disease diagnostic markers, therapeutic agents, and drug delivery carriers. Finally, the future developmental trends in exosome isolation and enrichment technologies for disease diagnosis and treatment are discussed.
Collapse
|
26
|
Aksamitiene E, Park J, Marjanovic M, Boppart SA. Defining Biological Variability, Analytical Precision and Quantitative Biophysiochemical Characterization of Human Urinary Extracellular Vesicles. J Extracell Vesicles 2025; 14:e70087. [PMID: 40384173 PMCID: PMC12086329 DOI: 10.1002/jev2.70087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 04/18/2025] [Indexed: 05/20/2025] Open
Abstract
The magnitude of combined analytical errors of urinary extracellular vesicle (uEV) preparation and measurement techniques (CVA) has not been thoroughly investigated to determine whether it exceeds biological variations. We utilized technical replicates of human urine to assess the repeatability of uEV concentration and size measurements by nanoparticle tracking analysis (NTA) following differential velocity centrifugation (DC), silicon carbide, or polyethylene glycol uEV isolation methods. The DC method attained the highest precision. Consequently, DC-derived uEV size, most abundant protein levels, and optical redox ratio (ORR) were further assessed by dynamic light scattering (DLS), immunoblotting or multi-photon (SLAM) microscopy. Procedural errors primarily affected uEV counting and uEV-associated protein quantification, while instrumental errors contributed most to the total variability of NTA- and DLS-mediated uEV sizing processes. The intra-individual variability (CVI) of uEV counts assessed by NTA was smaller than inter-individual variability (CVG), resulting in an estimated index of individuality IOI < 0.6, suggesting that personalized reference interval (RI) is more suitable for interpretation of changes in subject's test results. Population-based RI was more appropriate for ORR (IOI > 1.4). The analytical performance of DC-NTA and DC-SLAM techniques met optimal CVA < 0.5 × CVI criteria, indicating their suitability for further testing in clinical laboratory settings.
Collapse
Affiliation(s)
- Edita Aksamitiene
- Beckman Institute for Advanced Science and TechnologyUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
- NIH/NIBIB P41 Center for Label‐Free Imaging and Multiscale BiophotonicsUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| | - Jaena Park
- Beckman Institute for Advanced Science and TechnologyUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
- NIH/NIBIB P41 Center for Label‐Free Imaging and Multiscale BiophotonicsUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
- Department of BioengineeringUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| | - Marina Marjanovic
- Beckman Institute for Advanced Science and TechnologyUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
- NIH/NIBIB P41 Center for Label‐Free Imaging and Multiscale BiophotonicsUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| | - Stephen A. Boppart
- Beckman Institute for Advanced Science and TechnologyUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
- NIH/NIBIB P41 Center for Label‐Free Imaging and Multiscale BiophotonicsUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
- Department of BioengineeringUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
- Department of Electrical and Computer EngineeringUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
- Cancer Center at IllinoisUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
- Carle Illinois College of MedicineUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
- Interdisciplinary Health Sciences InstituteUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| |
Collapse
|
27
|
Dalali S, Kaviani F, Mahdevar M, Oroujalian A, Peymani M. Analysing glycolysis-related genes reveals the prognostic and diagnostic relevance of IER3 and AGRN in colorectal cancer. Genes Genomics 2025; 47:509-520. [PMID: 40048143 DOI: 10.1007/s13258-025-01618-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 01/04/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND Colorectal cancer (CRC) is a significant global health issue, with early detection being critical to improving patient survival. Dysregulation of the glycolysis pathway plays a pivotal role in CRC progression, but specific gene-level mechanisms remain underexplored. OBJECTIVE This study aimed to investigate the role of glycolysis-related genes in CRC development and identify potential diagnostic and prognostic biomarkers. METHODS We utilized The Cancer Genome Atlas (TCGA) dataset to perform differential expression analysis of glycolysis-related genes in CRC. Protein-protein interaction (PPI) network analysis was conducted to identify central hub genes. The diagnostic potential of selected genes was evaluated using ROC curve analysis, while their expression levels were validated through RT-qPCR. RESULTS IER3 and AGRN were identified as significantly upregulated genes associated with reduced survival rates in CRC patients. PPI analysis revealed their roles as central hub genes within the glycolysis pathway. ROC curve analysis demonstrated their ability to distinguish CRC patients from healthy individuals. Validation through RT-qPCR confirmed their significant overexpression in CRC samples, highlighting their involvement in disease progression. CONCLUSION IER3 and AGRN are critical components of the glycolysis pathway, driving CRC development and progression while also showing potential as biomarkers for predicting outcomes, diagnosing CRC, and serving as treatment targets.
Collapse
Affiliation(s)
- Samaneh Dalali
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Fatemeh Kaviani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Mohammad Mahdevar
- Genius Gene, Genetics and Biotechnology Company, Isfahan, Iran
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Andisheh Oroujalian
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Maryam Peymani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran.
| |
Collapse
|
28
|
Sharma R, Yadav J, Bhat SA, Musayev A, Myrzagulova S, Sharma D, Padha N, Saini M, Tuli HS, Singh T. Emerging Trends in Neuroblastoma Diagnosis, Therapeutics, and Research. Mol Neurobiol 2025; 62:6423-6466. [PMID: 39804528 DOI: 10.1007/s12035-024-04680-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 12/20/2024] [Indexed: 03/29/2025]
Abstract
This review explores the current understanding and recent advancements in neuroblastoma, one of the most common extracranial solid pediatric cancers, accounting for ~ 15% of childhood cancer-related mortality. The hallmarks of NBL, including angiogenesis, metastasis, apoptosis resistance, cell cycle dysregulation, drug resistance, and responses to hypoxia and ROS, underscore its complex biology. The tumor microenvironment's significance in disease progression is acknowledged in this study, along with the pivotal role of cancer stem cells in sustaining tumor growth and heterogeneity. A number of molecular signatures are being studied in order to better understand the disease, with many of them serving as targets for the development of new therapeutics. This includes inhibitor therapies for NBL patients, which notably concentrate on ALK signaling, MDM2, PI3K/Akt/mTOR, Wnt, and RAS-MAPK pathways, along with regulators of epigenetic mechanisms. Additionally, this study offers an extensive understanding of the molecular therapies used, such as monoclonal antibodies and CAR-T therapy, focused on both preclinical and clinical studies. Radiation therapy's evolving role and the promise of stem cell transplantation-mediated interventions underscore the dynamic landscape of NBL treatment. This study has also emphasized the recent progress in the field of diagnosis, encompassing the adoption of artificial intelligence and liquid biopsy as a non-intrusive approach for early detection and ongoing monitoring of NBL. Furthermore, the integration of innovative treatment approaches such as CRISPR-Cas9, and cancer stem cell therapy has also been emphasized in this review.
Collapse
Affiliation(s)
- Rishabh Sharma
- Translational Oncology Laboratory, Department of Zoology, Hansraj College, Delhi University, New Delhi, 110007, India
- Amity Stem Cell Institute, Amity Medical School, Amity University, Haryana, 122412, India
| | - Jaya Yadav
- Translational Oncology Laboratory, Department of Zoology, Hansraj College, Delhi University, New Delhi, 110007, India
- Amity Stem Cell Institute, Amity Medical School, Amity University, Haryana, 122412, India
| | - Sajad Ahmad Bhat
- Asfendiyarov Kazakh National Medical University, Almaty, 050000, Kazakhstan
- Department of Biochemistry, NIMS University, Rajasthan, Jaipur, 303121, India
| | - Abdugani Musayev
- Asfendiyarov Kazakh National Medical University, Almaty, 050000, Kazakhstan
| | | | - Deepika Sharma
- Translational Oncology Laboratory, Department of Zoology, Hansraj College, Delhi University, New Delhi, 110007, India
| | - Nipun Padha
- Translational Oncology Laboratory, Department of Zoology, Hansraj College, Delhi University, New Delhi, 110007, India
- Department of Zoology, Cluster University of Jammu, Jammu, 180001, India
| | - Manju Saini
- Translational Oncology Laboratory, Department of Zoology, Hansraj College, Delhi University, New Delhi, 110007, India
- Amity Stem Cell Institute, Amity Medical School, Amity University, Haryana, 122412, India
| | - Hardeep Singh Tuli
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, Haryana, 133207, India
| | - Tejveer Singh
- Translational Oncology Laboratory, Department of Zoology, Hansraj College, Delhi University, New Delhi, 110007, India.
