1
|
Wu J, Jiang S, Shen Q, Gong H. Postoperative metastatic Krukenberg tumors with ARID1A and KRAS mutations in a patient with gastric cancer treated with oxaliplatin and tegafur: A case report. Oncol Lett 2025; 29:262. [PMID: 40230423 PMCID: PMC11995681 DOI: 10.3892/ol.2025.15008] [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: 10/13/2024] [Accepted: 03/14/2025] [Indexed: 04/16/2025] Open
Abstract
Krukenberg tumors are a notably rare type of metastatic ovarian malignant tumor, often originating from the stomach. Due to their low incidence rate and the short survival time of patients, there is currently a lack of consensus on the diagnosis and treatment of this disease, as well as a deficiency in genomic analyses and research into the pathogenetic molecular mechanisms. In the present study, the case of a patient with gastric cancer who, 2 years after curative surgery and chemotherapy with oxaliplatin and tegafur, developed recurrent metastatic bilateral Krukenberg tumors with distant metastasis in the ovaries. During treatment, a total hysterectomy and bilateral salpingo-oophorectomy were performed, and intraoperative intraperitoneal chemotherapy with cisplatin (70 mg) was administered. Additionally, ureteroscopy and bilateral ureteral stent placement were conducted transurethrally. Post-surgery, assessments of the genomic alterations and microsatellite instability of the tumor revealed an AT-rich interaction domain 1A (ARID1A) exon c.4720delC mutation and a KRAS exon c.35G>C mutation. The potential pathogenic mechanisms and clinical significance of these mutations were then further discussed. Mutations in the ARID1A gene could increase the sensitivity of the patient to immune checkpoint inhibitor therapy. Additionally, the successful application of KRASG12C inhibitors in other cancer types offers a new approach for the targeted therapy of Krukenberg tumors. Therefore, the present study provides further evidence regarding the genomics of Krukenberg tumors, which may aid in the development of targeted treatment strategies.
Collapse
Affiliation(s)
- Jie Wu
- Department of Gynecology, Dongguan Songshan Lake Tungwah Hospital, Dongguan, Guangdong 523000, P.R. China
| | - Suzhen Jiang
- Department of Gynecology, Dongguan Songshan Lake Tungwah Hospital, Dongguan, Guangdong 523000, P.R. China
| | - Qingling Shen
- Department of Gynecology, Dongguan Songshan Lake Tungwah Hospital, Dongguan, Guangdong 523000, P.R. China
| | - Hongxia Gong
- Department of Gynecology, Dongguan Tungwah Hospital, Dongguan, Guangdong 523000, P.R. China
| |
Collapse
|
2
|
Su H, Gu X, Zhang W, Lin F, Lu X, Zeng X, Wang C, Chen W, Liu W, Tan P, Zou L, Gu B, Chen Q. Identification of Salivary Biomarkers in Colorectal Cancer by Integrating Olink Proteomics and Metabolomics. J Proteome Res 2025; 24:2542-2552. [PMID: 40183281 PMCID: PMC12054530 DOI: 10.1021/acs.jproteome.5c00091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 03/19/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025]
Abstract
Identifying novel biomarkers is crucial for early detection of colorectal cancer (CRC). Saliva, as a noninvasive sample, holds promise for CRC detection. Here, we used Olink proteomics and untargeted metabolomics to analyze saliva samples from CRC patients and healthy controls with the aim of identifying candidate biomarkers in CRC saliva. Univariate and multivariate analyses revealed 16 differentially expressed proteins (DEPs) and 40 differentially accumulated metabolites (DAMs). Pathway enrichment showed DEPs were mainly involved in cancer transcriptional dysregulation, Toll-like receptor signaling, and chemokine signaling. Metabolomics analysis highlighted significant changes in amino acid metabolites, particularly in pathways such as arginine biosynthesis, histidine metabolism, and cysteine and methionine metabolism. Random forest analysis and ELISA validation identified four potential biomarkers: succinate, l-methionine, GZMB, and MMP12. A combined protein-metabolite diagnostic model was developed using logistic regression, achieving an area under the curve of 0.933 (95% CI: 0.871-0.996) for the discovery cohort and 0.969 (95% CI: 0.918-1.000) for the validation cohort, effectively distinguishing CRC patients from healthy individuals. In conclusion, our study identified and validated a panel of noninvasive saliva-based biomarkers that could improve CRC screening and provide new insights into clinical CRC diagnosis.
Collapse
Affiliation(s)
- Hairong Su
- Second
Clinical Medical College, Guangzhou University
of Chinese Medicine, Guangzhou 510120, China
- State
Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University
of Chinese Medicine, Guangzhou 510120, China
| | - Xiangyu Gu
- Second
Clinical Medical College, Guangzhou University
of Chinese Medicine, Guangzhou 510120, China
- State
Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University
of Chinese Medicine, Guangzhou 510120, China
| | - Weizheng Zhang
- Biological
Resource Center, Guangzhou 11th People’s
Hospital, Guangzhou 510530, China
| | - Fengye Lin
- Department
of Clinical Laboratory, Sichuan Taikang
Hospital, Chengdu 610213, China
| | - Xinyi Lu
- Second
Clinical Medical College, Guangzhou University
of Chinese Medicine, Guangzhou 510120, China
- State
Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University
of Chinese Medicine, Guangzhou 510120, China
| | - Xuan Zeng
- Second
Clinical Medical College, Guangzhou University
of Chinese Medicine, Guangzhou 510120, China
- State
Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University
of Chinese Medicine, Guangzhou 510120, China
| | - Chuyang Wang
- Second
Clinical Medical College, Guangzhou University
of Chinese Medicine, Guangzhou 510120, China
- State
Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University
of Chinese Medicine, Guangzhou 510120, China
| | - Weicheng Chen
- Second
Clinical Medical College, Guangzhou University
of Chinese Medicine, Guangzhou 510120, China
- State
Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University
of Chinese Medicine, Guangzhou 510120, China
| | - Wofeng Liu
- Biological
Resource Center, Guangzhou 11th People’s
Hospital, Guangzhou 510530, China
| | - Ping Tan
- Department
of Gastrointestinal Surgery, Guangdong Provincial
Hospital of Chinese Medicine, Guangzhou 510120, China
| | - Liaonan Zou
- Department
of Gastrointestinal Surgery, Guangdong Provincial
Hospital of Chinese Medicine, Guangzhou 510120, China
| | - Bing Gu
- Department
of Clinical Laboratory Medicine, Guangdong Provincial People’s
Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Qubo Chen
- Second
Clinical Medical College, Guangzhou University
of Chinese Medicine, Guangzhou 510120, China
- State
Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University
of Chinese Medicine, Guangzhou 510120, China
| |
Collapse
|
3
|
Mizuno S, Shigeta K, Hara R, Sakamoto K, Nakadai J, Baba H, Kikuchi H, Adachi Y, Shimada T, Suzumura H, Sugiura K, Matsui S, Seishima R, Okabayashi K, Kitagawa Y. Three timepoint perioperative CEA levels are a prognostic factor for recurrence after adjuvant chemotherapy in patients with Stage II and III colorectal cancer. Ann Gastroenterol Surg 2025; 9:496-504. [PMID: 40385331 PMCID: PMC12080205 DOI: 10.1002/ags3.12886] [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: 06/09/2024] [Revised: 10/08/2024] [Accepted: 11/02/2024] [Indexed: 05/20/2025] Open
Abstract
Aim To investigate the relationship between the three timepoint perioperative CEA (ttpCEA) calculated at three timepoints and recurrence during the perioperative period in Stage II and III colorectal cancer (CRC) patients. Methods We performed a multi-institutional retrospective analysis of patients with Stage II and III CRC who underwent surgery and adjuvant chemotherapy from 2010 to 2020. Patient data from three facilities were used as training data, and data from three other facilities were used as validation data. The primary endpoint was the time to recurrence (TTR). Results A total of 538 patients were included for the training data. To validate the feasibility of ttpCEA, 329 patients were included for the validation data. Training data patients were categorized as ttpCEA low (n = 365) and ttpCEA high (n = 173). The 5-y TTR was significantly greater in the ttpCEA-low subgroup than in the ttpCEA-high subgroup (84.3% vs. 69.6%, respectively; p < 0.001). Validation data patients were categorized as ttpCEA low (n = 221) and ttpCEA high (n = 108). The 5-y TTR was significantly greater in the ttpCEA-low subgroup than in the ttpCEA-high subgroup (82.9% vs. 68.7%, respectively; p = 0.003). Conclusion The ttpCEA calculated from perioperative CEA levels at different timepoints was a prognostic factor for recurrence in Stage II and III CRC patients who underwent adjuvant chemotherapy according to both the training and validation data.
Collapse
Affiliation(s)
- Shodai Mizuno
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Kohei Shigeta
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Ryosuke Hara
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Kyoko Sakamoto
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | | | - Hideo Baba
- Department of SurgerySaitama City HospitalSaitamaJapan
| | - Hiroto Kikuchi
- Department of SurgeryHiratsuka City HospitalHiratsukaKanagawaJapan
| | - Yoko Adachi
- Department of SurgeryNational Hospital Organization Tokyo Medical CenterTokyoJapan
| | - Takehiro Shimada
- Department of SurgeryNational Hospital Organization Tokyo Medical CenterTokyoJapan
| | | | | | - Shimpei Matsui
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Ryo Seishima
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Koji Okabayashi
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Yuko Kitagawa
- Department of SurgeryKeio University School of MedicineTokyoJapan
| |
Collapse
|
4
|
Lindgren M, Ljuslinder I, Jonsson P, Nyström H. Type IV collagen, carcinoembryonic antigen, osteopontin, and hepatocyte growth factor as biomarkers for liver metastatic colorectal cancer. Int J Biol Markers 2025:3936155251329590. [PMID: 40289465 DOI: 10.1177/03936155251329590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
IntroductionDiagnosis and monitoring of metastatic colorectal cancer (mCRC) depend on diagnostic imaging. Circulating carcinoembryonic antigen (CEA) can be analyzed but no optimal, non-invasive biomarker exists. Circulating collagen IV (COL IV) is a promising biomarker in patients with colorectal liver metastases (CLM). This study aimed to evaluate COL IV and other cancer-related and stroma-derived proteins as biomarkers for mCRC.Materials & methodsPlasma COL IV and 10 other proteins were analyzed with ELISA and Luminex multiplex assays.ResultsmCRC patients have elevated levels of circulating COL IV, CEA, interleukin-8 (IL-8), hepatocyte growth factor (HGF), cytokeratin-19 fragments (CYFRA 21-1), osteopontin (OPN), and migration inhibitory factor (MIF) compared to primary CRC (pCRC) patients. COL IV is elevated in mCRC patients compared to healthy individuals. Levels of COL IV, CEA, OPN, CYFRA 21-1, IL-8, and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) were dependent on the metastatic site. OPN, CEA, and HGF are very good at discriminating between mCRC patients and pCRC controls. COL IV is very good at distinguishing between mCRC patients and healthy controls. The combination of OPN + CEA is superior at detecting mCRC than CEA alone. High HGF and COL IV levels correlate to poor prognosis.ConclusionOPN, CEA, and HGF are potential biomarkers for mCRC. COL IV is a potential biomarker for CLM. The combination of OPN with CEA is superior to CEA alone in detecting mCRC. Levels of circulating proteins depend on metastatic localization, implying that a combination of markers is better than single markers in detecting mCRC disease. High levels of COL IV and HGF have potential prognostic value.
Collapse
Affiliation(s)
- Moa Lindgren
- Department of Diagnostics and Intervention/Surgery, Umeå University, SE-901 85, Sweden
| | - Ingrid Ljuslinder
- Department of Diagnostics and Intervention/Oncology, Umeå University, SE-901 87, Sweden
| | - Pär Jonsson
- Department of Chemistry, Umeå University, SE-907 36, Sweden
| | - Hanna Nyström
- Department of Diagnostics and Intervention/Surgery, Umeå University, SE-901 85, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, SE-901 87, Sweden
| |
Collapse
|
5
|
Zhou S, Qin Z, Cai S, Ma T, Lin L, Feng L, Gao X, Ma D. Prognostic value of immune infiltration in colorectal cancer: Development of a histopathology-related immunoscore via multiplexed immunohistochemistry. Surgery 2025; 182:109350. [PMID: 40233469 DOI: 10.1016/j.surg.2025.109350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 02/27/2025] [Accepted: 03/10/2025] [Indexed: 04/17/2025]
Abstract
BACKGROUND Our objective was to evaluate the prognostic value of immune infiltration within the intratumoral and peritumoral tissues and to establish a novel histopathology-related immunoscore associated with postoperative colorectal cancer prognosis. METHODS In the tissue microarrays, a total of 104 patients with colorectal cancer were enrolled and randomly assigned to the derivation cohort (n = 61) or the validation cohort (n = 43). Eighteen prognostic immune biomarkers in both intratumoral and peritumoral tissues were examined by the multiplexed immunohistochemistry method, with quantification performed through digital pathology. The histopathology-related immunoscore score was constructed using least absolute shrinkage and selection operator Cox analysis by selected immune features. On the basis of the Cox regression analysis, 3 predictive models were established. Harrell C-statistics were used to assess the performance of those models. RESULTS The area under the curve was 0.743 (confidence interval, 0.457-1.000) in the derivation cohort and 0.739 (confidence interval, 0.538-0.940) in the validation cohort. Subsequently, the groups were classified on the basis of the optimal cutoff value, with the high-risk group exhibiting a poorer prognosis. Furthermore, 3 predictive clinical models were constructed, incorporating the significant risk factors and histopathology-related immunoscore score. The first model incorporating both histopathology-related immunoscore score and statistically significant factors identified through univariate analysis demonstrated superior predictive capability for survival across all 3 models, with an area under the curve of 0.852 and C-index of 0.837. CONCLUSION The histopathology-related immunoscore score offers a novel means of estimating of survival in patients with colorectal cancer. These findings indicated that the immunoscore and the clinical factors might serve as complementary tools to TNM staging to improve the accuracy of patient survival prediction.
Collapse
Affiliation(s)
- Shiqi Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China; Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou, China
| | - Zhaofu Qin
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou, China
| | - Shunv Cai
- Department of Anesthesiology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Ting Ma
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou, China
| | - Luyi Lin
- Department of Radiology, Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Longhai Feng
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou, China
| | - Xinyi Gao
- Department of Radiology, Zhejiang Cancer Hospital, Hangzhou, China.
| | - Dening Ma
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou, China; Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China.
| |
Collapse
|
6
|
Yeo D, Seyfi D, Bastian A, Strauss H, Leach A, Klemm V, Pirrello A, Spring K, Saxena P, Wahlroos S, Sutherland S, Grimison P, Park JS, Sandroussi C, Rasko JE. Portal venous circulating tumor cells as a biomarker for relapse prediction in resected pancreatic cancer. Cell Mol Life Sci 2025; 82:155. [PMID: 40208273 PMCID: PMC11985722 DOI: 10.1007/s00018-025-05669-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 03/16/2025] [Accepted: 03/19/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND Pancreatic cancer is an aggressive disease with poor prognosis. The only potentially curative treatment option is surgical resection, however recurrence is common. Biomarkers to detect minimal residual disease, assist with risk stratification, relapse and real time monitoring, are required. Circulating tumor cells (CTCs) are a promising liquid biopsy biomarker for solid tumors. However, their role in monitoring minimal residual disease in pancreatic cancer remains to be determined. Our study aimed to investigate whether detection and enumeration of CTCs could predict recurrence and provide monitoring of disease status. METHOD Participants planned for Whipple procedure or partial pancreatectomy were enrolled in this prospective pilot study. Intraoperatively, 7.5 mL of portal and peripheral venous blood were collected, and peripheral venous blood was also collected post-surgery. CTC identification and enumeration were performed using the AccuCyte-CyteFinder platform and CellSieve microfiltration. RESULTS Of 29 participants, 20 were confirmed to have epithelial cancer by histopathology, where 15 had pancreatic ductal adenocarcinoma. In those with epithelial cancer, CTCs were detected intraoperatively in 75% of portal venous blood samples, in contrast to 40% detected in peripheral venous blood (median: 6 and 0 per 7.5mL respectively). Only portal venous CTC detection was predictive of pancreatic ductal adenocarcinoma relapse. The positive (> 5) portal venous CTC group had a 6.67 times higher risk of recurring (odds ratio = 20.43, sensitivity = 1.00, specificity = 0.625). Detection of peripheral venous CTCs post-surgery was also correlated with relapse in a small subset of patients. CONCLUSIONS If validated, CTCs may provide a prognostic and monitoring biomarker in patients with pancreatic cancer undergoing surgery.