- Division of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences, (INMAS-DRDO), New Delhi, Delhi, 110054, India.
| |
Collapse
|
29
|
Atia GA, Abdal Dayem A, Taher ES, Alghonemy WY, Cho SG, Aldarmahi AA, Haque MA, Alshambky A, Taymour N, Ibrahim AM, Zaghamir DE, Elmorsy EM, Hetta HF, Mohamed ME, Abass KS, Khanday S, Abdeen A. Urine-derived stem cells: a sustainable resource for advancing personalized medicine and dental regeneration. Front Bioeng Biotechnol 2025; 13:1571066. [PMID: 40357329 PMCID: PMC12066649 DOI: 10.3389/fbioe.2025.1571066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
Urine-based therapy, an ancient practice, has been utilized across numerous civilizations to address a wide range of ailments. Urine was considered a priceless resource in numerous traditional therapeutic applications due to its reported medicinal capabilities. While the utilization of urine treatment is contentious and lacks significant support from modern healthcare, the discovery of urine-derived stem cells (UDSCs) has introduced a promising avenue for cell-based therapy. UDSCs offer a noninvasive and easily repeatable collection method, making them a practical and viable option for therapeutic applications. Research has shown that UDSCs contribute to organ preservation by promoting revascularization and decreasing inflammatory reactions in many diseases and conditions. This review will outline the contemporary status of UDSCs research and explore their potential applications in both fundamental science and medical practice.
Collapse
Affiliation(s)
- Gamal A. Atia
- Department of Oral Medicine, Periodontology, and Diagnosis, Faculty of Dentistry, Suez Canal University, Ismailia, Egypt
| | - Ahmed Abdal Dayem
- Department of Stem Cell and Regenerative Biotechnology, School of Advanced Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, and Institute of Health, Aging & Society, Konkuk University, Seoul, Republic of Korea
| | - Ehab S. Taher
- Department of Basic and Clinical Medical Sciences, Faculty of Dentistry, Zarqa University, Zarqa, Jordan
| | - Wafaa Y. Alghonemy
- Department of Basic and Clinical Medical Sciences, Faculty of Dentistry, Zarqa University, Zarqa, Jordan
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, School of Advanced Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, and Institute of Health, Aging & Society, Konkuk University, Seoul, Republic of Korea
- R&D Team, StemExOne Co., Ltd., Seoul, Republic of Korea
| | - Ahmed A. Aldarmahi
- Department of Basic Science, College of Science and Health Professions, King Saud bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
- National Guard- Health Affairs, King Abdullah International Medical Research Centre, Jeddah, Saudi Arabia
| | - Md Azizul Haque
- Department of Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea
| | - Abeer Alshambky
- Molecular Therapeutics Program, Fox Chase Cancer Center, Temple University, Philadelphia, PA, United States
- Department of Biochemistry, Animal Health Research Institute, Cairo, Egypt
| | - Noha Taymour
- Department of Substitutive Dental Sciences, College of Dentistry, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Ateya M. Ibrahim
- College of Nursing, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Donia E. Zaghamir
- College of Nursing, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Ekramy M. Elmorsy
- Center for Health Research, Northern Border University, Arar, Saudi Arabia
| | - Helal F. Hetta
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Mohamed E. Mohamed
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh, Saudi Arabia
| | - Kasim S. Abass
- Department of Physiology, Biochemistry, and Pharmacology, College of Veterinary Medicine, University of Kirkuk, Kirkuk, Iraq
| | - Shifan Khanday
- Department of Biomedical Sciences, Dubai Medical College for Girls, Dubai Medical University, Dubai, United Arab Emirates
| | - Ahmed Abdeen
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Benha University, Toukh, Egypt
| |
Collapse
|
30
|
Massaro G, Venturini J, Rossini D, Vannini A, Brugia M, Lavacchi D, Conticello C, Valle I, Ravizza D, Pillozzi S, Antonuzzo L. Beneath the surface of colorectal cancer: Unmasking the evolving nature of (Neo)RAS. Crit Rev Oncol Hematol 2025; 211:104746. [PMID: 40294875 DOI: 10.1016/j.critrevonc.2025.104746] [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: 10/26/2024] [Revised: 04/23/2025] [Accepted: 04/24/2025] [Indexed: 04/30/2025] Open
Abstract
Metastatic colorectal cancer (mCRC) remains a major clinical challenge, despite therapeutic advancements. Mutations in KRAS and NRAS (RAS) oncogenes drive resistance to anti-EGFR drugs, necessitating RAS mutational analysis prior to treatment. While tissue biopsy remains the gold standard for molecular profiling, it has limitations such as invasiveness, intra-tumoral heterogeneity, and delayed results. Liquid biopsy (LB), on the other hand, offers a non-invasive alternative by analyzing circulating tumor DNA (ctDNA) and it provides a dynamic view of molecular changes over time. Indeed, ctDNA analysis has expanded the understanding of the mCRC's molecular landscape, revealing that RAS mutated (MT) subclones undergo both a positive and a negative selection during treatment. This negative selection has been described as the "NeoRAS WT phenomenon." The temporary disappearance of RAS mutations opens "RAS WT windows," thus making potential candidates for anti-EGFR therapies even patients initially diagnosed as RAS MT. This review examines numerous studies investigating the clinical significance of the "NeoRas WT phenomenon" as a distinct pathological entity. It also highlights the key limitations arising from the variability in study designs, detecting methods and ctDNA shedding rates. The results of ongoing prospective trials are necessary to determine whether NeoRAS WT stands as a reliable marker for guiding anti-EGFR treatment strategies in RAS-mutated patients.
Collapse
Affiliation(s)
- Giulia Massaro
- Clinical Oncology Unit, Careggi University Hospital, Florence, Italy
| | - Jacopo Venturini
- Clinical Oncology Unit, Careggi University Hospital, Florence, Italy
| | - Daniele Rossini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy; Department of Health Science, University of Florence, Florence, Italy.
| | - Agnese Vannini
- Clinical Oncology Unit, Careggi University Hospital, Florence, Italy
| | - Marco Brugia
- Clinical Oncology Unit, Careggi University Hospital, Florence, Italy
| | - Daniele Lavacchi
- Clinical Oncology Unit, Careggi University Hospital, Florence, Italy
| | | | - Irene Valle
- Clinical Oncology Unit, Careggi University Hospital, Florence, Italy
| | - Delia Ravizza
- Clinical Oncology Unit, Careggi University Hospital, Florence, Italy
| | - Serena Pillozzi
- Department of Experimental and Clinical Biomedical Sciences 'Mario Serio', University of Florence, Florence, Italy
| | - Lorenzo Antonuzzo
- Clinical Oncology Unit, Careggi University Hospital, Florence, Italy; Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy; Department of Health Science, University of Florence, Florence, Italy
| |
Collapse
|
31
|
Parums DV. A Review of Circulating Tumor DNA (ctDNA) and the Liquid Biopsy in Cancer Diagnosis, Screening, and Monitoring Treatment Response. Med Sci Monit 2025; 31:e949300. [PMID: 40259565 PMCID: PMC12032849 DOI: 10.12659/msm.949300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2025] [Accepted: 04/08/2025] [Indexed: 04/23/2025] Open
Abstract
The concept of liquid biopsy is based on the knowledge that blood or secretions from the body contain tumor cells, nucleic acids, cellular components, and tumor metabolites. Detection of circulating DNA (ctDNA) in liquid biopsy material shows the most promise due to the advances in DNA technologies that have made detection and sample screening possible. Clinical trials have begun to evaluate ctDNA monitoring for response to cancer treatment in clinical settings for non-small cell lung cancer (NSCLC), breast cancer, and colorectal cancer (CRC). However, most liquid biopsy tests introduced into the clinic have only been able to identify one or two features of tumor DNA, which limits the specificity of the test. In early 2025, a study led by a research team at Oxford University identified a new blood test, TriOx, developed using machine learning to detect multiple types of cancer at an early stage. These new blood tests may revolutionize oncology and make early cancer detection as routine as other diagnostic blood tests, such as blood glucose testing. This article aims to review ctDNA and liquid biopsy in the diagnosis, early detection, and monitoring of treatment response in cancer.
Collapse
|
32
|
Wang X, Che Y, Liu S, Ma B, Liu Y, Lei J, Yuan L, Zhou Y, Ying J, Zhang Y, Tian C, Zhu T, Qi L, Jiang Y, Fang X. Single-Molecule Liquid Biopsy Detects Low- and High-Abundance Protein Markers Simultaneously for Pancreatic Cancer Diagnosis. Anal Chem 2025; 97:8385-8393. [PMID: 40194997 DOI: 10.1021/acs.analchem.4c07031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Simultaneous analysis of multiple biomarkers can typically improve the sensitivity and specificity of a disease diagnosis. Low-abundance serum proteins have recently emerged as a novel class of biomarkers for diseases. Due to the low concentration, the low-abundance protein analysis relies on single-molecule immunoassay, which has a very limited dynamic range. As a result, simultaneous analysis of low- and high-abundance protein markers requires multiple instruments, which demands larger sample volumes and is cost-/labor-consuming. To overcome these limitations, we developed a single-molecule imaging technique that can detect low- and high-abundance protein markers simultaneously in one chip. By employing a hybrid biomarker capture strategy that involves both glass surface and bead immobilization, our method greatly extended the detection range of the single-molecule assay. We used the method for pancreatic cancer diagnosis and analyzed three serum biomarkers of different abundances, including LIF, CA19-9, and CA125. Combined analysis of the three biomarkers yielded exceptional sensitivity and specificity (AUC = 0.996), which is better than using any of the markers alone, including CA19-9 that is used in clinical practice (AUC = 0.804). Overall, we demonstrated a simple and cost-effective method that greatly extended the dynamic range of single-molecule imaging while maintaining the sensitivity, which has great potential in various clinical applications.
Collapse
Affiliation(s)
- Xinjian Wang
- School of Life Sciences, Faculty of Medicine, Tianjin University, Tianjin 300350, P. R. China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P. R. China
| | - Yudong Che
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P. R. China
| | - Songlin Liu
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P. R. China
| | - Bochen Ma
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P. R. China
| | - Yijun Liu
- School of Life Sciences, Faculty of Medicine, Tianjin University, Tianjin 300350, P. R. China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P. R. China
| | - Jiao Lei
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P. R. China
| | - Liang Yuan
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P. R. China
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, P. R. China
| | - Yiwen Zhou
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P. R. China
| | - Jieer Ying
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P. R. China
| | - Yimin Zhang
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P. R. China
| | - Chen Tian
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, P. R. China
| | - Tao Zhu
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P. R. China
| | - Lubin Qi
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P. R. China
| | - Yifei Jiang
- School of Life Sciences, Faculty of Medicine, Tianjin University, Tianjin 300350, P. R. China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P. R. China
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, P. R. China
| | - Xiaohong Fang
- School of Life Sciences, Faculty of Medicine, Tianjin University, Tianjin 300350, P. R. China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P. R. China
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, P. R. China
| |
Collapse
|
33
|
Nell RJ, Versluis M, Menger NV, Gelmi MC, Vu THK, Verdijk RM, Luyten GPM, Jager MJ, van der Velden PA. Digital PCR-based genetic profiling from vitreous fluid as liquid biopsy for primary uveal melanoma: a proof-of-concept study. J Exp Clin Cancer Res 2025; 44:124. [PMID: 40240901 PMCID: PMC12004579 DOI: 10.1186/s13046-025-03374-y] [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: 10/21/2024] [Accepted: 03/25/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Uveal melanoma is an aggressive ocular malignancy. Early molecular characterisation of primary tumours is crucial to identify those at risk of metastatic dissemination. Although tumour biopsies are being taken, liquid biopsies of ocular fluids may form a less invasive but relatively unexplored alternative. In this study, we aim to evaluate the DNA content of vitreous fluid from eyes with a uveal melanoma to obtain molecular tumour information. METHODS DNA was isolated from 65 vitreous fluid samples from enucleated eyes with a uveal melanoma and studied using digital PCR. Primary and additional driver mutations (in GNAQ, GNA11, PLCB4, CYSLTR2, BAP1, SF3B1 and EIF1AX) were investigated using accustomed targeted and drop-off assays. The copy numbers of chromosome 3p and 8q were measured using multiplex and single-nucleotide polymorphism-based assays. Our findings were compared to the molecular profile of matched primary tumours and to the clinicopathological tumour characteristics. RESULTS Almost all (63/65) vitreous fluids had measurable levels of DNA, but melanoma-cell derived DNA (containing the primary driver mutation) was detected in 45/65 samples (median proportion 15.5%, range 0.03-94.4%) and was associated with a larger tumour prominence, but not with any of the molecular tumour subtypes. Among the vitreous fluids with melanoma-cell derived DNA, not all samples harboured (analysable) other mutations or had sufficient statistical power to measure copy numbers. Still, additional mutations in BAP1, SF3B1 and EIF1AX were detected in 15/17 samples and chromosome 3p and 8q copy numbers matched the primary tumour in 19/21 and 18/20 samples, respectively. Collectively, a clinically-relevant molecular classification of the primary tumour could be inferred from 29/65 vitreous fluids. CONCLUSIONS This proof-of-concept study shows that substantial amounts of DNA could be detected in vitreous fluids from uveal melanoma patients, including melanoma-cell derived DNA in 69% of the samples. Prognostically-relevant genetic alterations of the primary tumour could be identified in 45% of the patients. A follow-up study is needed to evaluate our approach in a prospective clinical context. Additionally, our work highlights improved possibilities to sensitively analyse scarce and heterogeneous tumour biopsies, with potential application in other malignancies.
Collapse
Affiliation(s)
- R J Nell
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands.
| | - M Versluis
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands
| | - N V Menger
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands
| | - M C Gelmi
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands
| | - T H K Vu
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands
| | - R M Verdijk
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Pathology, Section Ophthalmic Pathology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - G P M Luyten
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands
| | - M J Jager
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands
| | - P A van der Velden
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
34
|
Selvaggi F, Lopetuso LR, delli Pizzi A, Melchiorre E, Murgiano M, Taraschi AL, Cotellese R, Diana M, Vivarelli M, Mocchegiani F, Catalano T, Aceto GM. Diagnosis of Cholangiocarcinoma: The New Biological and Technological Horizons. Diagnostics (Basel) 2025; 15:1011. [PMID: 40310432 PMCID: PMC12025943 DOI: 10.3390/diagnostics15081011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/30/2025] [Accepted: 04/07/2025] [Indexed: 05/02/2025] Open
Abstract
The diagnosis of cholangiocarcinoma (CCA) remains challenging. Although new technologies have been developed and validated, their routine use in clinical practice is needed. Conventional cytology obtained during endoscopic retrograde cholangiopancreatography-guided brushings is the first-line technique for the diagnosis of CCA, but it has shown limited sensitivity when combined with endoscopic ultrasound-guided biopsy. Other diagnostic tools have been proposed for the diagnosis of CCA, with their respective advantages and limitations. Cholangioscopy with biopsy or cytology combined with FISH analysis, intraductal biliary ultrasound and confocal laser microscopy have made significant advances in the last decade. More recently, developments in the analytical "omics" sciences have allowed the mapping of the microbiota of patients with CCA, and liquid biopsy with proteomic and extracellular vesicle analysis has allowed the identification of new biomarkers that can be incorporated into the predictive diagnostics. Furthermore, in the preoperative setting, radiomics, radiogenomics and the integrated use of artificial intelligence may provide new useful foundations for integrated diagnosis and personalized therapy for hepatobiliary diseases. This review aims to evaluate the current diagnostic approaches and innovative translational research that can be integrated for the diagnosis of CCA.