Collapse
Affiliation(s)
- Dannel Yeo
- Li Ka Shing Cell & Gene Therapy Program, The University of Sydney, Camperdown, Australia
- Precision Oncology Laboratory, Cancer Innovations, Centenary Institute, Camperdown, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
- Cell and Molecular Therapies, Royal Prince Alfred Hospital, Sydney Local Health District, Camperdown, Australia
| | - Doruk Seyfi
- Department of Hepatobiliary and Upper Gastrointestinal Surgery, Royal Prince Alfred Hospital, Sydney Local Health District, Camperdown, Australia
- Upper Gastrointestinal Surgery, Chris O'Brien Lifehouse, Camperdown, Australia
| | - Althea Bastian
- Li Ka Shing Cell & Gene Therapy Program, The University of Sydney, Camperdown, Australia
- Precision Oncology Laboratory, Cancer Innovations, Centenary Institute, Camperdown, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
- Cell and Molecular Therapies, Royal Prince Alfred Hospital, Sydney Local Health District, Camperdown, Australia
| | - Heidi Strauss
- Li Ka Shing Cell & Gene Therapy Program, The University of Sydney, Camperdown, Australia
- Precision Oncology Laboratory, Cancer Innovations, Centenary Institute, Camperdown, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
- Cell and Molecular Therapies, Royal Prince Alfred Hospital, Sydney Local Health District, Camperdown, Australia
| | - Anna Leach
- Li Ka Shing Cell & Gene Therapy Program, The University of Sydney, Camperdown, Australia
- Precision Oncology Laboratory, Cancer Innovations, Centenary Institute, Camperdown, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
- Cell and Molecular Therapies, Royal Prince Alfred Hospital, Sydney Local Health District, Camperdown, Australia
| | - Vera Klemm
- Li Ka Shing Cell & Gene Therapy Program, The University of Sydney, Camperdown, Australia
- Precision Oncology Laboratory, Cancer Innovations, Centenary Institute, Camperdown, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
- Cell and Molecular Therapies, Royal Prince Alfred Hospital, Sydney Local Health District, Camperdown, Australia
| | - Anthony Pirrello
- Li Ka Shing Cell & Gene Therapy Program, The University of Sydney, Camperdown, Australia
- Precision Oncology Laboratory, Cancer Innovations, Centenary Institute, Camperdown, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
| | - Kevin Spring
- Medical Oncology Group, Liverpool Clinical School, School of Medicine, Western Sydney University and Ingham Institute for Applied Medical Research, Liverpool, Australia
- Concord Institute of Academic Surgery, Sydney Local Health District, Concord, Australia
- South-West Sydney Clinical Campuses, UNSW Medicine & Health, Sydney, Australia
| | - Payal Saxena
- Division of Gastroenterology, Department of Medicine, Royal Prince Alfred Hospital, Sydney Local Health District, Camperdown, Australia
- Gastroenterology, Chris O'Brien Lifehouse, Camperdown, Australia
| | - Sara Wahlroos
- Medical Oncology, Chris O'Brien Lifehouse, Camperdown, Australia
| | - Sarah Sutherland
- Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
- Medical Oncology, Chris O'Brien Lifehouse, Camperdown, Australia
| | - Peter Grimison
- Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
- Medical Oncology, Chris O'Brien Lifehouse, Camperdown, Australia
| | - Jin-Soo Park
- Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
- Department of Hepatobiliary and Upper Gastrointestinal Surgery, Royal Prince Alfred Hospital, Sydney Local Health District, Camperdown, Australia
- Upper Gastrointestinal Surgery, Chris O'Brien Lifehouse, Camperdown, Australia
| | - Charbel Sandroussi
- Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
- Department of Hepatobiliary and Upper Gastrointestinal Surgery, Royal Prince Alfred Hospital, Sydney Local Health District, Camperdown, Australia
- Upper Gastrointestinal Surgery, Chris O'Brien Lifehouse, Camperdown, Australia
| | - John Ej Rasko
- Li Ka Shing Cell & Gene Therapy Program, The University of Sydney, Camperdown, Australia.
- Precision Oncology Laboratory, Cancer Innovations, Centenary Institute, Camperdown, Australia.
- Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia.
- Cell and Molecular Therapies, Royal Prince Alfred Hospital, Sydney Local Health District, Camperdown, Australia.
| |
Collapse
|
7
|
Rosu MC, Tarta C, Moldovan S, Neamtu AA, Ardelean A, Capitanio M, Herczeg D, Faur IF, Bende R, Pilat L, Prunoiu VM, Neamtu C, Totolici BD. Integrating TNF-α with Established Tumor Markers to Enhance Prognostic Accuracy in Gastric Cancer: A Prospective Observational Study. Biomedicines 2025; 13:928. [PMID: 40299527 PMCID: PMC12025002 DOI: 10.3390/biomedicines13040928] [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: 02/25/2025] [Revised: 03/26/2025] [Accepted: 03/31/2025] [Indexed: 04/30/2025] Open
Abstract
Background/Objectives: Gastric cancer remains a leading cause of cancer mortality worldwide. Reliable biomarkers are crucial for early detection, prognostication, and therapy monitoring. While classical tumor markers such as carcinoembryonic antigen (CEA), cancer antigen (CA)19-9, CA72-4, and alpha-fetoprotein (AFP) are used in clinical practice, their accuracy can be limited. Tumor necrosis factor alpha (TNF-α) is an inflammatory cytokine implicated in tumor progression, yet its relationship with established gastric cancer tumor markers has not been fully clarified. This study aimed to determine whether elevated TNF-α correlates with key tumor markers and disease stage in gastric cancer. Methods: In this prospective observational study, we enrolled 80 gastric cancer patients and 20 non-neoplastic controls. Baseline clinical data, laboratory parameters, and tumor markers (CEA, CA19-9, CA72-4, AFP) were recorded. TNF-α concentrations were measured using enzyme-linked immunosorbent assays. Correlation analyses and multivariate regression were performed to assess the relationship of TNF-α with tumor markers, inflammatory indices, and disease stage. Results: TNF-α was significantly elevated in gastric cancer patients (median 4.5 pg/mL) compared to controls (2.9 pg/mL). TNF-α showed a robust correlation with CA19-9 (rho = 0.502) and CA72-4 (rho = 0.385), and a moderate correlation with CEA (rho = 0.279). TNF-α concentrations were highest in Stage IV disease and in the intestinal-type histology. In regression analysis, only CA19-9 and CA72-4 remained independent predictors of TNF-α after controlling for clinical confounders. Conclusions: TNF-α is strongly associated with CA19-9 and CA72-4 and rises with advancing stage, highlighting its potential as an adjunct marker for assessing gastric cancer burden. These findings provide a rationale for further research on TNF-α as both a prognostic biomarker and a possible therapeutic target in gastric cancer.
Collapse
Affiliation(s)
- Mihai Catalin Rosu
- Surgery Department, Clinical County Emergency Hospital of Arad, Andrenyi Karoly Str., No. 2-4, 310037 Arad, Romania; (M.C.R.); (A.-A.N.); (A.A.); (D.H.); (C.N.); (B.D.T.)
- Faculty of Medicine, “Vasile Goldis” Western University of Arad, Liviu Rebreanu Str., Nr. 86, 310045 Arad, Romania; (S.M.); (L.P.)
| | - Cristi Tarta
- Department X, Discipline of General Surgery II, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, No. 2, 300041 Timisoara, Romania; (M.C.); (I.-F.F.)
| | - Silviu Moldovan
- Faculty of Medicine, “Vasile Goldis” Western University of Arad, Liviu Rebreanu Str., Nr. 86, 310045 Arad, Romania; (S.M.); (L.P.)
| | - Andreea-Adriana Neamtu
- Surgery Department, Clinical County Emergency Hospital of Arad, Andrenyi Karoly Str., No. 2-4, 310037 Arad, Romania; (M.C.R.); (A.-A.N.); (A.A.); (D.H.); (C.N.); (B.D.T.)
- Department of Toxicology, “Victor Babes” University of Medicine and Pharmacy, E. Murgu Square, No. 2, 300041 Timisoara, Romania
- Research Centre for Pharmaco-Toxicological Evaluation, “Victor Babes” University of Medicine and Pharmacy, E. Murgu Square, No. 2, 300041 Timisoara, Romania
| | - Andrei Ardelean
- Surgery Department, Clinical County Emergency Hospital of Arad, Andrenyi Karoly Str., No. 2-4, 310037 Arad, Romania; (M.C.R.); (A.-A.N.); (A.A.); (D.H.); (C.N.); (B.D.T.)
- Faculty of Medicine, “Vasile Goldis” Western University of Arad, Liviu Rebreanu Str., Nr. 86, 310045 Arad, Romania; (S.M.); (L.P.)
| | - Marco Capitanio
- Department X, Discipline of General Surgery II, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, No. 2, 300041 Timisoara, Romania; (M.C.); (I.-F.F.)
| | - Diana Herczeg
- Surgery Department, Clinical County Emergency Hospital of Arad, Andrenyi Karoly Str., No. 2-4, 310037 Arad, Romania; (M.C.R.); (A.-A.N.); (A.A.); (D.H.); (C.N.); (B.D.T.)
| | - Ionut-Flaviu Faur
- Department X, Discipline of General Surgery II, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, No. 2, 300041 Timisoara, Romania; (M.C.); (I.-F.F.)
| | - Renata Bende
- Center for Advanced Research in Gastroenterology and Hepatology, Department of Internal Medicine II, Division of Gastroenterology and Hepatology, “Victor Babes” University of Medicine and Pharmacy Timisoara, E. Murgu Square, No. 2, 300041 Timisoara, Romania;
| | - Luminita Pilat
- Faculty of Medicine, “Vasile Goldis” Western University of Arad, Liviu Rebreanu Str., Nr. 86, 310045 Arad, Romania; (S.M.); (L.P.)
| | - Virgiliu Mihai Prunoiu
- Clinical Department No. 10, General Surgery, University of Medicine and Pharmacy “Carol Davila”, 050474 Bucharest, Romania;
- Department of Oncological Surgery, Oncological Institute “Prof. Dr. Alexandru Trestioreanu”, 022328 Bucharest, Romania
| | - Carmen Neamtu
- Surgery Department, Clinical County Emergency Hospital of Arad, Andrenyi Karoly Str., No. 2-4, 310037 Arad, Romania; (M.C.R.); (A.-A.N.); (A.A.); (D.H.); (C.N.); (B.D.T.)
- Faculty of Medicine, “Vasile Goldis” Western University of Arad, Liviu Rebreanu Str., Nr. 86, 310045 Arad, Romania; (S.M.); (L.P.)
| | - Bogdan Dan Totolici
- Surgery Department, Clinical County Emergency Hospital of Arad, Andrenyi Karoly Str., No. 2-4, 310037 Arad, Romania; (M.C.R.); (A.-A.N.); (A.A.); (D.H.); (C.N.); (B.D.T.)
- Faculty of Medicine, “Vasile Goldis” Western University of Arad, Liviu Rebreanu Str., Nr. 86, 310045 Arad, Romania; (S.M.); (L.P.)
| |
Collapse
|
8
|
Holowatyj AN, Overman MJ, Votanopoulos KI, Lowy AM, Wagner P, Washington MK, Eng C, Foo WC, Goldberg RM, Hosseini M, Idrees K, Johnson DB, Shergill A, Ward E, Zachos NC, Shelton D. Defining a 'cells to society' research framework for appendiceal tumours. Nat Rev Cancer 2025; 25:293-315. [PMID: 39979656 DOI: 10.1038/s41568-024-00788-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/12/2024] [Indexed: 02/22/2025]
Abstract
Tumours of the appendix - a vestigial digestive organ attached to the colon - are rare. Although we estimate that around 3,000 new appendiceal cancer cases are diagnosed annually in the USA, the challenges of accurately diagnosing and identifying this tumour type suggest that this number may underestimate true population incidence. In the current absence of disease-specific screening and diagnostic imaging modalities, or well-established risk factors, the incidental discovery of appendix tumours is often prompted by acute presentations mimicking appendicitis or when the tumour has already spread into the abdominal cavity - wherein the potential misclassification of appendiceal tumours as malignancies of the colon and ovaries also increases. Notwithstanding these diagnostic difficulties, our understanding of appendix carcinogenesis has advanced in recent years. However, there persist considerable challenges to accelerating the pace of research discoveries towards the path to improved treatments and cures for patients with this group of orphan malignancies. The premise of this Expert Recommendation article is to discuss the current state of the field, to delineate unique challenges for the study of appendiceal tumours, and to propose key priority research areas that will deliver a more complete picture of appendix carcinogenesis and metastasis. The Appendix Cancer Pseudomyxoma Peritonei (ACPMP) Research Foundation Scientific Think Tank delivered a consensus of core research priorities for appendiceal tumours that are poised to be ground-breaking and transformative for scientific discovery and innovation. On the basis of these six research areas, here, we define the first 'cells to society' research framework for appendix tumours.
Collapse
Affiliation(s)
- Andreana N Holowatyj
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA.
- Vanderbilt University School of Medicine, Nashville, TN, USA.
| | - Michael J Overman
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Andrew M Lowy
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Patrick Wagner
- Division of Surgical Oncology, Allegheny Health Network Cancer Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - Mary K Washington
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cathy Eng
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - Wai Chin Foo
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Mojgan Hosseini
- Department of Pathology, University of California, San Diego, San Diego, CA, USA
| | - Kamran Idrees
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Douglas B Johnson
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - Ardaman Shergill
- Department of Medicine, University of Chicago Medical Center, Chicago, IL, USA
| | - Erin Ward
- Section of Surgical Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Nicholas C Zachos
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Deborah Shelton
- Appendix Cancer Pseudomyxoma Peritonei (ACPMP) Research Foundation, Springfield, PA, USA
| |
Collapse
|
9
|
Dai XF, Yang YX, Yang BZ. Glycosylation editing: an innovative therapeutic opportunity in precision oncology. Mol Cell Biochem 2025; 480:1951-1967. [PMID: 38861100 DOI: 10.1007/s11010-024-05033-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 05/06/2024] [Indexed: 06/12/2024]
Abstract
Cancer is still one of the most arduous challenges in the human society, even though humans have found many ways to try to conquer it. With our incremental understandings on the impact of sugar on human health, the clinical relevance of glycosylation has attracted our attention. The fact that altered glycosylation profiles reflect and define different health statuses provide novel opportunities for cancer diagnosis and therapeutics. By reviewing the mechanisms and critical enzymes involved in protein, lipid and glycosylation, as well as current use of glycosylation for cancer diagnosis and therapeutics, we identify the pivotal connection between glycosylation and cellular redox status and, correspondingly, propose the use of redox modulatory tools such as cold atmospheric plasma (CAP) in cancer control via glycosylation editing. This paper interrogates the clinical relevance of glycosylation on cancer and has the promise to provide new ideas for laboratory practice of cold atmospheric plasma (CAP) and precision oncology therapy.
Collapse
Affiliation(s)
- Xiao-Feng Dai
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China.
| | - Yi-Xuan Yang
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
| | - Bo-Zhi Yang
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
| |
Collapse
|
10
|
Zhang Y, Wu D, Zhang Z, Ma J, Jiao S, Ma X, Li J, Meng Y, Zhao Z, Chen H, Jiang Z, Wang G, Liu H, Xi Y, Zhou H, Wang X, Guan X. Impact of lymph node metastasis on immune microenvironment and prognosis in colorectal cancer liver metastasis: insights from multiomics profiling. Br J Cancer 2025; 132:513-524. [PMID: 39753715 PMCID: PMC11920064 DOI: 10.1038/s41416-024-02921-2] [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] [Received: 08/13/2024] [Revised: 11/15/2024] [Accepted: 11/26/2024] [Indexed: 02/19/2025] Open
Abstract
BACKGROUND This study aimed to investigate the prognostic impact of lymph node metastasis (LNM) on patients with colorectal cancer liver metastasis (CRLM) and elucidate the underlying immune mechanisms using multiomics profiling. METHODS We enrolled patients with CRLM from the US Surveillance, Epidemiology, and End Results (SEER) cohort and a multicenter Chinese cohort, integrating bulk RNA sequencing, single-cell RNA sequencing and proteomics data. The cancer-specific survival (CSS) and immune profiles of the tumor-draining lymph nodes (TDLNs), primary tumors and liver metastasis were compared between patients with and without LNM. Pathological evaluations were used to assess immune cell infiltration and histological features. RESULTS The CRLM patients with LNM had significantly shorter CSS than patients without LNM in two large cohorts. Our results showed that nonmetastatic TDLNs exhibited a greater abundance of immune cells, including CD4+ T cells, CD8+ T cells, and CD19+ B cells, whereas metastatic TDLNs were enriched with fibroblasts, endothelial cells, and macrophages. Immunohistochemical analysis confirmed elevated levels of CD3+ T cells, CD8+ T cells, and CD19+ B cells in nonmetastatic TDLNs. The presence of nonmetastatic TDLNs was associated with enhanced antitumor immune responses in primary tumors, characterized by a higher Klintrup-Makinen (KM) grade and the presence of tertiary lymphoid structures. Furthermore, liver metastasis in patients with nonmetastatic TDLNs were predominantly of the desmoplastic growth pattern (dHGP), while those with metastatic TDLNs were predominantly of the replacement growth pattern (rHGP). CONCLUSIONS This research highlights the adverse prognostic impact of LNM on patients with CRLM and reveals potential related mechanisms through multiomics analysis. Our research paves the way for further refinement of the AJCC TNM staging system for CRLM in clinical practice.
Collapse
Affiliation(s)
- Yueyang Zhang
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Deng Wu
- College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China
| | - Zhen Zhang
- Department of Pathology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Jian Ma
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuai Jiao
- Department of Colorectal Surgery, Shanxi Province Cancer Hospital/Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Xiaolong Ma
- Department of Colorectal Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Jiangtao Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yongsheng Meng
- Department of Tumor Biobank, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/ Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Zhixun Zhao
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haipeng Chen
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zheng Jiang
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guiyu Wang
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin, China
| | - Haiyi Liu
- Department of Colorectal Surgery, Shanxi Province Cancer Hospital/Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Yanfeng Xi
- Department of Pathology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China.
| | - Haitao Zhou
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Xishan Wang
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Department of Colorectal Surgery, Shanxi Province Cancer Hospital/Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China.
| | - Xu Guan
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Department of Colorectal Surgery, Shanxi Province Cancer Hospital/Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
11
|
Younis M, Kumar P, Sudershan A, Gezici S, Angral C, Muruganantham B, Mohan G, Behlam I, Digra SK, Anand V. Serum level of carcinoembryonic antigen and risk of colorectal cancer: a case-control study from South Indian population. THE NUCLEUS 2025. [DOI: 10.1007/s13237-025-00543-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 02/11/2025] [Indexed: 05/04/2025] Open
|
12
|
Jin Z, Li Y, Yi H, Wang M, Wang C, Du S, Zeng W, Zong Z. Pathogenetic development, diagnosis and clinical therapeutic approaches for liver metastasis from colorectal cancer (Review). Int J Oncol 2025; 66:22. [PMID: 39950314 PMCID: PMC11844340 DOI: 10.3892/ijo.2025.5728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 01/10/2025] [Indexed: 02/23/2025] Open
Abstract
Colorectal cancer (CRC) is a prevalent malignancy and a significant proportion of patients with CRC develop liver metastasis (CRLM), which is a major contributor to CRC‑related mortality. The present review aimed to comprehensively examine the pathogenetic development and diagnosis of CRLM and the clinical therapeutic approaches for treatment of this disease. The molecular mechanisms underlying CRLM were discussed, including the role of the tumour microenvironment and epithelial‑mesenchymal transition. The present review also highlighted the importance of early detection and the current challenges in predicting the development of CRLM. Various treatment strategies were reviewed, including surgical resection, chemotherapy and immunotherapy, and the potential of novel therapies, such as selective internal radiation therapy and Traditional Chinese Medicine. Despite recent advancements in treatment options, the treatment of CRLM remains a therapeutic challenge due to the complexity of the liver microenvironment and the heterogeneity of CRC. The present review emphasized the need for a multidisciplinary approach and the integration of emerging therapies to improve patient outcomes.
Collapse
Affiliation(s)
- Zhenhua Jin
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- The Second Clinical Medical College of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yin Li
- Huan Kui Academy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Hao Yi
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- The Second Clinical Medical College of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Menghui Wang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Huan Kui Academy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Chaofeng Wang
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Shaokun Du
- The Second Clinical Medical College of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Wenjuan Zeng
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Huan Kui Academy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhen Zong
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
13
|
Fahrmann JF, Yip-Schneider M, Vykoukal J, Spencer R, Dennison JB, Do KA, Long JP, Maitra A, Zhang J, Schmidt CM, Hanash S, Irajizad E. Lead time trajectory of blood-based protein biomarkers for detection of pancreatic cancer based on repeat testing. Cancer Lett 2025; 612:217450. [PMID: 39793753 DOI: 10.1016/j.canlet.2025.217450] [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: 10/23/2024] [Revised: 12/06/2024] [Accepted: 01/07/2025] [Indexed: 01/13/2025]
Abstract
In the current study, we assessed whether repeated measurements of a panel of protein biomarkers with relevance to pancreatic ductal adenocarcinoma (PDAC) improves lead time performance for earlier detection over a single timepoint measurement. Specifically, CA125, CEA, LRG1, REG3A, THBS2, TIMP1, TNRFSF1A as well as CA19-9 were assayed in serially collected pre-diagnostic plasma from 242 PDAC cases and 242 age- and sex-matched non-case control participants in the PLCO cohort. We compared performance estimates of a parametric empirical Bayes (PEB) algorithm, which incorporates participant biomarker history, to that of a single-threshold (ST) method. We demonstrated improvements in AUC estimates (2-13 %) for all biomarkers when considering the PEB approach compared to ST. For CA19-9, the PEBCA19-9 yielded an AUC of 0.88 when at least one repeat measurement was within 3 years of clinical diagnosis. At a specificity of 98.5 %, the PEBCA19-9 identified 15 of the 41 PDAC cases and signaled positive at an average lead-time of 1.09 years whereas the ST approach captured 11 of the 41 PDAC cases with an average positive signal at 0.48 years. Among CA19-9 low individuals, a PEB algorithm based on repeat measurements of TIMP1 yielded an additional 14 % sensitivity at 98.5 % specificity. An adaptive algorithm that considers repeated CA19-9 measurements improves sensitivity and lead-time detection of PDAC compared to a single-threshold method. Additional protein biomarkers may improve sensitivity for earlier detection of PDAC among cases with low CA19-9.
Collapse
Affiliation(s)
- Johannes F Fahrmann
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Jody Vykoukal
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rachelle Spencer
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jennifer B Dennison
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kim-Anh Do
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA, 77030
| | - James P Long
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA, 77030
| | - Anirban Maitra
- Department of Translational Molecular Pathology and Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA, 77030
| | - Jianjun Zhang
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN, USA.
| | - C Max Schmidt
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Samir Hanash
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Ehsan Irajizad
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA, 77030.
| |
Collapse
|
14
|
Peters GW, Thomas G, Applegarth JA, Wasvary J, Bohler F, Callahan RE, Bergeron S, Wasvary HJ. The Effect of the Adoption of the National Accreditation Program for Rectal Cancer Process on Compliance Standards at a Single Institution. Am Surg 2025; 91:345-350. [PMID: 39402893 DOI: 10.1177/00031348241292730] [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: 03/20/2025]
Abstract
Background: The National Accreditation Program for Rectal Cancer (NAPRC) was developed to enhance the quality of rectal cancer care in the United States. This project compared NAPRC compliance at a single tertiary care academic hospital before and after the institution adopted these standards in 2019. Methods: Rectal cancer patients from 2016 to 2023 who met NAPRC eligibility criteria were retrospectively reviewed for compliance with pre-selected patient care standards. Patients diagnosed prior to August 1, 2019 (pre-NAPRC) were compared with those diagnosed afterward (post-NAPRC) to determine whether compliance with these standards differed following the institution's adoption of new guidelines. Results: This study included 353 patients, 146 pre-NAPRC and 207 post-NAPRC. The post-NAPRC group demonstrated significantly higher compliance with pretreatment standards compared to the pre-NAPRC group, including attaining magnetic resonance imaging (MRI) (P = .015), computed tomography (CT) (P < .001), and a carcinoembryonic antigen (CEA) level (P < .001). Postoperative standards were more frequently met in the post-NAPRC group regarding the photographing of surgical specimens (P < .001). No significant differences were observed in confirming a tissue diagnosis, starting treatment within a 60-day timeframe, or completing surgical pathology reports. Prior to initiation of the NAPRC process, the institution had achieved accreditation-level compliance in 2 of the 7 standards. Within 2 years of adopting NAPRC standards, complete compliance was met in 6 of the 7 measures. Conclusions: A single institution's adoption of NAPRC standards improved compliance with multiple rectal cancer care standards, achieving near-complete accreditation level compliance within 2 years.
Collapse
Affiliation(s)
- Garrett W Peters
- Oakland University William Beaumont School of Medicine, Rochester, MI, USA
| | - Gregory Thomas
- Division of Colon and Rectal Surgery, Department of Surgery, Corewell Health William Beaumont University Hospital, Royal Oak, MI, USA
| | - Jacob A Applegarth
- Division of Colon and Rectal Surgery, Department of Surgery, Corewell Health William Beaumont University Hospital, Royal Oak, MI, USA
| | - Joanna Wasvary
- Oakland University William Beaumont School of Medicine, Rochester, MI, USA
| | - Forrest Bohler
- Oakland University William Beaumont School of Medicine, Rochester, MI, USA
| | - Rose E Callahan
- Division of Colon and Rectal Surgery, Department of Surgery, Corewell Health William Beaumont University Hospital, Royal Oak, MI, USA
| | - Shelli Bergeron
- Division of Colon and Rectal Surgery, Department of Surgery, Corewell Health William Beaumont University Hospital, Royal Oak, MI, USA
| | - Harry J Wasvary
- Oakland University William Beaumont School of Medicine, Rochester, MI, USA
- Division of Colon and Rectal Surgery, Department of Surgery, Corewell Health William Beaumont University Hospital, Royal Oak, MI, USA
| |
Collapse
|
15
|
Ghabra S, Chang D, Sugarbaker PH. Preoperative tumor marker elevations in colorectal cancer patients with peritoneal metastases should be used to help select patients for cytoreductive surgery. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:109720. [PMID: 40023022 DOI: 10.1016/j.ejso.2025.109720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/04/2025] [Accepted: 02/19/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND Tumor markers are used routinely for surveillance in patients treated for colorectal cancer (CRC). However, the prognostic implications of elevated preoperative tumor markers in patients treated for CRC and peritoneal metastases (PM) has not been well defined. The utility of carcinoembryonic antigen (CEA), carbohydrate antigen 19-9 (CA19-9) and cancer antigen 125 (CA125) to predict outcome for these patients is reported. METHODS Clinical and histologic features plus preoperative tumor markers were recorded within 1 week prior to CRS. Impact on overall survival of these factors was analyzed by univariate and multivariate analysis. RESULTS Two hundred and four patients were in our database and 140 patients (75.3 %) had at least a single elevated preoperative tumor marker. In an analysis of clinical and histologic parameters preoperatively, a poorly differentiated tumor, signet ring morphology, a peritoneal cancer index (PCI) of ≥16 and an incomplete cytoreduction had a negative impact on median survival. In a multivariate analysis of clinical and histologic features together with tumor markers, an elevated CA19-9 and CA125 was independently associated with reduced overall survival (HR 2.7, p < 0.0001 and HR 2.2, p = 0.005), respectively. Quantitative assessment of CEA (HR 0.5, p = 0.0094) and CA19-9 (HR 4.9, p < 0.001) greater than x10 ULN showed reduced survival. CONCLUSION Preoperative assessment of symptoms and histopathology, PCI and a complete CRS combined with tumor markers CEA, CA19-9 and CA125 are independent prognostic indicators for selection by the multidisciplinary team of CRC PM patients for CRS and HIPEC. All three tumor markers are needed for a meaningful assessment.
Collapse
Affiliation(s)
- Shadin Ghabra
- Department of Surgery, MedStar Washington Hospital Center, Washington, DC, USA
| | | | - Paul H Sugarbaker
- Department of Surgery, MedStar Washington Hospital Center, Washington, DC, USA.
| |
Collapse
|
16
|
Malik AK, Davidson BR, Manas DM. Surgical management, including the role of transplantation, for intrahepatic and peri-hilar cholangiocarcinoma. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:108248. [PMID: 38467524 DOI: 10.1016/j.ejso.2024.108248] [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: 02/26/2024] [Revised: 02/28/2024] [Accepted: 03/03/2024] [Indexed: 03/13/2024]
Abstract
Intrahepatic and peri-hilar cholangiocarcinoma are life threatening disease with poor outcomes despite optimal treatment currently available (5-year overall survival following resection 20-35%, and <10% cured at 10-years post resection). The insidious onset makes diagnosis difficult, the majority do not have a resection option and the high recurrence rate post-resection suggests that occult metastatic disease is frequently present. Advances in perioperative management, such as ipsilateral portal vein (and hepatic vein) embolisation methods to increase the future liver remnant volume, genomic profiling, and (neo)adjuvant therapies demonstrate great potential in improving outcomes. However multiple areas of controversy exist. Surgical resection rate and outcomes vary between centres with no global consensus on how 'resectable' disease is defined - molecular profiling and genomic analysis could potentially identify patients unlikely to benefit from resection or likely to benefit from targeted therapies. FDG-PET scanning has also improved the ability to detect metastatic disease preoperatively and avoid futile resection. However tumours frequently invade major vasculo-biliary structures, with resection and reconstruction associated with significant morbidity and mortality even in specialist centres. Liver transplantation has been investigated for very selected patients for the last decade and yet the selection algorithm, surgical approach and both value of both neoadjuvant and adjuvant therapies remain to be clarified. In this review, we discuss the contemporary management of intrahepatic and peri-hilar cholangiocarcinoma.
Collapse
Affiliation(s)
- Abdullah K Malik
- Department of HPB and Transplant Surgery, Freeman Hospital, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK; NIHR Blood and Transplant Research Unit, Newcastle University and Cambridge University, Newcastle Upon Tyne, UK.
| | - Brian R Davidson
- Department of HPB and Liver Transplant Surgery, Royal Free Hospital, Royal Free London NHS Foundation Trust, London, UK; Division of Surgery and Interventional Sciences, University College London, London, UK
| | - Derek M Manas
- Department of HPB and Transplant Surgery, Freeman Hospital, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK; NIHR Blood and Transplant Research Unit, Newcastle University and Cambridge University, Newcastle Upon Tyne, UK; NHS Blood and Transplant, Bristol, UK
| |
Collapse
|
17
|
Keshvari A, Tafti SMA, Keramati MR, Fazeli MS, Kazemeini A, Behboudi B, Asbagh RA, Mirzasadeghi A. Preoperative Carcinoembryonic Antigen as a Predictor of 5-Year Survival in Rectal Cancer: Proposing a New Prognostic Cutoff. J Gastrointest Cancer 2025; 56:46. [PMID: 39826023 DOI: 10.1007/s12029-025-01175-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2025] [Indexed: 01/20/2025]
Abstract
PURPOSE Carcinoembryonic antigen (CEA) is an important prognostic factor for rectal cancer. This study aims to introduce a novel cutoff point for CEA within the normal range to improve prognosis prediction and enhance patient stratification in rectal cancer patients. METHODS A total of 316 patients with stages I to III rectal cancer who underwent surgical tumor resection were enrolled. The Cox proportional hazards regression model was used to evaluate the impact of preoperative CEA level and other co-variates on overall survival (OS). The Youden Index method was used for CEA optimal cutoff estimation. RESULTS The mean follow-up period was 46.47 months. In risk-adjusted Cox proportional analysis, higher preoperative CEA levels (HR 1.17, CI 1.131.21; P < 0.001), and T-stage were associated with poor OS. The mean preoperative CEA level was significantly higher in patients with positive lymphovascular invasion (LVI) and perineural invasion (PNI) (CI: 1.06-2.45 and 0.75-2.33, respectively, P < 0.001, t test). Pathologic complete response (pCR) occurred in 71 (22.4%) cases. Patients with pCR had lower levels of preoperative CEA than non-pCR group (P = 0.002, CEApCR-CEAnonpCR = - 1.3; t test). Using Youden Index, the estimated optimal CEA cutoff value for predicting OS was 2.8 ng/mL (sensitivity 90%; specificity 78.5%). Lower preoperative CEA levels predict higher pCR rates, aiding patient stratification and planning. CONCLUSION Preoperative CEA may play a role in the prediction of pCR in rectal cancer. Considering the CEA level of 2.8 ng/ml, as a newly defined cutoff point, patients with a worse prognosis can be identified prior to operation. PNI, along with LVI as independent predictors, may be contemplated as prognostic indicators to improve treatment strategies.
Collapse
Affiliation(s)
- Amir Keshvari
- Colorectal Research Center, Imam Khomeini Hospital complex, Tehran University of Medical Sciences, Keshavarz Blvd, Tehran, Iran
- Division of Colorectal Surgery, Department of Surgery, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mohsen Ahmadi Tafti
- Colorectal Research Center, Imam Khomeini Hospital complex, Tehran University of Medical Sciences, Keshavarz Blvd, Tehran, Iran
- Division of Colorectal Surgery, Department of Surgery, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Keramati
- Colorectal Research Center, Imam Khomeini Hospital complex, Tehran University of Medical Sciences, Keshavarz Blvd, Tehran, Iran
- Division of Colorectal Surgery, Department of Surgery, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Sadegh Fazeli
- Colorectal Research Center, Imam Khomeini Hospital complex, Tehran University of Medical Sciences, Keshavarz Blvd, Tehran, Iran
- Division of Colorectal Surgery, Department of Surgery, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Kazemeini
- Colorectal Research Center, Imam Khomeini Hospital complex, Tehran University of Medical Sciences, Keshavarz Blvd, Tehran, Iran
- Division of Colorectal Surgery, Department of Surgery, Tehran University of Medical Sciences, Tehran, Iran
| | - Behnam Behboudi
- Colorectal Research Center, Imam Khomeini Hospital complex, Tehran University of Medical Sciences, Keshavarz Blvd, Tehran, Iran
- Division of Colorectal Surgery, Department of Surgery, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Akbari Asbagh
- Colorectal Research Center, Imam Khomeini Hospital complex, Tehran University of Medical Sciences, Keshavarz Blvd, Tehran, Iran
- Division of Colorectal Surgery, Department of Surgery, Tehran University of Medical Sciences, Tehran, Iran
| | - Anahita Mirzasadeghi
- Colorectal Research Center, Imam Khomeini Hospital complex, Tehran University of Medical Sciences, Keshavarz Blvd, Tehran, Iran.