Collapse
Affiliation(s)
- Federico Selvaggi
- ASL2 Lanciano-Vasto-Chieti, Unit of General Surgery, 66100 Chieti, Italy
- Villa Serena Foundation for Research, 65013 Città Sant’Angelo, Italy; (R.C.); (G.M.A.)
| | - Loris Riccardo Lopetuso
- Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario Gemelli IRCCS, 00136 Roma, Italy; (L.R.L.); (M.M.)
- Dipartimento di Scienze della Vita della Salute e delle Professioni Sanitarie, Università degli Studi Link, 00165 Roma, Italy
| | - Andrea delli Pizzi
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio”, 66100 Chieti, Italy;
- ITAB—Institute for Advanced Biomedical Technologies, University “G. d’Annunzio”, 66100 Chieti, Italy
| | - Eugenia Melchiorre
- University “G. d’Annunzio” Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy;
| | - Marco Murgiano
- Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario Gemelli IRCCS, 00136 Roma, Italy; (L.R.L.); (M.M.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | | | - Roberto Cotellese
- Villa Serena Foundation for Research, 65013 Città Sant’Angelo, Italy; (R.C.); (G.M.A.)
| | - Michele Diana
- Department of Surgery, University Hospital of Geneva, 1205 Geneva, Switzerland;
| | - Marco Vivarelli
- Department of Experimental and Clinical Medicine, Polytechnic University of Marche, 60126 Ancona, Italy; (M.V.); (F.M.)
| | - Federico Mocchegiani
- Department of Experimental and Clinical Medicine, Polytechnic University of Marche, 60126 Ancona, Italy; (M.V.); (F.M.)
| | - Teresa Catalano
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy;
| | - Gitana Maria Aceto
- Villa Serena Foundation for Research, 65013 Città Sant’Angelo, Italy; (R.C.); (G.M.A.)
- Department of Science, University “G. d’Annunzio” Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy
| |
Collapse
|
35
|
Tang R, Luo S, Liu H, Sun Y, Liu M, Li L, Ren H, Angele MK, Börner N, Yu K, Guo Z, Yin G, Luo H. Circulating Tumor Microenvironment in Metastasis. Cancer Res 2025; 85:1354-1367. [PMID: 39992721 PMCID: PMC11997552 DOI: 10.1158/0008-5472.can-24-1241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 11/12/2024] [Accepted: 02/19/2025] [Indexed: 02/26/2025]
Abstract
Activation of invasion and metastasis is a central hallmark of cancer, contributing to the primary cause of death for patients with cancer. In the multistep metastatic process, cancer cells must infiltrate the circulation, survive, arrest at capillary beds, extravasate, and form metastatic clones in distant organs. However, only a small proportion of circulating tumor cells (CTC) successfully form metastases, with transit of CTCs in the circulation being the rate-limiting step. The fate of CTCs is influenced by the circulating tumor microenvironment (cTME), which encompasses factors affecting their biological behaviors in the circulation. This liquid and flowing microenvironment differs significantly from the primary TME or the premetastatic niche. This review summarizes the latest advancements in identifying the biophysical cues, key components, and biological roles of the cTME, highlighting the network among biophysical attributes, blood cells, and nonblood factors in cancer metastasis. In addition to the potential of the cTME as a therapeutic target for inhibiting metastasis, the cTME could also represent as a biomarker for predicting patient outcomes and developing strategies for treating cancer.
Collapse
Affiliation(s)
- Rui Tang
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Shujuan Luo
- Department of Obstetrics, Women and Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Hui Liu
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yan Sun
- Department of Cell Biology and Medical Genetics, Basic Medical School, Chongqing Medical University, Chongqing, China
| | - Manran Liu
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Lu Li
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Haoyu Ren
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Martin K. Angele
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich Munich, Germany
| | - Nikolaus Börner
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich Munich, Germany
| | - Keda Yu
- Department of Breast Surgery, Precision Cancer Medicine Center, Fudan University Shanghai Cancer Center, Shanghai, P.R. China
| | - Zufeng Guo
- Center for Novel Target and Therapeutic Intervention, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Guobing Yin
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Haojun Luo
- Department of Thyroid and Breast Surgery, Renji Hospital, School of Medicine, Chongqing University, Chongqing, China
| |
Collapse
|
36
|
Liu SV, Nagasaka M, Atz J, Solca F, Müllauer L. Oncogenic gene fusions in cancer: from biology to therapy. Signal Transduct Target Ther 2025; 10:111. [PMID: 40223139 PMCID: PMC11994825 DOI: 10.1038/s41392-025-02161-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 12/06/2024] [Accepted: 01/16/2025] [Indexed: 04/15/2025] Open
Abstract
Oncogenic gene fusions occur across a broad range of cancers and are a defining feature of some cancer types. Cancers driven by gene fusion products tend to respond well to targeted therapies, where available; thus, detection of potentially targetable oncogenic fusions is necessary to select optimal treatment. Detection methods include non-sequencing methods, such as fluorescence in situ hybridization and immunohistochemistry, and sequencing methods, such as DNA- and RNA-based next-generation sequencing (NGS). While NGS is an efficient way to analyze multiple genes of interest at once, economic and technical factors may preclude its use in routine care globally, despite several guideline recommendations. The aim of this review is to present a summary of oncogenic gene fusions, with a focus on fusions that affect tyrosine kinase signaling, and to highlight the importance of testing for oncogenic fusions. We present an overview of the identification of oncogenic gene fusions and therapies approved for the treatment of cancers harboring gene fusions, and summarize data regarding treating fusion-positive cancers with no current targeted therapies and clinical studies of fusion-positive cancers. Although treatment options may be limited for patients with rare alterations, healthcare professionals should identify patients most likely to benefit from oncogenic gene fusion testing and initiate the appropriate targeted therapy to achieve optimal treatment outcomes.
Collapse
Affiliation(s)
- Stephen V Liu
- Division of Hematology and Oncology, Georgetown University, Washington, DC, USA.
| | - Misako Nagasaka
- Division of Hematology Oncology, Department of Medicine, University of California Irvine School of Medicine, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, Orange, CA, USA
| | - Judith Atz
- Boehringer Ingelheim International GmbH, Ingelheim am Rhein, Germany
| | - Flavio Solca
- Boehringer Ingelheim RCV GmbH & Co.KG, Vienna, Austria
| | - Leonhard Müllauer
- Department of Pathology, Medical University of Vienna, 1090, Vienna, Austria
| |
Collapse
|
37
|
Zhan J, Cai Y, Cheng P, Zheng L, Pu K. Body fluid diagnostics using activatable optical probes. Chem Soc Rev 2025; 54:3906-3929. [PMID: 40084539 DOI: 10.1039/d4cs01315h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
In vitro diagnostics often detects biomarkers in body fluids (such as blood, urine, sputum, and cerebrospinal fluids) to identify life-threatening diseases at an early stage, monitor overall health, or provide information to help cure, treat, or prevent diseases. Most clinically used optical in vitro diagnostic tests utilize dye-labeled biomolecules for biomarker recognition and signal readout, which typically involve complex steps and long processing times. Activatable optical probes (AOPs), which spontaneously activate their optical signals only in the presence of disease biomarkers, offer higher signal-to-background ratios and improved detection specificity. They also have the potential to simplify detection procedures by eliminating multiple washing steps. In this review, we summarize recent advances in the use of AOPs for pre-clinical and clinical body fluid diagnostics across various diseases, including cancer, nephro-urological disorders, infectious diseases, and digestive diseases. We begin by discussing the molecular design strategies of AOPs to achieve different optical signal readouts and biomarker specificity. We then highlight their diagnostic applications in various disease models and body fluids. Finally, we address the challenges and future perspectives of AOPs in enhancing body fluid diagnostics and advancing precision medicine.