- Division of Colorectal Surgery, Department of Surgery, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
18
|
Zhao X, Wang Y, Zhou L, Ye A, Zhu Q. Changes of CA19-9 levels and related influencing factors in patients with type 2 diabetes mellitus after antidiabetic therapy. Sci Rep 2025; 15:1264. [PMID: 39779798 PMCID: PMC11711652 DOI: 10.1038/s41598-025-85807-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 01/06/2025] [Indexed: 01/11/2025] Open
Abstract
Abnormalities of carbohydrate antigen 19 - 9 (CA19-9) are common in patients with type 2 diabetes mellitus (T2DM), and in some patients, CA19-9 returns to normal level after glycemic control. The aim of this study was to investigate the factors associated with CA19-9 levels in patients with T2DM and the associated influences on the degree of reduction of CA19-9 levels after antidiabetic therapy (AT). This study was an observational cross-sectional study. A total of 213 patients with T2DM were enrolled in this study, of whom 105 with abnormal CA19-9 and 108 with normal CA19-9. Socio-demographic information, complete blood counts, biochemical indicators, thyroid function indicators, and CA19-9 level were collected separately for each subject. Levels of glycosylated hemoglobin, type A1C (HbA1c), fasting blood glucose (FBG) were significantly higher in T2DM patients with abnormal CA19-9 compared to patients with normal CA19-9 (both FDR < 0.001). CA19-9 level was significantly and positively correlated with neutrophil/lymphocyte ratio (NLR) (r = 0.16, P = 0.02), monocyte/lymphocyte ratio (MLR) (r = 0.16, P = 0.02), and FBG (r = 0.38, P < 0.001), while significantly and negatively correlated with free triiodothyronine (FT3) (r=-0.22, P = 0.002) and albumin count (r=-0.18, P = 0.007). After AT, the degree of decrease in CA19-9 level in T2DM patients with abnormal CA19-9 was significantly positively correlated with degree of decrease in FBG (r = 0.33, P < 0.001), as well as CA19-9 level before AT (r = 0.73, P < 0.001), NLR (r = 0.20, P = 0.04), and MLR (r = 0.25, P = 0.01). In this study, we investigated the influencing factors associated with CA19-9 level and the factors influencing degree of CA19-9 reduction after AT in T2DM patients with abnormal CA19-9.
Collapse
Affiliation(s)
- Xiafei Zhao
- Affiliated Xiaoshan Hospital, Hangzhou Normal University/Zhejiang Xiaoshan Hospital, 728 Yucai North Road, Hangzhou, 311200, China
| | - Yan Wang
- Affiliated Xiaoshan Hospital, Hangzhou Normal University/Zhejiang Xiaoshan Hospital, 728 Yucai North Road, Hangzhou, 311200, China
| | - Ling Zhou
- Affiliated Xiaoshan Hospital, Hangzhou Normal University/Zhejiang Xiaoshan Hospital, 728 Yucai North Road, Hangzhou, 311200, China
| | - Aili Ye
- Affiliated Xiaoshan Hospital, Hangzhou Normal University/Zhejiang Xiaoshan Hospital, 728 Yucai North Road, Hangzhou, 311200, China.
| | - Quanfeng Zhu
- Affiliated Xiaoshan Hospital, Hangzhou Normal University/Zhejiang Xiaoshan Hospital, 728 Yucai North Road, Hangzhou, 311200, China.
| |
Collapse
|
19
|
Athanasiou A, Kureshi N, Wittig A, Sterner M, Huber R, Palma NA, King T, Schiess R. Biomarker Discovery for Early Detection of Pancreatic Ductal Adenocarcinoma (PDAC) Using Multiplex Proteomics Technology. J Proteome Res 2025; 24:315-322. [PMID: 39699878 PMCID: PMC11705213 DOI: 10.1021/acs.jproteome.4c00752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/27/2024] [Accepted: 12/13/2024] [Indexed: 12/20/2024]
Abstract
Early detection of pancreatic ductal adenocarcinoma (PDAC) can improve survival but is hampered by the absence of early disease symptoms. Imaging remains key for surveillance but is cumbersome and may lack sensitivity to detect small tumors. CA19-9, the only FDA-approved blood biomarker for PDAC, is insufficiently sensitive and specific to be recommended for surveillance. We aimed to discover a blood-based protein signature to improve PDAC detection in our main target population consisting of stage I or II PDAC patients (n = 75) and various controls including healthy controls (n = 50), individuals at high risk (genetic and familial) for PDAC (n = 47), or those under surveillance for an intraductal papillary mucinous neoplasm (n = 36). Roughly 3000 proteins were measured using Olink multiplex technology and conventional immunoassays. Machine learning combined biomarker candidates into 4- to 6-plex signatures. These signatures significantly (p < 0.001) outperformed CA19-9 with 84% sensitivity at 95% specificity, compared to CA19-9's sensitivity of 53% in the target population. Exploratory analysis was performed in new-onset diabetes (n = 81) and chronic pancreatitis (n = 50) patients. In conclusion, 41 promising biomarker candidates across multiple signatures were identified using proteomics technology and will be further tested in an independent cohort.
Collapse
Affiliation(s)
| | - Natasha Kureshi
- Immunovia
Inc., 26 Forest Street,
Suite 110, Marlborough, Massachusetts 01752, United States
| | - Anja Wittig
- Proteomedix
AG, Wagistrasse 23, CH-8952 Schlieren, Switzerland
| | - Maria Sterner
- Immunovia
AB, Medicon Village,
Scheelevägen 8, SE-223 63 Lund, Sweden
| | - Ramy Huber
- Proteomedix
AG, Wagistrasse 23, CH-8952 Schlieren, Switzerland
| | - Norma A. Palma
- Immunovia
Inc., 26 Forest Street,
Suite 110, Marlborough, Massachusetts 01752, United States
| | - Thomas King
- Immunovia
Inc., 26 Forest Street,
Suite 110, Marlborough, Massachusetts 01752, United States
| | - Ralph Schiess
- Proteomedix
AG, Wagistrasse 23, CH-8952 Schlieren, Switzerland
| |
Collapse
|
20
|
Zhang L, Li YW, Xie T, Sun K, Huang X, Xiong W, Liu RJ. Potential role of P4HB in the tumor microenvironment and its clinical prognostic value: a comprehensive pan-cancer analysis and experimental validation with a focus on KIRC. Cancer Cell Int 2025; 25:1. [PMID: 39754183 PMCID: PMC11697512 DOI: 10.1186/s12935-024-03575-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 11/13/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND Tumor microenvironment (TME) plays a crucial role in tumor growth and metastasis. Exploring biomarkers that are significantly associated with TME can help guide individualized treatment of patients. METHODS We analyzed the expression and survival of P4HB in pan-cancer through the TCGA database, and verified the protein level of P4HB by the HPA database. In addition, we used the Metascape database to construct protein-protein interaction networks and the single-cell Sequencing database for functional analysis. An immune cell infiltration analysis was performed to explore the potential role of P4HB in TME. We further analyze the relationship between P4HB and immune checkpoint molecules to explore the role of P4HB in immune checkpoint blockade therapy. Finally, the oncogenic role of P4HB in RCC cells was validated using colony formation and wound healing assays. RESULTS RNA and protein levels of P4HB were extensively up-regulated in pan-cancer. However, high P4HB expression was associated with poor survival in KIRC. The clinical relevance analyses of P4HB suggested that high P4HB expression was associated with advanced clinical TNM stage. Moreover, multivariate cox regression analysis indicated that P4HB (HR = 1.372, 95% CI 1.047-1.681, P = 0.019) was an independent risk factor for OS in KIRC. Functional analysis revealed that P4HB is involved in hypoxia, TME and immune system processes. Our study also found that high P4HB expression was significantly correlated with elevated infiltration levels in CD8 + T cells and M2 macrophages. The results of colony formation and wound healing assays showed that knockdown of P4HB inhibited the RCC growth and migration. CONCLUSIONS P4HB is a specific biomarker for KIRC prognosis and is significantly associated with clinical characteristics. In addition, P4HB may play an influential role in TME and is a biomarker for ICB therapy.
Collapse
Affiliation(s)
- Linxue Zhang
- School of Materials and Energy, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Yu-Wei Li
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, China
| | - Tianyi Xie
- Department of Neuroscience, Kenneth P. Dietrich School of Arts & Science, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ke Sun
- School of Materials and Energy, University of Electronic Science and Technology of China, Chengdu, 610054, China.
| | - Xiang Huang
- Department of Urology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Wei Xiong
- Department of Urology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Rui-Ji Liu
- Department of Urology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
21
|
Lopes M, Figueiredo V, Mendes A, Amaral M, Delgado Alves J. Lung Adenocarcinoma Presenting With Elevated Serum Carbohydrate Antigen 19-9 Levels: A Case Report. Cureus 2025; 17:e77786. [PMID: 39981495 PMCID: PMC11841819 DOI: 10.7759/cureus.77786] [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] [Accepted: 01/21/2025] [Indexed: 02/22/2025] Open
Abstract
Carbohydrate antigen 19-9 (CA 19-9) is a tumor marker usually used for the diagnosis and follow-up of pancreatic, gastric, and hepatobiliary malignancies. However, it has a low specificity and can be elevated in a wide array of other conditions. CA 19-9 elevation in lung tumors seems to be associated with the worst prognosis, but its role in this condition is not fully established yet. We present the case of a 78-year-old woman with mild consumptive symptoms, consisting of unselective anorexia and weight loss, and isolated high levels of CA 19-9. Hepatobiliary and gastrointestinal lesions were excluded, and a suspicious lesion was found in the right lung. A biopsy, which was only possible several months after presentation due to the location and small dimensions of the lesion, confirmed the diagnosis of lung adenocarcinoma. After an initial indolent course, the patient evolved with disease progression and is currently under treatment with palliative chemotherapy. This case illustrates the thorough investigation performed to clarify the elevation of a tumor biomarker measured in a pauci-symptomatic patient, which led to the identification of a rare and unsuspected etiology of high levels of CA 19-9.
Collapse
Affiliation(s)
- Mónica Lopes
- Department of Internal Medicine IV, Hospital Professor Doutor Fernando Fonseca, Amadora, PRT
| | - Vera Figueiredo
- Department of Pulmonology, Hospital Professor Doutor Fernando Fonseca, Amadora, PRT
| | - Ana Mendes
- Department of Oncology, Hospital Professor Doutor Fernando Fonseca, Amadora, PRT
| | - Marta Amaral
- Department of Internal Medicine IV, Hospital Professor Doutor Fernando Fonseca, Amadora, PRT
| | - José Delgado Alves
- Department of Internal Medicine IV, Hospital Professor Doutor Fernando Fonseca, Amadora, PRT
| |
Collapse
|
22
|
Xu R, Chi H, Zhang Q, Li X, Hong Z. Enhancing the diagnostic accuracy of colorectal cancer through the integration of serum tumor markers and hematological indicators with machine learning algorithms. Clin Transl Oncol 2025; 27:299-308. [PMID: 38902493 DOI: 10.1007/s12094-024-03564-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 06/09/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND Colorectal cancer has a high incidence and mortality rate due to a low rate of early diagnosis. Therefore, efficient diagnostic methods are urgently needed. PURPOSE This study assesses the diagnostic effectiveness of Carbohydrate Antigen 19-9 (CA19-9), Carcinoembryonic Antigen (CEA), Alpha-fetoprotein (AFP), and Cancer Antigen 125 (CA125) serum tumor markers for colorectal cancer (CRC) and investigates a machine learning-based diagnostic model incorporating these markers with blood biochemical indices for improved CRC detection. METHOD Between January 2019 and December 2021, data from 800 CRC patients and 697 controls were collected; 52 patients and 63 controls attending the same hospital in 2022 were collected as an external validation set. Markers' effectiveness was analyzed individually and collectively, using metrics like ROC curve AUC and F1 score. Variables chosen through backward regression, including demographics and blood tests, were tested on six machine learning models using these metrics. RESULT In the case group, the levels of CEA, CA199, and CA125 were found to be higher than those in the control group. Combining these with a fourth serum marker significantly improved predictive efficacy over using any single marker alone, achieving an Area Under the Curve (AUC) value of 0.801. Using stepwise regression (backward), 17 variables were meticulously selected for evaluation in six machine learning models. Among these models, the Gradient Boosting Machine (GBM) emerged as the top performer in the training set, test set, and external validation set, boasting an AUC value of over 0.9, indicating its superior predictive power. CONCLUSION Machine learning models integrating tumor markers and blood indices offer superior CRC diagnostic accuracy, potentially enhancing clinical practice.
Collapse
Affiliation(s)
- Rongxuan Xu
- Department of Epidemiology and Health Statistics, Dalian Medical University, Dalian, China
| | | | - Qian Zhang
- Department of Epidemiology and Health Statistics, Dalian Medical University, Dalian, China
| | - Xiaofeng Li
- Department of Epidemiology and Health Statistics, Dalian Medical University, Dalian, China.
| | - Zhijun Hong
- The Health Management Center, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China.
| |
Collapse
|
23
|
Chen Y, Mi Y, Tan S, Chen Y, Liu S, Lin S, Yang C, Hong W, Li W. CEA-induced PI3K/AKT pathway activation through the binding of CEA to KRT1 contributes to oxaliplatin resistance in gastric cancer. Drug Resist Updat 2025; 78:101179. [PMID: 39644827 DOI: 10.1016/j.drup.2024.101179] [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: 09/12/2024] [Revised: 11/28/2024] [Accepted: 12/01/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND The serum level of carcinoembryonic antigen (CEA) has prognostic value in patients with gastric cancer (GC) receiving oxaliplatin-based chemotherapy. As the molecular functions of CEA are increasingly uncovered, its role in regulating oxaliplatin resistance in GC attracts attention. METHODS The survival analysis adopted the KaplanMeier method. Effects of CEA on proliferative capacity were investigated using CCK8, colony formation, and xenograft assays. Oxaliplatin sensitivity was identified through IC50 detection, apoptosis analysis, comet assay, organoid culture model, and xenograft assay. Multi-omics approaches were utilized to explore CEA's downstream effects. The binding of CEA to KRT1 was confirmed through proteomic analysis and Co-IP, GST pull-down, and immunofluorescence colocalization assays. Furthermore, small molecule inhibitors were identified using virtual screening and surface plasmon resonance. RESULTS Starting from clinical data, we confirmed that CEA demonstrated superior ability to predict the prognosis of patients with GC who received oxaliplatin-based chemotherapy, particularly in predicting recurrence-free survival based on serum CEA level. In vitro and in vivo experiments revealed CEAhigh GC cells presented increased proliferative capacity and decreased oxaliplatin sensitivity. The resistance phenotype was transmitted through secreted CEA. Multi-omics analysis revealed that CEA activated the PI3K/AKT pathway by binding to KRT1, leading to oxaliplatin resistance. Finally, the small molecule inhibitor evacetrapib, which competitively inhibits the CEA-KRT1 interaction, was identified and validated in vitro. CONCLUSIONS In summary, the CEA-KRT1-PI3K/AKT axis regulates oxaliplatin sensitivity in GC cells. Treatment with small molecule inhibitors such as evacetrapib to inhibit this interaction constitutes a novel therapeutic strategy.
Collapse
Affiliation(s)
- Yifan Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350013, China; Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Yulong Mi
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350013, China; Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Song Tan
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350013, China; Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Yizhen Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350013, China; Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Shaolin Liu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350013, China; Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Shengtao Lin
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350013, China; Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Changshun Yang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350013, China; Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Weifeng Hong
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou 310005, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310005, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou 310005, China.
| | - Weihua Li
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350013, China; Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Fuzhou 350001, China.
| |
Collapse
|
24
|
Hussung S, Hess ME, Haghighi EB, Wittel UA, Boerries M, Fritsch RM. Integrated Analysis of Cell-Free DNA and Novel Protein Biomarkers for Stratification and Therapy Monitoring in Stage IV Pancreatic Cancer: A Preliminary Study. Diagnostics (Basel) 2024; 15:49. [PMID: 39795577 PMCID: PMC11720586 DOI: 10.3390/diagnostics15010049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 12/25/2024] [Accepted: 12/26/2024] [Indexed: 01/13/2025] Open
Abstract
Background: Given the poor prognosis of metastatic pancreatic adenocarcinoma (mPDAC), closer disease monitoring through liquid biopsy, most frequently based on serial measurements of cell-free mutated KRAS (KRASmut cfDNA), has become a highly active research focus, aimed at improving patients' long-term outcomes. However, most of the available data show only a limited predictive and prognostic value of single-parameter-based methods. We hypothesized that a combined longitudinal analysis of KRASmut cfDNA and novel protein biomarkers could improve risk stratification and molecular monitoring of patients with mPDAC. Methods: We prospectively collected 160 plasma samples from 47 patients with mPDAC at our institution. Highly sensitive single-target ddPCR assays were employed to detect and quantify KRASmut cfDNA. Additionally, analysis of ten protein biomarkers was performed through Enzyme-linked Immunosorbent Assay (ELISA), and Carbohydrate-Antigen 19-9 (CA 19-9) dynamics were registered. Results: KRASmut cfDNA was detectable in 37/47 (78.7%) patients throughout the course of study, and CA 19-9 levels were elevated in 40 out of 47 (85.1%) patients. KRASmut cfDNA increase at the time of the first follow-up could predict inferior progression-free survival (PFS) (Hazard ratio (HR) = 3.40, p = 0.0003) and overall survival (OS) (HR = 4.91, p < 0.0001). In contrast to CA 19-9 kinetics, which were not predictive of outcome, integrated analysis of KRASmut cfDNA combined with six evaluated circulating protein biomarkers allowed basal risk stratification at the time of the first follow-up (HR = 10.2, p = 0.0014). Conclusions: A combined longitudinal analysis of KRASmut cfDNA with selected protein biomarkers offers significantly improved prognostic value for patients with mPDAC compared to single-parameter methods. This innovative approach is a step forward in the molecular monitoring of mPDAC and should be validated in further prospective studies.
Collapse
Affiliation(s)
- Saskia Hussung
- Department of Medicine I (Hematology, Oncology and Stem Cell Transplantation), Freiburg University Medical Center, 79106 Freiburg, Germany;
- Department of Medical Oncology and Hematology, Zurich University Hospital, 8091 Zurich, Switzerland
| | - Maria E. Hess
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (M.E.H.); (E.B.H.); (M.B.)
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Elham Bavafaye Haghighi
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (M.E.H.); (E.B.H.); (M.B.)
| | - Uwe A. Wittel
- Department of Surgery, Freiburg University Medical Center, 79106 Freiburg, Germany;
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; (M.E.H.); (E.B.H.); (M.B.)
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Comprehensive Cancer Center Freiburg (CCCF), Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Ralph M. Fritsch
- Department of Medicine I (Hematology, Oncology and Stem Cell Transplantation), Freiburg University Medical Center, 79106 Freiburg, Germany;
- Department of Medical Oncology and Hematology, Zurich University Hospital, 8091 Zurich, Switzerland
- Department of Surgery, Freiburg University Medical Center, 79106 Freiburg, Germany;
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
25
|
Su QX, Zheng ZJ, Xie YH, Chu LY, Lin YW, Liu YQ, Li XX, Peng YH, Xu YW, Xie JJ. The diagnostic value of serum Ephrin-A1 in patients with colorectal cancer. Sci Rep 2024; 14:31194. [PMID: 39732744 DOI: 10.1038/s41598-024-82540-2] [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] [Received: 07/21/2024] [Accepted: 12/05/2024] [Indexed: 12/30/2024] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignant tumors worldwide, with a high incidence rate and mortality. The analysis of serum biomarkers for colorectal cancer diagnosis has attracted more and more attention because of its low cost, repeatability, and quantification. This study was aimed to evaluate the diagnostic performance of serum Ephrin-A1 in patients with CRC. We retrospectively analyzed CRC cases in a test cohort (121 patients and 108 controls) and validated them in a validation cohort (119 patients and 118 controls). The concentration of Ephrin-A1 in serum was detected by Enzyme-linked immunosorbent assay (ELISA) and the diagnostic performance of serum Ephrin-A1 was evaluated by receiver operating characteristic (ROC) analysis. In the test cohort, serum Ephrin-A1 levels in patients with all-stage CRC and early-stage CRC were significantly higher than those in healthy controls. The area under the ROC curve (AUC), sensitivity and specificity of all-stage CRC and early-stage CRC were 0.709 (95% CI 0.644-0.775) and 0.660 (95% CI 0.530-0.790), 48.76% and 45.00%, 81.48% and 81.48%, respectively. Similar results were observed in the validation cohort. Serum Ephrin-A1 might be served as a potential biomarker in the diagnosis of CRC.
Collapse
Affiliation(s)
- Qi-Xin Su
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, Institute of Biochemistry & Molecular Biology, Guangdong Medical University, Dongguan, China
| | - Ze-Jun Zheng
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, Institute of Biochemistry & Molecular Biology, Guangdong Medical University, Dongguan, China
| | - Ying-Hua Xie
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, China
| | - Ling-Yu Chu
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, China
- Department of Clinical Laboratory Medicine, The Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Yi-Wei Lin
- Department of Clinical Laboratory Medicine, The Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Yin-Qiao Liu
- Department of Clinical Laboratory Medicine, The Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Xin-Xin Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Yu-Hui Peng
- Department of Clinical Laboratory Medicine, The Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Yi-Wei Xu
- Department of Clinical Laboratory Medicine, The Cancer Hospital of Shantou University Medical College, Shantou, China.
| | - Jian-Jun Xie
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, Institute of Biochemistry & Molecular Biology, Guangdong Medical University, Dongguan, China.
| |
Collapse
|
26
|
Bockorny B, Muthuswamy L, Huang L, Hadisurya M, Maria Lim C, Tsai LL, Gill RR, Wei JL, Bullock AJ, Grossman JE, Besaw RJ, Narasimhan S, Tao WA, Perea S, Sawhney MS, Freedman SD, Hildago M, Iliuk A, Muthuswamy SK. A large-scale proteomics resource of circulating extracellular vesicles for biomarker discovery in pancreatic cancer. eLife 2024; 12:RP87369. [PMID: 39693144 DOI: 10.7554/elife.87369] [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] [Indexed: 12/19/2024] Open
Abstract
Pancreatic cancer has the worst prognosis of all common tumors. Earlier cancer diagnosis could increase survival rates and better assessment of metastatic disease could improve patient care. As such, there is an urgent need to develop biomarkers to diagnose this deadly malignancy. Analyzing circulating extracellular vesicles (cEVs) using 'liquid biopsies' offers an attractive approach to diagnose and monitor disease status. However, it is important to differentiate EV-associated proteins enriched in patients with pancreatic ductal adenocarcinoma (PDAC) from those with benign pancreatic diseases such as chronic pancreatitis and intraductal papillary mucinous neoplasm (IPMN). To meet this need, we combined the novel EVtrap method for highly efficient isolation of EVs from plasma and conducted proteomics analysis of samples from 124 individuals, including patients with PDAC, benign pancreatic diseases and controls. On average, 912 EV proteins were identified per 100 µL of plasma. EVs containing high levels of PDCD6IP, SERPINA12, and RUVBL2 were associated with PDAC compared to the benign diseases in both discovery and validation cohorts. EVs with PSMB4, RUVBL2, and ANKAR were associated with metastasis, and those with CRP, RALB, and CD55 correlated with poor clinical prognosis. Finally, we validated a seven EV protein PDAC signature against a background of benign pancreatic diseases that yielded an 89% prediction accuracy for the diagnosis of PDAC. To our knowledge, our study represents the largest proteomics profiling of circulating EVs ever conducted in pancreatic cancer and provides a valuable open-source atlas to the scientific community with a comprehensive catalogue of novel cEVs that may assist in the development of biomarkers and improve the outcomes of patients with PDAC.
Collapse
Affiliation(s)
- Bruno Bockorny
- Division of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, United States
- Harvard Medical School, Boston, United States
| | | | - Ling Huang
- Henry Ford Cancer Institute, Detroit, United States
| | - Marco Hadisurya
- Department of Biochemistry, Purdue University West Lafayette, West Lafayette, United States
| | | | - Leo L Tsai
- Harvard Medical School, Boston, United States
- Department of Radiology, Beth Israel Deaconess Medical Center, Boston, United States
| | - Ritu R Gill
- Harvard Medical School, Boston, United States
- Department of Radiology, Beth Israel Deaconess Medical Center, Boston, United States
| | - Jesse L Wei
- Harvard Medical School, Boston, United States
- Department of Radiology, Beth Israel Deaconess Medical Center, Boston, United States
| | - Andrea J Bullock
- Division of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, United States
- Harvard Medical School, Boston, United States
| | | | - Robert J Besaw
- Division of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, United States
| | | | - Weiguo Andy Tao
- Department of Biochemistry, Purdue University West Lafayette, West Lafayette, United States
| | - Sofia Perea
- Division of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, United States
| | - Mandeep S Sawhney
- Harvard Medical School, Boston, United States
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Boston, United States
| | - Steven D Freedman
- Harvard Medical School, Boston, United States
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Boston, United States
| | - Manuel Hildago
- Division of Hematology-Oncology, Weill Cornell Medical College, New York, United States
- New York-Presbyterian Hospital, New York, United States
| | - Anton Iliuk
- Tymora Analytical Operations, West Lafayette, United States
| | | |
Collapse
|
27
|
Piroozkhah M, Zabihi M, Jalali P, Salehi Z. Comprehensive Multi-Omics Analysis Reveals NPC2 and ITGAV Genes as Potential Prognostic Biomarkers in Gastrointestinal Cancers. Cancer Rep (Hoboken) 2024; 7:e70087. [PMID: 39690926 DOI: 10.1002/cnr2.70087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 10/23/2024] [Accepted: 12/03/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND Gastrointestinal cancers (GICs) continue to dominate in terms of both incidence and mortality worldwide. Due to the absence of efficient and accurate prognostic biomarkers, the prognosis and treatment outcomes of many GICs are poor. Identifying biomarkers to predict individual clinical outcomes efficiently is a fundamental challenge in clinical oncology. Although several biomarkers have been continually discovered, their predictive accuracy is relatively modest, and their therapeutic use is restricted. In light of this, the discovery of reliable biomarkers for predicting prognosis and outcome in GIC is urgently required. MATERIALS AND METHODS We evaluated the Human Protein Atlas dataset and identified NPC Intracellular Cholesterol Transporter 2 (NPC2) and Integrin Subunit Alpha V (ITGAV) as probable poor predictive genes for these cancers. In addition, we used the GEPIA2, cBioPortal, UALCAN, LinkedOmics, STRING, Enrichr, TISDB, TIMER2.0, hTFTarget, miRTarBase, circBank, and drug-gene interaction database databases to conduct a comprehensive and systematic analysis of the NPC2 and ITGAV genes. RESULT Our results found high expression levels of NPC2 and ITGAV in most GICs. The aforementioned gene expressions were linked to several clinicopathological characteristics of GICs as well as poorer prognosis in LIHC and STAD. The most common alteration type of NPC2 was amplification, and for ITGAV was deep deletion. Significant promotor hypermethylation was also seen in NPC2 and ITGAV in PAAD and COAD, respectively. For the immunologic significance, NPC2 and ITGAV were positively correlated with the abundance of tumor-infiltrating lymphocytes and macrophages. Furthermore, various immunomodulators showed strong correlations with the expression of these genes. There were currently 10 small molecule drugs targeting ITGAV. CONCLUSION Consequently, our bioinformatics analysis showed that NPC2 and ITGAV might be used as potential biomarkers to determine the prognosis of various GICs and are also related to immune infiltration.
Collapse
Affiliation(s)
- Moein Piroozkhah
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Zabihi
- Institute of Biochemistry and Biophysics (IBB), Department of Bioinformatics, Laboratory of Complex Biological Systems and Bioinformatics (CBB), University of Tehran, Tehran, Iran
| | - Pooya Jalali
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Salehi
- Hematology, Oncology and Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
28
|
Yang S, Ma R, Wu J. The diagnostic value of combining preoperative serum CA19-9, ALBI score, and 18 F-FDG PET/CT imaging in preoperative resectability of pancreatic cancer. Nucl Med Commun 2024; 45:1061-1068. [PMID: 39363633 DOI: 10.1097/mnm.0000000000001910] [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: 10/05/2024]
Abstract
OBJECTIVE Pancreatic cancer is an increasing cause of cancer-related mortality, with persistently low survival rates. We investigated the clinical diagnostic value of the combination of preoperative serum carbohydrate antigen 19-9 (CA19-9), albumin-bilirubin (ALBI) score, and 18 F-fluoro-2-deoxy- d -glucose PET integrated with computed tomography ( 18 F-FDG PET/CT) imaging in pancreatic cancer preoperative resectability. METHODS This study included 143 pancreatic cancer patients, including 68 preoperative resectable and 75 preoperative unresectable pancreatic cancer patients. Meanwhile, 67 patients with non-pancreatic cancer were included as the control group. The clinical data were collected. Serum CA19-9 level was measured by ELISA. The levels of total bilirubin and albumin were determined using a biochemical analyzer, with the ALBI score calculated. All patients underwent 18 F-FDG PET/CT imaging. The consistency of the diagnosis was evaluated by the Kappa test. Logistic univariate and multivariate regression analyses were performed. The diagnostic efficacy of these parameters was evaluated using receiver operating characteristic (ROC) curves, and the optimal ROC curve thresholds were obtained using the Youden index. RESULTS The preoperative serum CA19-9 and ALBI score of patients with preoperative resectable pancreatic cancer were increased, which helped diagnose preoperative resectable pancreatic cancer. 18 F-FDG PET/CT imaging had diagnostic value for preoperative resectable pancreatic cancer. Preoperative serum CA19-9, ALBI score, and 18 F-FDG PET/CT imaging were independent influencing factors for pancreatic cancer preoperative resectability, and their combination had higher diagnostic value for preoperative resectable pancreatic cancer than any single of these indexes. CONCLUSION The combination of preoperative serum CA19-9, ALBI score, and 18 F-FDG PET/CT imaging had high diagnostic value for pancreatic cancer preoperative resectability.
Collapse
Affiliation(s)
- Shuli Yang
- Imaging Diagnosis Teaching and Research Office, Henan Medical College
- Department of Nuclear Medicine, Zhengzhou Central Hospital of Zhengzhou University, Zhengzhou, China
| | - Ruixue Ma
- Imaging Diagnosis Teaching and Research Office, Henan Medical College
- Department of Nuclear Medicine, Zhengzhou Central Hospital of Zhengzhou University, Zhengzhou, China
| | - Jing Wu
- Imaging Diagnosis Teaching and Research Office, Henan Medical College
- Department of Nuclear Medicine, Zhengzhou Central Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
29
|
Sakuraba S, Koizumi A, Iwasawa T, Ito T, Kato K. Serum EphA2 as a Promising Biomarker for the Early Detection and Diagnosis of Colorectal Cancer. Biomolecules 2024; 14:1504. [PMID: 39766211 PMCID: PMC11673381 DOI: 10.3390/biom14121504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 11/19/2024] [Accepted: 11/24/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND EphA2, a receptor-type tyrosine kinase, is overexpressed in several cancers, including colorectal cancer (CRC), and can be detected as soluble EphA2 in serum. This study aimed to investigate the relationship between soluble EphA2 and CRC. METHODS Serum samples were collected from 65 patients with CRC and 19 healthy individuals. Time-series changes in soluble EphA2 levels were measured in CRC cell lines to verify the release of EphA2 into the culture medium. RESULTS Soluble EphA2 levels were significantly higher in patients than in healthy individuals (p < 0.0001). Specifically, even in early-stage cancer, there was a notable difference between healthy individuals and patients with Stage I CRC (p = 0.00298), highlighting the potential of EphA2 as a biomarker for early detection. Additionally, correlations were observed with tumor size (p = 0.0346), depth of invasion (p = 0.0311), and lymphatic invasion (p = 0.0431). A receiver operating characteristic (ROC) analysis yielded an area under the curve (AUC) of 0.90 with 93.8% sensitivity and 78.9% specificity at a cutoff value of 448 pg/mL. CONCLUSIONS These findings suggest that serum EphA2 could serve as a valuable biomarker for the early detection of CRC, offering a practical and minimally invasive alternative to conventional tumor markers.
Collapse
Affiliation(s)
- Shunsuke Sakuraba
- Department of Surgery, Juntendo University Medical School, Shizuoka Hospital, Shizuoka 410-2295, Japan; (S.S.); (T.I.)
- Shizuoka Medical Research Center for Disaster, Shizuoka 410-2295, Japan;
| | - Akihiro Koizumi
- Department of Surgery, Juntendo University Medical School, Shizuoka Hospital, Shizuoka 410-2295, Japan; (S.S.); (T.I.)
| | - Takumi Iwasawa
- Shizuoka Medical Research Center for Disaster, Shizuoka 410-2295, Japan;
- Institute of Life Innovation Studies, Toyo University, Tokyo 115-8650, Japan
| | - Tomoaki Ito
- Department of Surgery, Juntendo University Medical School, Shizuoka Hospital, Shizuoka 410-2295, Japan; (S.S.); (T.I.)
- Shizuoka Medical Research Center for Disaster, Shizuoka 410-2295, Japan;
| | - Kazunori Kato
- Institute of Life Innovation Studies, Toyo University, Tokyo 115-8650, Japan
- Department of Nutrition Sciences, Graduate School of Health and Sports Sciences, Toyo University, Tokyo 115-8650, Japan
| |
Collapse
|
30
|
Kędzierska M, Bańkosz M. Role of Proteins in Oncology: Advances in Cancer Diagnosis, Prognosis, and Targeted Therapy-A Narrative Review. J Clin Med 2024; 13:7131. [PMID: 39685591 DOI: 10.3390/jcm13237131] [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: 10/27/2024] [Revised: 11/19/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Modern oncology increasingly relies on the role of proteins as key components in cancer diagnosis, prognosis, and targeted therapy. This review examines advancements in protein biomarkers across several cancer types, including breast cancer, lung cancer, ovarian cancer, and hepatocellular carcinoma. These biomarkers have proven critical for early detection, treatment response monitoring, and tailoring personalized therapeutic strategies. The article highlights the utility of targeted therapies, such as tyrosine kinase inhibitors and monoclonal antibodies, in improving treatment efficacy while minimizing systemic toxicity. Despite these advancements, challenges like tumor resistance, variability in protein expression, and diagnostic heterogeneity persist, complicating universal application. The review underscores future directions, including the integration of artificial intelligence, advanced protein analysis technologies, and the development of combination therapies to overcome these barriers and refine personalized cancer treatment.