Collapse
Affiliation(s)
- Jie Zhan
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Yanbin Cai
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Penghui Cheng
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 211198, China.
| | - Lei Zheng
- Department of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, 636921, Singapore, Singapore
| |
Collapse
|
38
|
Tataranu LG. Liquid Biopsy as a Diagnostic and Monitoring Tool in Glioblastoma. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:716. [PMID: 40283007 PMCID: PMC12028463 DOI: 10.3390/medicina61040716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Revised: 03/26/2025] [Accepted: 03/31/2025] [Indexed: 04/29/2025]
Abstract
Glioblastoma (GBM) is the most prevalent and aggressive primary central nervous system (CNS) tumor in adults. GBMs exhibit genetic and epigenetic heterogeneity, posing difficulties in surveillance and being associated with high rates of recurrence and mortality. Nevertheless, due to the high infiltrating ability of glioblastoma cells, and regardless of the considerable progress made in radiotherapeutic, chemotherapeutic, and surgical protocols, the treatment of GBM is still inefficient. Conventional diagnostic approaches, such as neuroimaging techniques and tissue biopsies, which are invasive maneuvers, present certain challenges and limitations in providing real-time information, and are incapable of differentiating pseudo-progression related to treatment from real tumor progression. Liquid biopsy, the analysis of biomarkers such as nucleic acids (DNA/RNA), circulating tumor cells (CTCs), extracellular vesicles (EVs), or tumor-educated platelets (TEPs) that are present in body fluids, provides a minimally invasive and dynamic method of diagnosis and continuous monitoring for GBM. It represents a new preferred approach that enables a superior manner to obtain data on possible tumor risk, prognosis, and recurrence assessment. This article is a literature review that aims to provide updated information about GBM biomarkers in body fluids and to analyze their clinical efficiency.
Collapse
Affiliation(s)
- Ligia Gabriela Tataranu
- Department of Neurosurgery, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
- Department of Neurosurgery, Bagdasar-Arseni Emergency Clinical Hospital, 041915 Bucharest, Romania
| |
Collapse
|
39
|
Barata PC, Zarrabi KK, Bex A, Grivas P, Hermann K, Hofman MS, Li R, Lopez-Beltran A, Padani AR, Powles T, Taplin ME, Loriot Y. Novel Methods to Assess Tumor Burden and Minimal Residual Disease in Genitourinary Cancers. Eur Urol 2025; 87:412-423. [PMID: 39638730 DOI: 10.1016/j.eururo.2024.11.011] [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: 07/29/2024] [Revised: 10/23/2024] [Accepted: 11/06/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND AND OBJECTIVE Advances in molecular diagnostics have ushered in a new era for patients with prostate, renal, and urothelial cancers, with novel radiographic and molecular modalities for the assessment of disease burden and minimal residual disease (MRD). Conventional imaging has a limited threshold for disease detection and is often unable to discern clinically occult disease with varying risks of false-negative or false-positive findings depending on the disease state and type of imaging. METHODS We provide an overview of emerging radiographic and molecular tools in development within the genitourinary (GU) disease space. A literature review of contemporary basic, translational, and clinical research studies was performed, covering the timeframe of 1980-2024 through the MEDLINE (via PubMed) and Scopus databases. We highlight select examples of emerging technologies and biomarker-informed clinical trials, which aim to quantify disease at lower thresholds and have the potential for integrating MRD in clinical practice for GU patients. KEY FINDINGS AND LIMITATIONS The development of novel radiotracers, such as prostate-specific membrane antigen or carbonic anhydrase IX, is being evaluated in both clinical practice and trial setting, aiming to change the management of these tumors. Molecular tools including circulating tumor cells and byproducts such as plasma and urine cell-free circulating tumor DNA provide the opportunity for MRD detection. MRD capture on single-cell or cellular byproducts can serve as a conduit for genomic and transcriptomic analyses, providing insight into the molecular underpinnings and clonal evolution of disease. CONCLUSIONS AND CLINICAL IMPLICATIONS While the full potential for MRD applications has yet to be realized, we are witnessing the emergence of novel techniques aimed at MRD detection and the rapid development of elegantly designed studies implementing iterative detection of MRD as means to provide biological rationale and tailor therapeutic options in GU tumors.
Collapse
Affiliation(s)
- Pedro C Barata
- Division of Solid Tumor Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland, OH, USA.
| | - Kevin K Zarrabi
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Axel Bex
- The Royal Free London NHS Foundation Trust, London, UK; UCL Division of Surgery and Interventional Science, University College London, London, UK; Department of Urology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Petros Grivas
- Department of Medicine, Division of Hematology Oncology, University of Washington, Seattle, WA, USA; Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Ken Hermann
- Department of Nuclear Medicine, University of Duisburg-Essen, German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany
| | - Michael S Hofman
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Roger Li
- Department of GU Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Antonio Lopez-Beltran
- Department of Morphological Sciences, Unit of Anatomic Pathology, University of Cordoba Medical School, Cordoba, Spain
| | - Anwar R Padani
- Paul Strickland Scanner Centre, Mount Vernon Cancer Centre, London, UK
| | - Thomas Powles
- Barts Cancer Institute, Experimental Cancer Medicine Centre, Queen Mary University of London, St. Bartholomew's Hospital, London, UK
| | - Mary-Ellen Taplin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Yohann Loriot
- Department of Cancer Medicine and INSERM U981, Université Paris-Sud, Université Paris-Saclay, Gustave Roussy, Villejuif, France
| |
Collapse
|
40
|
Araujo-Abad S, Berna JM, Lloret-Lopez E, López-Cortés A, Saceda M, de Juan Romero C. Exosomes: from basic research to clinical diagnostic and therapeutic applications in cancer. Cell Oncol (Dordr) 2025; 48:269-293. [PMID: 39298081 PMCID: PMC11997007 DOI: 10.1007/s13402-024-00990-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2024] [Indexed: 09/21/2024] Open
Abstract
Cancer continues to pose a global threat despite potent anticancer drugs, often accompanied by undesired side effects. To enhance patient outcomes, sophisticated multifunctional approaches are imperative. Small extracellular vesicles (EVs), a diverse family of naturally occurring vesicles derived from cells, offer advantages over synthetic carriers. Among the EVs, the exosomes are facilitating intercellular communication with minimal toxicity, high biocompatibility, and low immunogenicity. Their tissue-specific targeting ability, mediated by surface molecules, enables precise transport of biomolecules to cancer cells. Here, we explore the potential of exosomes as innovative therapeutic agents, including cancer vaccines, and their clinical relevance as biomarkers for clinical diagnosis. We highlight the cargo possibilities, including nucleic acids and drugs, which make them a good delivery system for targeted cancer treatment and contrast agents for disease monitoring. Other general aspects, sources, and the methodology associated with therapeutic cancer applications are also reviewed. Additionally, the challenges associated with translating exosome-based therapies into clinical practice are discussed, together with the future prospects for this innovative approach.