Collapse
Affiliation(s)
- Magdalena Kędzierska
- Department of Chemotherapy, Medical University of Lodz, Copernicus Memorial Hospital of Lodz, 90-549 Lodz, Poland
| | - Magdalena Bańkosz
- CUT Doctoral School, Faculty of Materials Engineering and Physics, Department of Material Engineering, Cracow University of Technology, 37 Jana Pawla II Av., 31-864 Krakow, Poland
| |
Collapse
|
31
|
Habeeb IF, Alao TE, Delgado D, Buffone A. When a negative (charge) is not a positive: sialylation and its role in cancer mechanics and progression. Front Oncol 2024; 14:1487306. [PMID: 39628991 PMCID: PMC11611868 DOI: 10.3389/fonc.2024.1487306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 10/10/2024] [Indexed: 12/06/2024] Open
Abstract
Sialic acids and sialoglycans are critical actors in cancer progression and metastasis. These terminal sugar residues on glycoproteins and glycolipids modulate key cellular processes such as immune evasion, cell adhesion, and migration. Aberrant sialylation is driven by overexpression of sialyltransferases, resulting in hypersialylation on cancer cell surfaces as well as enhancing tumor aggressiveness. Sialylated glycans alter the structure of the glycocalyx, a protective barrier that fosters cancer cell detachment, migration, and invasion. This bulky glycocalyx also increases membrane tension, promoting integrin clustering and downstream signaling pathways that drive cell proliferation and metastasis. They play a critical role in immune evasion by binding to Siglecs, inhibitory receptors on immune cells, which transmit signals that protect cancer cells from immune-mediated destruction. Targeting sialylation pathways presents a promising therapeutic opportunity to understand the complex roles of sialic acids and sialoglycans in cancer mechanics and progression, which is crucial for developing novel diagnostic and therapeutic strategies that can disrupt these processes and improve cancer treatment outcomes.
Collapse
Affiliation(s)
- Issa Funsho Habeeb
- Department of Biomedical Engineering, New Jersey Institute of Technlogy, Newark, NJ, United States
| | - Toheeb Eniola Alao
- Department of Biomedical Engineering, New Jersey Institute of Technlogy, Newark, NJ, United States
| | - Daniella Delgado
- Department of Biomedical Engineering, New Jersey Institute of Technlogy, Newark, NJ, United States
| | - Alexander Buffone
- Department of Biomedical Engineering, New Jersey Institute of Technlogy, Newark, NJ, United States
- Chemical and Materials Engineering, New Jersey Institute of Technlogy, Newark, NJ, United States
| |
Collapse
|
32
|
Trentin L, Basile D, Lazzari E, Fietta E, Rossi A, Graziani F, Cappetta A, Simionato F, D'Amore E, Perbellini O, Aprile G. Implementation of a MSRE ddPCR method for the detection of methylated WIF1 and NPY genes in colorectal cancer patients. TUMORI JOURNAL 2024; 110:375-385. [PMID: 39101541 DOI: 10.1177/03008916241261675] [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: 08/06/2024]
Abstract
BACKGROUND Colorectal cancer is a worldwide leading cause of death accounting for high-rate mortality. Mutations in the epidermal growth factor receptor and RAS/MAPK pathways, as well as altered methylation genes profiles, have been described as molecular mechanisms promoting and sustaining tumour development and progression. Aberrant methylation is a well-known epigenetic mechanism involved in gene regulation; particularly several genes were reported as hypermethylated in CRC. Recently, it was shown that epigenetic alterations in genes such as neuropeptide y, proenkephalin and Wnt inhibitory factor 1 can be used as promising disease biomarkers. Almost all methods developed for the DNA methylation analysis combined next generation sequencing, conventional qRT-PCR or ddPCR with the prior DNA modification with sodium bisulfite. METHODS AND RESULTS We implemented a ddPCR method to assess the methylation status of Wnt inhibitory factor 1 and neuropeptide y using the methylation sensitive restriction enzyme approach that does not impact on DNA quality and guarantees the discrimination of DNA methylation independent of bisulfite conversion. CONCLUSIONS We showed that this method is robust and sensitive also allowing the monitoring of CRC disease progression when applied to circulating free DNA samples from liquid biopsies, proving to be a fast and easy to implement assay to be used for the monitoring of the methylation pattern of clinically relevant target genes.
Collapse
Affiliation(s)
- Luca Trentin
- AULSS 8 Berica Ospedale San Bortolo, Laboratory of Haematology, Vicenza, Italy
- AULSS 2 Marca Trevigiana, Histocompatibility Laboratory, Treviso, Italy
| | - Debora Basile
- AULSS 8 Berica Ospedale San Bortolo, UOC Oncologia, Vicenza, Italy
- Unit of Medical Oncology, Ospedale San Giovanni di Dio, Crotone, Italy
| | - Elena Lazzari
- AULSS 8 Berica Ospedale San Bortolo, UOC Anatomia Patologica, Vicenza, Italy
| | - Elena Fietta
- AULSS 8 Berica Ospedale San Bortolo, Laboratory of Haematology, Vicenza, Italy
| | - Alice Rossi
- AULSS 8 Berica Ospedale San Bortolo, UOC Oncologia, Vicenza, Italy
| | | | | | | | - Emanuele D'Amore
- AULSS 8 Berica Ospedale San Bortolo, UOC Anatomia Patologica, Vicenza, Italy
| | - Omar Perbellini
- AULSS 8 Berica Ospedale San Bortolo, Laboratory of Haematology, Vicenza, Italy
| | - Giuseppe Aprile
- AULSS 8 Berica Ospedale San Bortolo, UOC Oncologia, Vicenza, Italy
| |
Collapse
|
33
|
Tang H, Li YX, Lian JJ, Ng HY, Wang SSY. Personalized treatment using predictive biomarkers in solid organ malignancies: A review. TUMORI JOURNAL 2024; 110:386-404. [PMID: 39091157 DOI: 10.1177/03008916241261484] [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: 08/04/2024]
Abstract
In recent years, the influence of specific biomarkers in the diagnosis and prognosis of solid organ malignancies has been increasingly prominent. The relevance of the use of predictive biomarkers, which predict cancer response to specific forms of treatment provided, is playing a more significant role than ever before, as it affects diagnosis and initiation of treatment, monitoring for efficacy and side effects of treatment, and adjustment in treatment regimen in the long term. In the current review, we explored the use of predictive biomarkers in the treatment of solid organ malignancies, including common cancers such as colorectal cancer, breast cancer, lung cancer, prostate cancer, and cancers associated with high mortalities, such as pancreatic cancer, liver cancer, kidney cancer and cancers of the central nervous system. We additionally analyzed the goals and types of personalized treatment using predictive biomarkers, and the management of various types of solid organ malignancies using predictive biomarkers and their relative efficacies so far in the clinical settings.
Collapse
|
34
|
González A, Badiola I, Fullaondo A, Rodríguez J, Odriozola A. Personalised medicine based on host genetics and microbiota applied to colorectal cancer. ADVANCES IN GENETICS 2024; 112:411-485. [PMID: 39396842 DOI: 10.1016/bs.adgen.2024.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Colorectal cancer (CRC) ranks second in incidence and third in cancer mortality worldwide. This situation, together with the understanding of the heterogeneity of the disease, has highlighted the need to develop a more individualised approach to its prevention, diagnosis and treatment through personalised medicine. This approach aims to stratify patients according to risk, predict disease progression and determine the most appropriate treatment. It is essential to identify patients who may respond adequately to treatment and those who may be resistant to treatment to avoid unnecessary therapies and minimise adverse side effects. Current research is focused on identifying biomarkers such as specific mutated genes, the type of mutations and molecular profiles critical for the individualisation of CRC diagnosis, prognosis and treatment guidance. In addition, the study of the intestinal microbiota as biomarkers is being incorporated due to the growing scientific evidence supporting its influence on this disease. This article comprehensively addresses the use of current and emerging diagnostic, prognostic and predictive biomarkers in precision medicine against CRC. The effects of host genetics and gut microbiota composition on new approaches to treating this disease are discussed. How the gut microbiota could mitigate the side effects of treatment is reviewed. In addition, strategies to modulate the gut microbiota, such as dietary interventions, antibiotics, and transplantation of faecal microbiota and phages, are discussed to improve CRC prevention and treatment. These findings provide a solid foundation for future research and improving the care of CRC patients.
Collapse
Affiliation(s)
- Adriana González
- Hologenomics Research Group, Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country, Spain
| | - Iker Badiola
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Asier Fullaondo
- Hologenomics Research Group, Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country, Spain
| | | | - Adrian Odriozola
- Hologenomics Research Group, Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country, Spain.
| |
Collapse
|
35
|
Van Rossum HH, Holdenrieder S, Yun YM, Patel D, Thelen M, Song J, Unsworth N, Partridge K, Moore M, Cui W, Ramanathan L, Meng QH, Ballieux BEPB, Sturgeon C, Vesper H. External quality assessment-based tumor marker harmonization simulation; insights in achievable harmonization for CA 15-3 and CEA. Clin Chem Lab Med 2024:cclm-2024-0696. [PMID: 39299928 DOI: 10.1515/cclm-2024-0696] [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: 06/14/2024] [Accepted: 08/31/2024] [Indexed: 09/22/2024]
Abstract
OBJECTIVES CA 15-3 and CEA are tumor markers used in routine clinical care for breast cancer and colorectal cancer, among others. Current measurement procedures (MP) for these tumor markers are considered to be insufficiently harmonized. This study investigated the achievable harmonization for CA 15-3 and CEA by using an in silico simulation of external quality assessment (EQA) data from multiple EQA programs using patient-pool based samples. METHODS CA 15-3 and CEA data from SKML (2021), UK NEQAS (2020-2021) and KEQAS (2020-2021) were used. A harmonization protocol was defined in which MPs that were considered equivalent were used to value assign EQA samples, and recalibration was only required if the MP had a bias of >5 % with value assigned EQA. Harmonization status was assessed by determining the mean level of agreement and residual variation by CV (%). RESULTS Only MPs from Abbott, Beckman, Roche and Siemens were available in all EQA programs. For CA 15-3, recalibration was proposed for Beckman MP only and for CEA, recalibration was proposed for Siemens MP only. When the harmonization procedures were applied, for CA 15-3 the pre-harmonization mean bias range per MP was reduced from -29.28 to 9.86 %, into -0.09-0.12 % after harmonization. For CEA, the mean bias range per MP was reduced from -23.78 to 2.00 % pre-harmonization to -3.13-1.42 % post-harmonization. CONCLUSIONS The present study suggests that a significant improvement in the harmonization status of CA 15-3 and CEA may be achieved by recalibration of a limited number of MPs.
Collapse
Affiliation(s)
- Huub H Van Rossum
- Department of Laboratory Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Stefan Holdenrieder
- Institute of Laboratory Medicine, Munich Biomarker Research Center, Deutsches Herzzentrum, Munich, Germany
| | - Yeo-Min Yun
- Department of Laboratory Medicine, Konkuk University Medical Center, Seoul, Republic of South Korea
| | - Dina Patel
- UK NEQAS Immunology, Immunochemistry & Allergy, Northern General Hospital, Sheffield, UK
| | - Marc Thelen
- SKML, Nijmegen, The Netherlands
- Department of Laboratory Medicine of the Radboud University Medical Center, Nijmegen, The Netherlands
| | - Junghan Song
- Department of Laboratory Medicine, Seoul National University Bundang Hospital and College of Medicine, Seongnam, Republic of South Korea
| | - Nick Unsworth
- UK NEQAS [Edinburgh], Department of Laboratory Medicine, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Katherine Partridge
- Therapeutic Reference Materials,Medicines and Healthcare Products Regulatory Agency (MHRA), South Mimms, Hertfordshire, UK
| | - Melanie Moore
- Therapeutic Reference Materials,Medicines and Healthcare Products Regulatory Agency (MHRA), South Mimms, Hertfordshire, UK
| | - Wei Cui
- Department of Laboratory Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lakshmi Ramanathan
- Clinical Chemistry Service, Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Qing H Meng
- Department of Laboratory Medicine, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bart E P B Ballieux
- Department of Clinical Chemistry, Leiden University Medical Center, Leiden, The Netherlands
| | - Catharine Sturgeon
- UK NEQAS [Edinburgh], Department of Laboratory Medicine, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Hubert Vesper
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, GA, USA
| |
Collapse
|
36
|
Ishizaki T, Sugimoto M, Kuboyama Y, Mazaki J, Kasahara K, Tago T, Udo R, Iwasaki K, Hayashi Y, Nagakawa Y. Stage-Specific Plasma Metabolomic Profiles in Colorectal Cancer. J Clin Med 2024; 13:5202. [PMID: 39274416 PMCID: PMC11396754 DOI: 10.3390/jcm13175202] [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: 08/11/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/16/2024] Open
Abstract
Background/Objectives: The objective of this study was to investigate the metabolomic profiles of patients with colorectal cancer (CRC) across various stages of the disease. Methods: The plasma samples were obtained from 255 subjects, including patients with CRC in stages I-IV, polyps, and controls. We employed capillary electrophoresis time-of-flight mass spectrometry and liquid chromatography triple quadrupole mass spectrometry to analyze hydrophilic metabolites comprehensively. The data were randomly divided into two groups, and consistent differences observed in both groups were analyzed. Results: Acetylated polyamines, such as N1-acetylspermine and N1, N12-diacetylspermine, consistently showed elevated concentrations in stage IV compared to stages I-III. Non-acetylated polyamines, including spermine and spermidine, exhibited increasing trends from polyp to stage IV. Other metabolites, such as histidine and o-acetylcarnitine, showed decreasing trends across stages. While acetylated polyamines have been reported as CRC detection markers, our findings suggest that they also possess diagnostic potential for distinguishing stage IV from other stages. Conclusions: This study showed stage-specific changes in metabolic profiles, including polyamines, of colorectal cancer.
Collapse
Affiliation(s)
- Tetsuo Ishizaki
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku 160-0023, Tokyo, Japan
| | - Masahiro Sugimoto
- Institute of Medical Science, Tokyo Medical University, Shinjuku 160-8402, Tokyo, Japan
- Institute for Advanced Biosciences, Keio University, Tsuruoka 997-0052, Yamagata, Japan
| | - Yu Kuboyama
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku 160-0023, Tokyo, Japan
| | - Junichi Mazaki
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku 160-0023, Tokyo, Japan
| | - Kenta Kasahara
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku 160-0023, Tokyo, Japan
| | - Tomoya Tago
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku 160-0023, Tokyo, Japan
| | - Ryutaro Udo
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku 160-0023, Tokyo, Japan
| | - Kenichi Iwasaki
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku 160-0023, Tokyo, Japan
| | - Yutaka Hayashi
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku 160-0023, Tokyo, Japan
| | - Yuichi Nagakawa
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku 160-0023, Tokyo, Japan
| |
Collapse
|
37
|
Fuller RN, Morcos A, Bustillos JG, Molina DC, Wall NR. Small non-coding RNAs and pancreatic ductal adenocarcinoma: Linking diagnosis, pathogenesis, drug resistance, and therapeutic potential. Biochim Biophys Acta Rev Cancer 2024; 1879:189153. [PMID: 38986720 DOI: 10.1016/j.bbcan.2024.189153] [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: 01/22/2024] [Revised: 07/03/2024] [Accepted: 07/05/2024] [Indexed: 07/12/2024]
Abstract
This review comprehensively investigates the intricate interplay between small non-coding RNAs (sncRNAs) and pancreatic ductal adenocarcinoma (PDAC), a devastating malignancy with limited therapeutic options. Our analysis reveals the pivotal roles of sncRNAs in various facets of PDAC biology, spanning diagnosis, pathogenesis, drug resistance, and therapeutic strategies. sncRNAs have emerged as promising biomarkers for PDAC, demonstrating distinct expression profiles in diseased tissues. sncRNA differential expression patterns, often detectable in bodily fluids, hold potential for early and minimally invasive diagnostic approaches. Furthermore, sncRNAs exhibit intricate involvement in PDAC pathogenesis, regulating critical cellular processes such as proliferation, apoptosis, and metastasis. Additionally, mechanistic insights into sncRNA-mediated pathogenic pathways illuminate novel therapeutic targets and interventions. A significant focus of this review is dedicated to unraveling sncRNA mechanisms underlying drug resistance in PDAC. Understanding these mechanisms at the molecular level is imperative for devising strategies to overcome drug resistance. Exploring the therapeutic landscape, we discuss the potential of sncRNAs as therapeutic agents themselves as their ability to modulate gene expression with high specificity renders them attractive candidates for targeted therapy. In summary, this review integrates current knowledge on sncRNAs in PDAC, offering a holistic perspective on their diagnostic, pathogenic, and therapeutic relevance. By elucidating the roles of sncRNAs in PDAC biology, this review provides valuable insights for the development of novel diagnostic tools and targeted therapeutic approaches, crucial for improving the prognosis of PDAC patients.