Collapse
Affiliation(s)
- Salomé Araujo-Abad
- Cancer Research Group, Faculty of Engineering and Applied Sciences, Universidad de Las Américas, Quito, 170124, Ecuador
| | - José Marcos Berna
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, Camí de l'Almazara 11, Elche, Alicante, 03203, Spain
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Avda. Universidad s/n, Ed. Torregaitán, Elche, Alicante, 03202, Spain
| | - Elena Lloret-Lopez
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, Camí de l'Almazara 11, Elche, Alicante, 03203, Spain
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Avda. Universidad s/n, Ed. Torregaitán, Elche, Alicante, 03202, Spain
| | - Andrés López-Cortés
- Cancer Research Group (CRG), Faculty of Medicine, Universidad de Las Américas, Quito, 170124, Ecuador
| | - Miguel Saceda
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, Camí de l'Almazara 11, Elche, Alicante, 03203, Spain
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Avda. Universidad s/n, Ed. Torregaitán, Elche, Alicante, 03202, Spain
| | - Camino de Juan Romero
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, Camí de l'Almazara 11, Elche, Alicante, 03203, Spain.
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Avda. Universidad s/n, Ed. Torregaitán, Elche, Alicante, 03202, Spain.
| |
Collapse
|
41
|
Wu Y, Qiao Y, Yang C, Chen Y, Shen X, Deng C, Yao Q, Sun N. Accelerated Exosomal Metabolic Profiling Enabled by Robust On-Target Array Sintering with Metal-Organic Frameworks. SMALL METHODS 2025; 9:e2401238. [PMID: 39263996 DOI: 10.1002/smtd.202401238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/06/2024] [Indexed: 09/13/2024]
Abstract
Pancreatic cancer is highly lethal, and survival chances improve only with early detection at a precancerous stage. However, there remains a significant gap in developing tools for large-scale, rapid screening. To this end, a high-throughput On-Target Array Extraction Platform (OTAEP) by direct sintering of a series of metal-organic frameworks (MOFs) for dual in situ extraction, encompassing both exosomes and their metabolic profiles, is developed. Based on the principle of geometry-dependent photothermal conversion efficiency and standard testing, the appropriate MOF functional unit is identified. This unit enables exosome enrichment within 10 min and metabolic fingerprint extraction in under 1 s of laser irradiation, with over five reuse. To further accelerate and enhance the quality of metabolic profile analysis, the application of Surrogate Variable Analysis to eliminate hidden confounding factors within the profiles is proposed, and five biomarkers demonstrated by MS/MS experiments are identified. These biomarkers enable early diagnosis, risk stratification, and staging of pancreatic cancer simultaneously, with sensitivity of 94.1%, specificity of 98.8%, and precision of 94.9%. This work represents a breakthrough for overcoming throughput challenges in large-scale testing and for addressing confounding factors in big data analysis.
Collapse
Affiliation(s)
- Yun Wu
- Department of Chemistry, Institutes of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200433, P. R. China
| | - Yiming Qiao
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Chenyu Yang
- Department of Chemistry, Institutes of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200433, P. R. China
| | - Yueying Chen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Xizhong Shen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Chunhui Deng
- Department of Chemistry, Institutes of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200433, P. R. China
| | - Qunyan Yao
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
- Department of Gastroenterology and Hepatology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, 361015, P. R. China
| | - Nianrong Sun
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| |
Collapse
|
42
|
Sigler GI, Murtha J, Varley PR. Diagnostic Advances and Novel Therapeutics in Peritoneal Metastasis. Surg Oncol Clin N Am 2025; 34:173-194. [PMID: 40015798 DOI: 10.1016/j.soc.2024.12.005] [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: 03/01/2025]
Abstract
Appropriate assessment of disease burden in patients with peritoneal surface malignancy (PSM) is critical for treatment decision-making, and conventional cross-sectional imaging (computed tomography and/or MRI) often underestimates burden of disease. Advances in imaging for PSM include novel functional imaging modalities that target cells unique to the tumor microenvironment. Novel alternative methods of diagnosis and disease monitoring are also potentially applicable to management of PSM. These include forms of "liquid biopsy" targeting circulating tumor DNA. Novel regional therapies include both new therapeutic agents (immune-based and nanoparticle-based), as well as new methods of delivery such as pressurized intraperitoneal aerosolized chemotherapy.
Collapse
Affiliation(s)
- Gregory I Sigler
- Division of Surgical Oncology, Department of General Surgery, Complex General Surgical Oncology, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Mail Code 7375, Madison, WI 53792, USA
| | - Jacqueline Murtha
- Department of General Surgery, General Surgery, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Mail Code 7375, Madison, WI 53792, USA
| | - Patrick R Varley
- Division of Surgical Oncology, Department of General Surgery, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Mail Code 7375, Madison, WI 53792, USA; William S. Middleton Memorial Veterans Affairs Hospital, Madison, WI, USA.
| |
Collapse
|
43
|
Sun Y, Puspanathan P, Lim T, Lin D. Advances and challenges in gastric cancer testing: the role of biomarkers. Cancer Biol Med 2025; 22:j.issn.2095-3941.2024.0386. [PMID: 40126094 PMCID: PMC11976707 DOI: 10.20892/j.issn.2095-3941.2024.0386] [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: 12/04/2024] [Accepted: 01/23/2025] [Indexed: 03/25/2025] Open
Abstract
Advances in the identification of molecular biomarkers and the development of targeted therapies have enhanced the prognosis of patients with advanced gastric cancer. Several established biomarkers have been widely integrated into routine clinical diagnostics of gastric cancer to guide personalized treatment. Human epidermal growth factor receptor 2 (HER2) was the first molecular biomarker to be used in gastric cancer with trastuzumab being the first approved targeted therapy for HER2-positive gastric cancer. Programmed death-ligand 1 positivity and microsatellite instability can guide the use of immunotherapies, such as pembrolizumab and nivolumab. More recently, zolbetuximab has been approved for patients with claudin 18.2-positive diseases in some countries. More targeted therapies, including savolitinib for MET-positive patients, are currently under clinical investigation. However, the clinical application of these diagnostic approaches could be hampered by many existing challenges, including invasive and costly sampling methods, variability in immunohistochemistry interpretation, high costs and long turnaround times for next-generation sequencing, the absence of standardized and clinically validated diagnostic cut-off values for some biomarkers, and tumor heterogeneity. Novel testing and analysis techniques, such as artificial intelligence-assisted image analysis and multiplex immunohistochemistry, and emerging therapeutic strategies, including combination therapies that integrate immune checkpoint inhibitors with targeted therapies, offer potential solutions to some of these challenges. This article reviews recent progress in gastric cancer testing, outlines current challenges, and explores future directions for biomarker testing and targeted therapy for gastric cancer.
Collapse
Affiliation(s)
- Yu Sun
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Pathology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | | | - Tony Lim
- Division of Pathology, Singapore General Hospital, Singapore 169608, Singapore
| | - Dongmei Lin
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
44
|
Li Y, Zhang X, Wang J, Wang K, Li B, Qiao X, He W, Cai J, Liu D, Yang LL. Leveraging adenosine triphosphate for cancer theranostics. Theranostics 2025; 15:4708-4733. [PMID: 40225571 PMCID: PMC11984400 DOI: 10.7150/thno.106291] [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: 11/03/2024] [Accepted: 02/20/2025] [Indexed: 04/15/2025] Open
Abstract
Manipulation of the biochemical composition of the tumor microenvironment (TME) is a thriving research area in cancer treatment. Adenosine triphosphate (ATP), a key biochemical component, serves as an energy source for cancer cell proliferation. Notably, ATP can also act as a potent signal transducer to prime anti-tumor immune responses. There is increasing attention given to both the tumor-promoting and tumor-inhibiting roles of ATP in the context of possible new treatments for cancer. ATP levels in the TME are known to be significantly greater than in non-tumor tissues. This disparity presents an opportunity to exploit the ATP response for the delivery of anti-tumor drugs and tumor diagnosis. In this article, we provide a comprehensive overview of the existing strategies and mechanisms for ATP-based therapy and cancer diagnosis. We also discuss the current challenges in the field and propose potential areas for future research, to provide researchers with insights to further investigate the potential of ATP in cancer theranostics.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jinghua Cai
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Danfeng Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Lei-Lei Yang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
45
|
Kotsifa E, Saffioti F, Mavroeidis VK. Cholangiocarcinoma: The era of liquid biopsy. World J Gastroenterol 2025; 31:104170. [PMID: 40124277 PMCID: PMC11924015 DOI: 10.3748/wjg.v31.i11.104170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/28/2025] [Accepted: 02/14/2025] [Indexed: 03/13/2025] Open
Abstract
Cholangiocarcinoma (CCA) is a highly aggressive and heterogeneous malignancy arising from the epithelial cells of the biliary tract. The limitations of the current methods in the diagnosis of CCA highlight the urgent need for new, accurate tools for early cancer detection, better prognostication and patient monitoring. Liquid biopsy (LB) is a modern and non-invasive technique comprising a diverse group of methodologies aiming to detect tumour biomarkers from body fluids. These biomarkers include circulating tumour cells, cell-free DNA, circulating tumour DNA, RNA and extracellular vesicles. The aim of this review is to explore the current and potential future applications of LB in CCA management, with a focus on diagnosis, prognostication and monitoring. We examine both its significant potential and the inevitable limitations associated with this technology. We conclude that LB holds considerable promise, but further research is necessary to fully integrate it into precision oncology for CCA.