Collapse
Affiliation(s)
- Ryan N Fuller
- Department of Basic Science, Division of Biochemistry, Center for Health Disparity and Mol. Med., Loma Linda University, Loma Linda, CA 92350, USA; Department of Radiation Medicine, James M. Slater, MD Proton Treatment and Research Center, Loma Linda University, Loma Linda, CA 92350, USA
| | - Ann Morcos
- Department of Basic Science, Division of Biochemistry, Center for Health Disparity and Mol. Med., Loma Linda University, Loma Linda, CA 92350, USA; Department of Radiation Medicine, James M. Slater, MD Proton Treatment and Research Center, Loma Linda University, Loma Linda, CA 92350, USA
| | - Joab Galvan Bustillos
- Department of Basic Science, Division of Biochemistry, Center for Health Disparity and Mol. Med., Loma Linda University, Loma Linda, CA 92350, USA; Division of Surgical Oncology, Department of Surgery, Loma Linda University, Loma Linda, CA 92350, USA
| | - David Caba Molina
- Division of Surgical Oncology, Department of Surgery, Loma Linda University, Loma Linda, CA 92350, USA
| | - Nathan R Wall
- Department of Basic Science, Division of Biochemistry, Center for Health Disparity and Mol. Med., Loma Linda University, Loma Linda, CA 92350, USA; Department of Radiation Medicine, James M. Slater, MD Proton Treatment and Research Center, Loma Linda University, Loma Linda, CA 92350, USA.
| |
Collapse
|
38
|
Hertz DL, Bousman CA, McLeod HL, Monte AA, Voora D, Orlando LA, Crutchley RD, Brown B, Teeple W, Rogers S, Patel JN. Recommendations for pharmacogenetic testing in clinical practice guidelines in the US. Am J Health Syst Pharm 2024; 81:672-683. [PMID: 38652504 DOI: 10.1093/ajhp/zxae110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Indexed: 04/25/2024] Open
Abstract
PURPOSE Pharmacogenetic testing can identify patients who may benefit from personalized drug treatment. However, clinical uptake of pharmacogenetic testing has been limited. Clinical practice guidelines recommend biomarker tests that the guideline authors deem to have demonstrated clinical utility, meaning that testing improves treatment outcomes. The objective of this narrative review is to describe the current status of pharmacogenetic testing recommendations within clinical practice guidelines in the US. SUMMARY Guidelines were reviewed for pharmacogenetic testing recommendations for 21 gene-drug pairs that have well-established drug response associations and all of which are categorized as clinically actionable by the Clinical Pharmacogenetics Implementation Consortium. The degree of consistency within and between organizations in pharmacogenetic testing recommendations was assessed. Relatively few clinical practice guidelines that provide a pharmacogenetic testing recommendation were identified. Testing recommendations for HLA-B*57:01 before initiation of abacavir and G6PD before initiation of rasburicase, both of which are included in drug labeling, were mostly consistent across guidelines. Gene-drug pairs with at least one clinical practice guideline recommending testing or stating that testing could be considered included CYP2C19-clopidogrel, CYP2D6-codeine, CYP2D6-tramadol, CYP2B6-efavirenz, TPMT-thiopurines, and NUDT15-thiopurines. Testing recommendations for the same gene-drug pair were often inconsistent between organizations and sometimes inconsistent between different guidelines from the same organization. CONCLUSION A standardized approach to evaluating the evidence of clinical utility for pharmacogenetic testing may increase the inclusion and consistency of pharmacogenetic testing recommendations in clinical practice guidelines, which could benefit patients and society by increasing clinical use of pharmacogenetic testing.
Collapse
Affiliation(s)
- Daniel L Hertz
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Chad A Bousman
- Department of Medical Genetics, University of Calgary, Calgary, AB, Canada
| | - Howard L McLeod
- Center for Precision Medicine and Functional Genomics, Utah Tech University, St. George, UT, USA
| | - Andrew A Monte
- Section of Pharmacology & Medical Toxicology, Department of Emergency Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Deepak Voora
- Duke Precision Medicine Program, Department of Medicine, Duke University, Durham, NC, USA
| | - Lori A Orlando
- Department of Medicine, Duke University, Durham, NC, USA
| | - Rustin D Crutchley
- Department of Pharmaceutical Sciences, College of Pharmacy, Manchester University, Fort Wayne, IN, USA
| | | | | | - Sara Rogers
- American Society of Pharmacovigilance, Houston, TX, and Texas A&M Health Science Center, Bryan, TX, USA
| | - Jai N Patel
- Department of Cancer Pharmacology and Pharmacogenomics, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA and Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, USA
| |
Collapse
|
39
|
Shah Y, Dahiya DS, Tiwari A, Kumar H, Gangwani MK, Ali H, Hayat U, Alsakarneh S, Singh S, Malik S, Sohail AH, Chandan S, Ali MA, Inamdar S. Advancements in Early Detection and Screening Strategies for Pancreatic Cancer: From Genetic Susceptibility to Novel Biomarkers. J Clin Med 2024; 13:4706. [PMID: 39200847 PMCID: PMC11355237 DOI: 10.3390/jcm13164706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 08/05/2024] [Accepted: 08/08/2024] [Indexed: 09/02/2024] Open
Abstract
Pancreatic cancer is a rare but lethal cancer due to its biologically aggressive nature, advanced stage at the time of diagnosis, and poor response to oncologic therapies. The risk of pancreatic cancer is significantly higher to 5% in certain high-risk individuals with inherited genetic susceptibility. Screening for pancreatic cancer in these individuals from high-risk groups can help with the early detection of pancreatic cancer as well as the detection of precursor lesions leading to early surgical resection and improved overall outcomes. The advancements in radiological imaging as well as advanced endoscopic procedures has made a significant impact on the early diagnosis, surveillance, and staging of pancreatic cancer. There is also a significant advancement in the development of biomarkers for the early detection of pancreatic cancer, which has also led to the development of liquid biopsy, allowing for microRNA detection in serum and circulating tumor cells. Various societies and organizations have provided guidelines for pancreatic cancer screening and surveillance in high-risk individuals. In this review, we aim to discuss the hereditary risk factors for developing pancreatic cancer, summarize the screening recommendations by different societies, and discuss the development of novel biomarkers and areas for future research in pancreatic cancer screening for high-risk individuals.
Collapse
Affiliation(s)
- Yash Shah
- Department of Internal Medicine, Trinity Health Oakland/Wayne State University, Pontiac, MI 48341, USA
| | - Dushyant Singh Dahiya
- Division of Gastroenterology, Hepatology & Motility, The University of Kansas School of Medicine, Kansas City, KS 66160, USA
| | - Angad Tiwari
- Department of Internal Medicine, Maharani Laxmi Bai Medical College, Jhansi 284001, Uttar Pradesh, India
| | - Harendra Kumar
- Department of Internal Medicine, Dow University of Health Sciences, Karachi 74200, Pakistan
| | - Manesh Kumar Gangwani
- Department of Gastroenterology and Hepatology, University of Arkansas For Medical Sciences, Little Rock, AR 72205, USA
| | - Hassam Ali
- Division of Gastroenterology, Hepatology & Nutrition, East Carolina University/Brody School of Medicine, Greenville, NC 27834, USA
| | - Umar Hayat
- Department of Internal Medicine, Geisinger Wyoming Valley Medical Center, Wilkes Barre, PA 18711, USA
| | - Saqr Alsakarneh
- Department of Internal Medicine, University of Missouri-Kansas City, Kansas City, MO 64110, USA
| | - Sahib Singh
- Department of Internal Medicine, Sinai Hospital, Baltimore, MD 21215, USA
| | - Sheza Malik
- Department of Internal Medicine, Rochester General Hospital, Rochester, NY 14621, USA
| | - Amir H. Sohail
- Department of Surgery, University of New Mexico, Albuquerque, NM 87131, USA
| | - Saurabh Chandan
- Center for Interventional Endoscopy (CIE), Advent Health, Orlando, FL 32803, USA
| | - Meer A. Ali
- Department of Gastroenterology and Hepatology, University of Arkansas For Medical Sciences, Little Rock, AR 72205, USA
| | - Sumant Inamdar
- Department of Gastroenterology and Hepatology, University of Arkansas For Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
40
|
Martínez-Espartosa D, Alegre E, Casero-Ramírez H, Díaz-Garzón J, Fernández-Calle P, Fuentes-Bullejos P, Varo N, González Á. Clinical utility of personalized reference intervals for CEA in the early detection of oncologic disease. Clin Chem Lab Med 2024; 0:cclm-2024-0546. [PMID: 39101454 DOI: 10.1515/cclm-2024-0546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/02/2024] [Indexed: 08/06/2024]
Abstract
OBJECTIVES Personalized reference intervals (prRI) have been proposed as a diagnostic tool for assessing measurands with high individuality. Here, we evaluate clinical performance of prRI using carcinoembryonic antigen (CEA) for cancer detection and compare it with that of reference change values (RCV) and other criteria recommended by clinical guidelines (e.g. 25 % of change between consecutive CEA results (RV25) and the cut-off point of 5 μg/L (CP5)). METHODS Clinical and analytical data from 2,638 patients collected over 19 years were retrospectively evaluated. A total 15,485 CEA results were studied. For each patient, we calculated prRI and RCV using computer algorithms based on the combination of different strategies to assess the number of CEA results needed, consideration of one or two limits of reference interval and the intraindividual biological variation estimate (CVI) used: (a) publicly available (CVI-EU), (b) CVI calculated using an indirect method (CVI-NOO) and (c) within-person BV (CVP). For each new result identified falling outside the prRI, exceeding the RCV interval, RV25 or CP5, we searched for records identifying the presence of tumour at 3 and 12 months after the test. The sensitivity, specificity and predictive power of each strategy were calculated. RESULTS PrRI approaches derived using CVI-EU, and both limits of reference interval achieve the best sensitivity (87.5 %) and NPV (99.3 %) at 3 and 12 months of all evaluated criteria. Only 3 results per patients are enough to calculate prRIs that reach this diagnostic performance. CONCLUSIONS PrRI approaches could be an effective tool to rule out new oncological findings during the active surveillance of patients.
Collapse
Affiliation(s)
| | - Estíbaliz Alegre
- Biochemistry Department, Clínica Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | | | - Jorge Díaz-Garzón
- Department of Laboratory Medicine, Hospital Universitario La Paz, Madrid, Spain
| | | | | | - Nerea Varo
- Biochemistry Department, Clínica Universidad de Navarra, Pamplona, Spain
| | - Álvaro González
- Biochemistry Department, Clínica Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| |
Collapse
|
41
|
Fabijanec M, Hulina-Tomašković A, Štefanović M, Verbanac D, Ćelap I, Somborac-Bačura A, Grdić Rajković M, Demirović A, Ramić S, Krušlin B, Rumora L, Čeri A, Koržinek M, Petrik J, Ljubičić N, Baršić N, Barišić K. MicroRNA-193a-3p as a Valuable Biomarker for Discriminating between Colorectal Cancer and Colorectal Adenoma Patients. Int J Mol Sci 2024; 25:8156. [PMID: 39125725 PMCID: PMC11311302 DOI: 10.3390/ijms25158156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/16/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Specific markers for colorectal cancer (CRC), preceded by colorectal adenoma (pre-CRC), are lacking. This study aimed to investigate whether microRNAs (miR-19a-3p, miR-92a-3p, miR-193a-3p, and miR-210-3p) from tissues and exosomes are potential CRC biomarkers and compare them to existing biomarkers, namely carcinoembryonic antigen (CEA) and carbohydrate antigen (CA) 19-9. MiRNA was isolated in the samples of 52 CRC and 76 pre-CRC patients. Expression levels were analyzed by RT-qPCR. When comparing pre-CRC and CRC tissue expression levels, only miR-193a-3p showed statistically significant result (p < 0.0001). When comparing the tissues and exosomes of CRC samples, a statistically significant difference was found for miR-193a-3p (p < 0.0001), miR-19a-3p (p < 0.0001), miR-92a-3p (p = 0.0212), and miR-210-3p (p < 0.0001). A receiver-operating characteristic (ROC) curve and area under the ROC curve (AUC) were used to evaluate the diagnostic value of CEA, CA 19-9, and miRNAs. CEA and CA 19-9 had good diagnostic values (AUCs of 0.798 and 0.668). The diagnostic value only of miR-193a-3p was highlighted (AUC = 0.725). The final logistic regression model, in which we put a combination of CEA concentration and the miR-193a-3p expression level in tissues, showed that using these two markers can distinguish CRC and pre-CRC in 71.3% of cases (AUC = 0.823). MiR-193a-3p from tissues could be a potential CRC biomarker.
Collapse
Affiliation(s)
- Marija Fabijanec
- Center for Applied Medical Biochemistry, Faculty of Pharmacy and Biochemistry, University of Zagreb, 10000 Zagreb, Croatia;
| | - Andrea Hulina-Tomašković
- Department of Medical Biochemistry and Hematology, Faculty of Pharmacy and Biochemistry, University of Zagreb, 10000 Zagreb, Croatia; (M.Š.); (D.V.); (I.Ć.); (A.S.-B.); (M.G.R.); (L.R.); (A.Č.); (M.K.); (J.P.); (K.B.)
| | - Mario Štefanović
- Department of Medical Biochemistry and Hematology, Faculty of Pharmacy and Biochemistry, University of Zagreb, 10000 Zagreb, Croatia; (M.Š.); (D.V.); (I.Ć.); (A.S.-B.); (M.G.R.); (L.R.); (A.Č.); (M.K.); (J.P.); (K.B.)
- Department of Clinical Chemistry, Sestre Milosrdnice University Hospital Center, 10000 Zagreb, Croatia
| | - Donatella Verbanac
- Department of Medical Biochemistry and Hematology, Faculty of Pharmacy and Biochemistry, University of Zagreb, 10000 Zagreb, Croatia; (M.Š.); (D.V.); (I.Ć.); (A.S.-B.); (M.G.R.); (L.R.); (A.Č.); (M.K.); (J.P.); (K.B.)
| | - Ivana Ćelap
- Department of Medical Biochemistry and Hematology, Faculty of Pharmacy and Biochemistry, University of Zagreb, 10000 Zagreb, Croatia; (M.Š.); (D.V.); (I.Ć.); (A.S.-B.); (M.G.R.); (L.R.); (A.Č.); (M.K.); (J.P.); (K.B.)