Collapse
Affiliation(s)
- Evgenia Kotsifa
- The Second Propaedeutic Department of Surgery, National and Kapodistrian University of Athens, General Hospital of Athens “Laiko”, Athens 11527, Greece
| | - Francesca Saffioti
- Department of Gastroenterology and Hepatology, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, United Kingdom
- University College London Institute for Liver and Digestive Health and Sheila Sherlock Liver Unit, Royal Free Hospital and University College London, London NW3 2QG, United Kingdom
- Division of Clinical and Molecular Hepatology, Department of Clinical and Experimental Medicine, University Hospital of Messina, Messina 98124, Italy
| | - Vasileios K Mavroeidis
- Department of Transplant Surgery, North Bristol NHS Trust, Southmead Hospital, Bristol BS10 5NB, United Kingdom
- Department of Gastrointestinal Surgery, North Bristol NHS Trust, Southmead Hospital, Bristol BS10 5NB, United Kingdom
- Department of HPB Surgery, Bristol Royal Infirmary, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol BS2 8HW, United Kingdom
| |
Collapse
|
46
|
Ghufran SM, Brown ML, Beierle EA. Role of exosomes in diagnosis, prognostication, and treatment of pediatric solid tumors. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200930. [PMID: 39895692 PMCID: PMC11783428 DOI: 10.1016/j.omton.2024.200930] [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] [Indexed: 02/04/2025]
Abstract
Cancer is the second leading cause of death in children, and solid tumors make up 30% of childhood cancers. Molecular profiling of pediatric solid tumors allows a personalized approach to therapy, but this approach mostly relies on surgical biopsy, which is invasive and carries the risk of complications. Liquid biopsy serves as a reliable alternative and a minimally invasive tool for diagnosing, prognosticating, and residual disease monitoring in childhood cancers. This review outlines the potential of exosomes as informative liquid biopsies in pediatric solid tumors. Studies highlighting the potential applications and clinical utility of exosomes and their molecular constituents as prognosticators and therapies in common childhood solid tumors, including neuroblastoma, medulloblastoma, sarcoma, and hepatoblastoma, have been overviewed. We also discuss the limitations and technical challenges of utilizing exosomes for pediatric solid tumors.
Collapse
Affiliation(s)
- Shaikh M. Ghufran
- University of Alabama at Birmingham, Department of Surgery, Division of Pediatric Surgery, Birmingham, AL 35233, USA
| | - Morgan L. Brown
- University of Alabama at Birmingham, Department of Surgery, Division of Pediatric Surgery, Birmingham, AL 35233, USA
| | - Elizabeth A. Beierle
- University of Alabama at Birmingham, Department of Surgery, Division of Pediatric Surgery, Birmingham, AL 35233, USA
| |
Collapse
|
47
|
Zhi Y, Wu J, Li R, Chang X, Liu S, Lu W, Zheng M, Liu B, Chen J, Zhang X, Huang Y. A combination of hepatic leukemia factor and circulating tumor cells serve as effective biomarkers for lung adenocarcinoma prognosis. PeerJ 2025; 13:e19092. [PMID: 40124619 PMCID: PMC11927566 DOI: 10.7717/peerj.19092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 02/11/2025] [Indexed: 03/25/2025] Open
Abstract
Background Lung adenocarcinoma (LUAD) is a highly malignant tumor with the highest mortality rate among all cancers. Early diagnosis and prognosis are important factors in treatment. Hepatic leukemia factor (HLF) is thought to be closely associated with lung cancer metastasis. It is downregulated in lung cancer tissues and negatively correlated with the number of metastasis-activating circulating tumor cells (CTCs) in the peripheral blood of patients. Method and Results In this study, we analyzed data from LUAD samples in TCGA and found that HLF was significantly upregulated in samples with EGFR mutations. Immunohistochemical (IHC) staining of 343 clinical samples also revealed a trend of HLF upregulation in patients with EGFR mutations. EGFR is one of the driver genes in non-small cell lung cancer (NSCLC), and the proportion in LUAD is as high as 50% in the East Asian population. In this study, EGFR mutation was not significantly correlated with the prognosis of LUAD patients and the number of CTC was also not related to EGFR mutation, but was closely related to HLF expression, with more CTCs being captured in the peripheral blood of patients with low expression of HLF (SI ≤ 4). By following up these 343 LUAD patients, high HLF expression (SI > 4) was found to be an independent protective factor for progression-free survival regardless of EGFR status (P < 0.001), whereas high CTC count (> 3) was an independent risk factor for recurrence or death in LUAD patients (P < 0.001). When low HLF and high CTCs coexisted, patients had the shortest median survival time. Patients with low HLF or high CTCs appeared alone had a moderate median survival time. Patients had the longest median survival time when HLF was high and CTCs were low. Conclusion In summary, we believe that HLF expression in cancer tissues and the number of CTCs can be used as effective biomarkers for predicting the prognosis of LUAD, which plays an important role in clinical diagnosis and prognosis judgment.
Collapse
Affiliation(s)
- Yaofeng Zhi
- Clinical Experimental Center, Jiangmen Engineering Technology Research Center of Clinical Biobank and Translational Research, Jiangmen Key Laboratory of Precision and Clinical Translation Medicine, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Jinhua Wu
- Department of Clinical Laboratory, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Ronggang Li
- Department of Pathology, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Xuefei Chang
- Clinical Experimental Center, Jiangmen Engineering Technology Research Center of Clinical Biobank and Translational Research, Jiangmen Key Laboratory of Precision and Clinical Translation Medicine, Jiangmen Central Hospital, Jiangmen, Guangdong, China
- Department of Pulmonary and Critical Care Medicine, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Silin Liu
- Clinical Experimental Center, Jiangmen Engineering Technology Research Center of Clinical Biobank and Translational Research, Jiangmen Key Laboratory of Precision and Clinical Translation Medicine, Jiangmen Central Hospital, Jiangmen, Guangdong, China
- Department of Pulmonary and Critical Care Medicine, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Wenjie Lu
- Clinical Experimental Center, Jiangmen Engineering Technology Research Center of Clinical Biobank and Translational Research, Jiangmen Key Laboratory of Precision and Clinical Translation Medicine, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Mingzhu Zheng
- Clinical Experimental Center, Jiangmen Engineering Technology Research Center of Clinical Biobank and Translational Research, Jiangmen Key Laboratory of Precision and Clinical Translation Medicine, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Baoyi Liu
- Clinical Experimental Center, Jiangmen Engineering Technology Research Center of Clinical Biobank and Translational Research, Jiangmen Key Laboratory of Precision and Clinical Translation Medicine, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Jiarong Chen
- Clinical Experimental Center, Jiangmen Engineering Technology Research Center of Clinical Biobank and Translational Research, Jiangmen Key Laboratory of Precision and Clinical Translation Medicine, Jiangmen Central Hospital, Jiangmen, Guangdong, China
- Department of Oncology, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Xin Zhang
- Clinical Experimental Center, Jiangmen Engineering Technology Research Center of Clinical Biobank and Translational Research, Jiangmen Key Laboratory of Precision and Clinical Translation Medicine, Jiangmen Central Hospital, Jiangmen, Guangdong, China
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong, China
- Collaborative Innovation Center for Antitumor Active Substance Research and Development, Guangdong Medical University, Dongguan, Guangdong, China
| | - Yanming Huang
- Clinical Experimental Center, Jiangmen Engineering Technology Research Center of Clinical Biobank and Translational Research, Jiangmen Key Laboratory of Precision and Clinical Translation Medicine, Jiangmen Central Hospital, Jiangmen, Guangdong, China
- Department of Pulmonary and Critical Care Medicine, Jiangmen Central Hospital, Jiangmen, Guangdong, China
| |
Collapse
|
48
|
Stojchevski R, Sutanto EA, Sutanto R, Hadzi-Petrushev N, Mladenov M, Singh SR, Sinha JK, Ghosh S, Yarlagadda B, Singh KK, Verma P, Sengupta S, Bhaskar R, Avtanski D. Translational Advances in Oncogene and Tumor-Suppressor Gene Research. Cancers (Basel) 2025; 17:1008. [PMID: 40149342 PMCID: PMC11940485 DOI: 10.3390/cancers17061008] [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: 02/10/2025] [Revised: 03/10/2025] [Accepted: 03/15/2025] [Indexed: 03/29/2025] Open
Abstract
Cancer, characterized by the uncontrolled proliferation of cells, is one of the leading causes of death globally, with approximately one in five people developing the disease in their lifetime. While many driver genes were identified decades ago, and most cancers can be classified based on morphology and progression, there is still a significant gap in knowledge about genetic aberrations and nuclear DNA damage. The study of two critical groups of genes-tumor suppressors, which inhibit proliferation and promote apoptosis, and oncogenes, which regulate proliferation and survival-can help to understand the genomic causes behind tumorigenesis, leading to more personalized approaches to diagnosis and treatment. Aberration of tumor suppressors, which undergo two-hit and loss-of-function mutations, and oncogenes, activated forms of proto-oncogenes that experience one-hit and gain-of-function mutations, are responsible for the dysregulation of key signaling pathways that regulate cell division, such as p53, Rb, Ras/Raf/ERK/MAPK, PI3K/AKT, and Wnt/β-catenin. Modern breakthroughs in genomics research, like next-generation sequencing, have provided efficient strategies for mapping unique genomic changes that contribute to tumor heterogeneity. Novel therapeutic approaches have enabled personalized medicine, helping address genetic variability in tumor suppressors and oncogenes. This comprehensive review examines the molecular mechanisms behind tumor-suppressor genes and oncogenes, the key signaling pathways they regulate, epigenetic modifications, tumor heterogeneity, and the drug resistance mechanisms that drive carcinogenesis. Moreover, the review explores the clinical application of sequencing techniques, multiomics, diagnostic procedures, pharmacogenomics, and personalized treatment and prevention options, discussing future directions for emerging technologies.
Collapse
Affiliation(s)
- Radoslav Stojchevski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA;
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Edward Agus Sutanto
- CUNY School of Medicine, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA;
| | - Rinni Sutanto
- New York Institute of Technology College of Osteopathic Medicine, Glen Head, NY 11545, USA;
| | - Nikola Hadzi-Petrushev
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (N.H.-P.)
| | - Mitko Mladenov
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (N.H.-P.)
| | - Sajal Raj Singh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | - Jitendra Kumar Sinha
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | - Shampa Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | | | - Krishna Kumar Singh
- Symbiosis Centre for Information Technology (SCIT), Rajiv Gandhi InfoTech Park, Hinjawadi, Pune 411057, Maharashtra, India;
| | - Prashant Verma
- School of Management, BML Munjal University, NH8, Sidhrawali, Gurugram 122413, Haryana, India
| | - Sonali Sengupta
- Department of Gastroenterology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Dimiter Avtanski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA;
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| |
Collapse
|
49
|
Madhan S, Dhar R, Devi A. Clinical Impact of Exosome Chemistry in Cancer. ACS APPLIED BIO MATERIALS 2025; 8:1862-1876. [PMID: 39936581 DOI: 10.1021/acsabm.4c01920] [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: 02/13/2025]
Abstract
As we progress into the 21st century, cancer stands as one of the most dreaded diseases. With approximately one in every four individuals facing a lifetime risk of developing cancer, cancer remains one of the most serious health challenges worldwide. Its multifaceted nature makes it an arduous and tricky problem to diagnose and treat. Over the years, researchers have explored plenty of approaches and avenues to improve cancer management. One notable strategy includes the study of extracellular vesicles (EVs) as potential biomarkers and therapeutics. Among these EVs, exosomes have emerged as particularly promising candidates due to their unique characteristic properties and functions. They are small membrane-bound vesicles secreted by cells carrying a cargo of biomolecules such as proteins, nucleic acids, and lipids. These vesicles play crucial roles in intercellular communication, facilitating the transfer of biological information between cell-to-cell communication. Exosomes transport cargoes such as DNA, RNA, proteins, and lipids involved in cellular reprogramming and promoting cancer. In this review, we explore the molecular composition of exosomes, significance of exosomes chemistry in cancer development, and its theranostic application as well as exosomes research complications and solutions.
Collapse
Affiliation(s)
- Shrishti Madhan
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu-603203, India
| | - Rajib Dhar
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu-603203, India
| | - Arikketh Devi
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu-603203, India
| |
Collapse
|
50
|
Cox M, Vitello D, Chawla A. Translating the multifaceted use of liquid biopsy to management of early disease in pancreatic adenocarcinoma. Front Oncol 2025; 15:1520717. [PMID: 40182037 PMCID: PMC11966063 DOI: 10.3389/fonc.2025.1520717] [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: 10/31/2024] [Accepted: 02/25/2025] [Indexed: 04/05/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a leading cause of cancer-related mortality, primarily due to late stage at diagnosis. This review examines the multifaceted applications of liquid biopsy and circulating tumor DNA (ctDNA) analysis in the diagnosis and management of PDAC. We review the current literature on the technological advancements in liquid biopsy analysis such as next generation sequencing (NGS) and digital droplet PCR (ddPCR) as well as multi-omics technologies, highlighting their potential for accurate molecular subtyping through ctDNA analysis. This review highlights the significant role of ctDNA in the assessment of tumor behavior, disease subtyping, prediction and monitoring of treatment response, and evaluation of minimal residual disease. We discuss the implications of integrating liquid biopsy techniques into clinical practice as well as its challenges and limitations. By drawing insights from recent studies, this review aims to provide a comprehensive overview of how liquid biopsy and ctDNA analysis can enhance early disease management strategies in PDAC. We underscore the need for additional prospective studies and clinical trials to validate its feasibility and accuracy in order to establish clinical utility, with the ultimate goal of routine incorporation into practice to improve patient outcomes and transform the treatment landscape for PDAC.
Collapse
Affiliation(s)
- Madison Cox
- Division of Surgical Oncology, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Northwestern Medicine Cancer Centers, Northwestern Medicine Regional Medical Group, Winfield, IL, United States
| | - Dominic Vitello
- Division of Surgical Oncology, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Northwestern Quality Improvement, Research and Education in Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Akhil Chawla
- Division of Surgical Oncology, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Northwestern Medicine Cancer Centers, Northwestern Medicine Regional Medical Group, Winfield, IL, United States
- Northwestern Quality Improvement, Research and Education in Surgery, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, United States
| |
Collapse
|