- Department of Clinical Chemistry, Sestre Milosrdnice University Hospital Center, 10000 Zagreb, Croatia
| | - Anita Somborac-Bačura
- Department of Medical Biochemistry and Hematology, Faculty of Pharmacy and Biochemistry, University of Zagreb, 10000 Zagreb, Croatia; (M.Š.); (D.V.); (I.Ć.); (A.S.-B.); (M.G.R.); (L.R.); (A.Č.); (M.K.); (J.P.); (K.B.)
| | - Marija Grdić Rajković
- Department of Medical Biochemistry and Hematology, Faculty of Pharmacy and Biochemistry, University of Zagreb, 10000 Zagreb, Croatia; (M.Š.); (D.V.); (I.Ć.); (A.S.-B.); (M.G.R.); (L.R.); (A.Č.); (M.K.); (J.P.); (K.B.)
| | - Alma Demirović
- Department of Pathology and Cytology “Ljudevit Jurak”, Sestre Milosrdnice University Hospital Center, 10000 Zagreb, Croatia; (A.D.); (B.K.)
| | - Snježana Ramić
- Department of Oncological Pathology, University Hospital for Tumors, Sestre Milosrdnice University Hospital Centre, 10000 Zagreb, Croatia;
| | - Božo Krušlin
- Department of Pathology and Cytology “Ljudevit Jurak”, Sestre Milosrdnice University Hospital Center, 10000 Zagreb, Croatia; (A.D.); (B.K.)
| | - Lada Rumora
- Department of Medical Biochemistry and Hematology, Faculty of Pharmacy and Biochemistry, University of Zagreb, 10000 Zagreb, Croatia; (M.Š.); (D.V.); (I.Ć.); (A.S.-B.); (M.G.R.); (L.R.); (A.Č.); (M.K.); (J.P.); (K.B.)
| | - Andrea Čeri
- Department of Medical Biochemistry and Hematology, Faculty of Pharmacy and Biochemistry, University of Zagreb, 10000 Zagreb, Croatia; (M.Š.); (D.V.); (I.Ć.); (A.S.-B.); (M.G.R.); (L.R.); (A.Č.); (M.K.); (J.P.); (K.B.)
| | - Martha Koržinek
- Department of Medical Biochemistry and Hematology, Faculty of Pharmacy and Biochemistry, University of Zagreb, 10000 Zagreb, Croatia; (M.Š.); (D.V.); (I.Ć.); (A.S.-B.); (M.G.R.); (L.R.); (A.Č.); (M.K.); (J.P.); (K.B.)
| | - József Petrik
- Department of Medical Biochemistry and Hematology, Faculty of Pharmacy and Biochemistry, University of Zagreb, 10000 Zagreb, Croatia; (M.Š.); (D.V.); (I.Ć.); (A.S.-B.); (M.G.R.); (L.R.); (A.Č.); (M.K.); (J.P.); (K.B.)
| | - Neven Ljubičić
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Sestre Milosrdnice University Hospital Center, 10000 Zagreb, Croatia; (N.L.)
| | - Neven Baršić
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Sestre Milosrdnice University Hospital Center, 10000 Zagreb, Croatia; (N.L.)
| | - Karmela Barišić
- Department of Medical Biochemistry and Hematology, Faculty of Pharmacy and Biochemistry, University of Zagreb, 10000 Zagreb, Croatia; (M.Š.); (D.V.); (I.Ć.); (A.S.-B.); (M.G.R.); (L.R.); (A.Č.); (M.K.); (J.P.); (K.B.)
| |
Collapse
|
42
|
Bockorny B, Muthuswamy L, Huang L, Hadisurya M, Lim CM, Tsai LL, Gill RR, Wei JL, Bullock AJ, Grossman JE, Besaw RJ, Narasimhan S, Tao WA, Perea S, Sawhney MS, Freedman SD, Hidalgo M, Iliuk A, Muthuswamy SK. A Large-Scale Proteomics Resource of Circulating Extracellular Vesicles for Biomarker Discovery in Pancreatic Cancer. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.03.13.23287216. [PMID: 36993200 PMCID: PMC10055460 DOI: 10.1101/2023.03.13.23287216] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Pancreatic cancer has the worst prognosis of all common tumors. Earlier cancer diagnosis could increase survival rates and better assessment of metastatic disease could improve patient care. As such, there is an urgent need to develop biomarkers to diagnose this deadly malignancy. Analyzing circulating extracellular vesicles (cEVs) using 'liquid biopsies' offers an attractive approach to diagnose and monitor disease status. However, it is important to differentiate EV-associated proteins enriched in patients with pancreatic ductal adenocarcinoma (PDAC) from those with benign pancreatic diseases such as chronic pancreatitis and intraductal papillary mucinous neoplasm (IPMN). To meet this need, we combined the novel EVtrap method for highly efficient isolation of EVs from plasma and conducted proteomics analysis of samples from 124 individuals, including patients with PDAC, benign pancreatic diseases and controls. On average, 912 EV proteins were identified per 100µL of plasma. EVs containing high levels of PDCD6IP, SERPINA12 and RUVBL2 were associated with PDAC compared to the benign diseases in both discovery and validation cohorts. EVs with PSMB4, RUVBL2 and ANKAR were associated with metastasis, and those with CRP, RALB and CD55 correlated with poor clinical prognosis. Finally, we validated a 7-EV protein PDAC signature against a background of benign pancreatic diseases that yielded an 89% prediction accuracy for the diagnosis of PDAC. To our knowledge, our study represents the largest proteomics profiling of circulating EVs ever conducted in pancreatic cancer and provides a valuable open-source atlas to the scientific community with a comprehensive catalogue of novel cEVs that may assist in the development of biomarkers and improve the outcomes of patients with PDAC.
Collapse
|
43
|
Fekry B, Ugartemendia L, Esnaola NF, Goetzl L. Extracellular Vesicles, Circadian Rhythms, and Cancer: A Comprehensive Review with Emphasis on Hepatocellular Carcinoma. Cancers (Basel) 2024; 16:2552. [PMID: 39061191 PMCID: PMC11274441 DOI: 10.3390/cancers16142552] [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: 06/20/2024] [Revised: 07/12/2024] [Accepted: 07/14/2024] [Indexed: 07/28/2024] Open
Abstract
This review comprehensively explores the complex interplay between extracellular vesicles (ECVs)/exosomes and circadian rhythms, with a focus on the role of this interaction in hepatocellular carcinoma (HCC). Exosomes are nanovesicles derived from cells that facilitate intercellular communication by transporting bioactive molecules such as proteins, lipids, and RNA/DNA species. ECVs are implicated in a range of diseases, where they play crucial roles in signaling between cells and their surrounding environment. In the setting of cancer, ECVs are known to influence cancer initiation and progression. The scope of this review extends to all cancer types, synthesizing existing knowledge on the various roles of ECVs. A unique aspect of this review is the emphasis on the circadian-controlled release and composition of exosomes, highlighting their potential as biomarkers for early cancer detection and monitoring metastasis. We also discuss how circadian rhythms affect multiple cancer-related pathways, proposing that disruptions in the circadian clock can alter tumor development and treatment response. Additionally, this review delves into the influence of circadian clock components on ECV biogenesis and their impact on reshaping the tumor microenvironment, a key component driving HCC progression. Finally, we address the potential clinical applications of ECVs, particularly their use as diagnostic tools and drug delivery vehicles, while considering the challenges associated with clinical implementation.
Collapse
Affiliation(s)
- Baharan Fekry
- McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (L.U.); (L.G.)
| | - Lierni Ugartemendia
- McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (L.U.); (L.G.)
| | - Nestor F. Esnaola
- Division of Surgical Oncology and Gastrointestinal Surgery, Department of Surgery, Houston Methodist Hospital, Houston, TX 77030, USA;
| | - Laura Goetzl
- McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (L.U.); (L.G.)
| |
Collapse
|
44
|
He D, Cui B, Lv H, Lu S, Zhu Y, Cheng Y, Dang L, Zhang H. Blood-Derived Extracellular Vesicles as a Promising Liquid Biopsy Diagnostic Tool for Early Cancer Detection. Biomolecules 2024; 14:847. [PMID: 39062561 PMCID: PMC11275243 DOI: 10.3390/biom14070847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
Cancer poses a significant public health challenge worldwide, and timely screening has the potential to mitigate cancer progression and reduce mortality rates. Currently, early identification of most tumors relies on imaging techniques and tissue biopsies. However, the use of low-cost, highly sensitive, non-invasive detection methods for early cancer screening has become more attractive. Extracellular Vesicles (EVs) released by all living cells contain distinctive biological components, such as nucleic acids, proteins, and lipids. These vesicles play crucial roles in the tumor microenvironment and intercellular communication during tumor progression, rendering liquid biopsy a particularly suitable method for diagnosis. Nevertheless, challenges related to purification methods and validation of efficacy currently hinder its widespread clinical implementation. These limitations underscore the importance of refining isolation techniques and conducting comprehensive investigations on EVs. This study seeks to evaluate the potential of liquid biopsy utilizing blood-derived EVs as a practical, cost-effective, and secure approach for early cancer detection.
Collapse
Affiliation(s)
- Dan He
- Laboratory of Animal Center, Medical Experiment Center, Shaanxi University of Chinese Medicine, Xianyang 712046, China; (D.H.); (S.L.); (Y.Z.)
| | - Bozhou Cui
- Department of Experimental Surgery, Tangdu Hospital, Fourth Military Medical University, Xi’an 710038, China;
| | - Hongkai Lv
- Department of Clinical Medicine of Second Clinical Medical School, Shaanxi University of Chinese Medicine, Xianyang 712046, China; (H.L.); (Y.C.)
| | - Shuxian Lu
- Laboratory of Animal Center, Medical Experiment Center, Shaanxi University of Chinese Medicine, Xianyang 712046, China; (D.H.); (S.L.); (Y.Z.)
| | - Yuan Zhu
- Laboratory of Animal Center, Medical Experiment Center, Shaanxi University of Chinese Medicine, Xianyang 712046, China; (D.H.); (S.L.); (Y.Z.)
| | - Yuqiang Cheng
- Department of Clinical Medicine of Second Clinical Medical School, Shaanxi University of Chinese Medicine, Xianyang 712046, China; (H.L.); (Y.C.)
| | - Lin Dang
- Basic Medical Academy, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Hong Zhang
- Laboratory of Animal Center, Medical Experiment Center, Shaanxi University of Chinese Medicine, Xianyang 712046, China; (D.H.); (S.L.); (Y.Z.)
| |
Collapse
|
45
|
He D, Wang K, Zhang Y, Jiang X, Chen H, Chen J, Liu D, Li G, Hu J, He X. Risk of advanced neoplasia after removal of colorectal adenomas with high-grade dysplasia. Surg Endosc 2024; 38:3783-3798. [PMID: 38806955 PMCID: PMC11219408 DOI: 10.1007/s00464-024-10898-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 05/02/2024] [Indexed: 05/30/2024]
Abstract
BACKGROUND Many studies reported the presence of adenomas with high-grade dysplasia (HGD) at index colonoscopy increased the incidence of advanced neoplasia (AN) and colorectal cancer (CRC) following. However, the conclusion remains obscure due to lack of studies on the specific population of adenomas with HGD. This study aimed to assess the long-term risk of AN and CRC after removal of adenomas with HGD. METHODS A total of 814 patients who underwent adenomas with HGD removal between 2010 and 2019 were retrospectively analyzed. The outcomes were the incidences of AN and CRC during surveillance colonoscopy. Cox proportional hazards models were utilized to identify risk factors associated with AN and CRC. RESULTS During more than 2000 person-years of follow-up, we found that AN and CRC incidence densities were 44.3 and 4.4 per 1000 person-years, respectively. The 10-year cumulative incidence of AN and CRC were 39.1% and 5.5%, respectively. In the multivariate model, synchronous low-risk polyps (HR 1.80, 95% CI 1.10-2.93) and synchronous high-risk polyps (HR 3.99, 95% CI 2.37-6.72) were risk factors for AN, whereas participation in surveillance colonoscopy visits (HR 0.56, 95% CI 0.36-0.88 for 1 visit; HR 0.10, 95% CI 0.06-0.19 for ≥ 2 visits) were associated with decreased AN incidence. Additionally, elevated baseline carcinoembryonic antigen (CEA) level (HR 10.19, 95% CI 1.77-58.59) was a risk factor for CRC, while participation in ≥ 2 surveillance colonoscopy visits (HR 0.11, 95% CI 0.02-0.56) were associated with decreased CRC incidence. Interestingly, for 11 patients who developed CRC after removal of adenomas with HGD, immunohistochemistry revealed that 8 cases (73%) were deficient mismatch repair CRCs. CONCLUSIONS Patients who have undergone adenoma with HGD removal are at higher risk of developing AN and CRC, while surveillance colonoscopy can reduce the risk. Patients with synchronous polyps, or with elevated baseline CEA level are considered high-risk populations and require more frequent surveillance.
Collapse
Affiliation(s)
- Degao He
- Department of Anorectal Surgery, Shenzhen Longhua District Central Hospital, Guanlan Avenue 187, Shenzhen, 518100, Guangdong, China.
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Road, Guangzhou, 510655, Guangdong, China.
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Road, Guangzhou, 510655, Guangdong, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Road, Guangzhou, 510655, Guangdong, China.
| | - Kai Wang
- Department of Anaesthesia, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Road, Guangzhou, 510655, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Road, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Road, Guangzhou, 510655, Guangdong, China
| | - Yanhong Zhang
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Road, Guangzhou, 510655, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Road, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Road, Guangzhou, 510655, Guangdong, China
| | - Xuefei Jiang
- Department of General Surgery (Institute of Gastroenterology), The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Road, Guangzhou, 510655, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Road, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Road, Guangzhou, 510655, Guangdong, China
| | - Hao Chen
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Road, Guangzhou, 510655, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Road, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Road, Guangzhou, 510655, Guangdong, China
| | - Junguo Chen
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Road, Guangzhou, 510655, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Road, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Road, Guangzhou, 510655, Guangdong, China
| | - Danlin Liu
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Road, Guangzhou, 510655, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Road, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Road, Guangzhou, 510655, Guangdong, China
| | - Guanman Li
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Road, Guangzhou, 510655, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Road, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Road, Guangzhou, 510655, Guangdong, China
| | - Jiancong Hu
- Department of General Surgery (Endoscopic Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Road, Guangzhou, 510655, Guangdong, China.
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Road, Guangzhou, 510655, Guangdong, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Road, Guangzhou, 510655, Guangdong, China.
| | - Xiaosheng He
- Department of General Surgery (Colorectal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Road, Guangzhou, 510655, Guangdong, China.
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Road, Guangzhou, 510655, Guangdong, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Road, Guangzhou, 510655, Guangdong, China.
| |
Collapse
|
46
|
Yuan C, Zeng L, Duan H, Suksatit B. Meta-analysis of the prognostic value of serum carcinoembryonic antigen in patients with colorectal cancer liver metastases after hepatectomy. Eur J Cancer Prev 2024; 33:334-346. [PMID: 37997904 DOI: 10.1097/cej.0000000000000859] [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: 11/25/2023]
Abstract
OBJECTIVES Carcinoembryonic antigen (CEA) is a broad-spectrum tumor marker for differential diagnosis, monitoring, and response assessment of a variety of malignancies. This meta-analysis was aimed at evaluating whether serum CEA could predict the prognosis in patients with colorectal cancer liver metastasis (CRCLM) before and after liver resection (LR). METHODS PubMed, Embase, Cochrane, and Web of Science were systematically searched to retrieve literature, with a search cutoff date of 27 February 2023. Articles were strictly screened for inclusion according to pre-specified inclusion and exclusion criteria. Data were pooled and analyzed using Stata 16.0. RESULTS This meta-analysis included 36 studies involving a total of 11 143 CRCLM patients. The results showed that a high pre-LR serum CEA level was correlated with poor overall survival (OS: HR = 1.61, 95% CI = 1.49-1.75, P < 0.001) and recurrence-free survival (RFS: HR = 1.27, 95% CI = 1.11-1.45, P < 0.001) in CRCLM patients. A high post-LR serum CEA level predicted poor overall survival (OS: HR = 2.66, 95% CI = 2.10-3.38, P < 0.001). CONCLUSION High preoperative and postoperative serum CEA levels in patients with CRCLM were significantly associated with poor prognosis, independent of treatment modality, mode of analysis, case origin, and cutoff value classification.
Collapse
Affiliation(s)
- Chenzhao Yuan
- Faculty of Nursing, Chiang Mai University, Chiang Mai, Thailand
| | - Lumin Zeng
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Hongxiang Duan
- Faculty of Nursing, Chiang Mai University, Chiang Mai, Thailand
- School of Nursing, Chengdu University, Chengdu, China
| | | |
Collapse
|
47
|
Arayici ME, İnal A, Basbinar Y, Olgun N. Evaluation of the diagnostic and prognostic clinical values of circulating tumor DNA and cell-free DNA in pancreatic malignancies: a comprehensive meta-analysis. Front Oncol 2024; 14:1382369. [PMID: 38983931 PMCID: PMC11231086 DOI: 10.3389/fonc.2024.1382369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 06/10/2024] [Indexed: 07/11/2024] Open
Abstract
BACKGROUND The diagnostic and prognostic clinical value of circulating tumor DNA (ctDNA) and cell-free DNA (cfDNA) in pancreatic malignancies are unclear. Herein, we aimed to perform a meta-analysis to evaluate ctDNA and cfDNA as potential diagnostic and prognostic biomarkers. METHODS PRISMA reporting guidelines were followed closely for conducting the current meta-analysis. The PubMed/Medline, Scopus, and Web of Science (WoS) databases were scanned in detail to identify eligible papers for the study. A quality assessment was performed in accordance with the REMARK criteria. The risk ratios (RRs) of the diagnostic accuracy of ctDNA compared to that of carbohydrate antigen 19.9 (CA 19.9) in all disease stages and the hazard ratios (HRs) of the prognostic role of ctDNA in overall survival (OS) were calculated with 95% confidence intervals (CIs). RESULTS A total of 18 papers were evaluated to assess the diagnostic accuracy and prognostic value of biomarkers related to pancreatic malignancies. The pooled analysis indicated that CA19.9 provides greater diagnostic accuracy across all disease stages than ctDNA or cfDNA (RR = 0.64, 95% CI: 0.50-0.82, p < 0.001). Additionally, in a secondary analysis focusing on prognosis, patients who were ctDNA-positive were found to have significantly worse OS (HR = 2.00, 95% CI: 1.51-2.66, p < 0.001). CONCLUSION The findings of this meta-analysis demonstrated that CA19-9 still has greater diagnostic accuracy across all disease stages than KRAS mutations in ctDNA or cfDNA. Nonetheless, the presence of detectable levels of ctDNA was associated with worse patient outcomes regarding OS. There is a growing need for further research on this topic. SYSTEMATIC REVIEW REGISTRATION https://doi.org/10.37766/inplasy2023.12.0092, identifier INPLASY2023120092.
Collapse
Affiliation(s)
- Mehmet Emin Arayici
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Dokuz Eylül University, İzmir, Türkiye
- Department of Public Health, Faculty of Medicine, Dokuz Eylül University, İzmir, Türkiye
| | - Abdullah İnal
- Department of General Surgery, Faculty of Medicine, İzmir Democracy University, İzmir, Türkiye
| | - Yasemin Basbinar
- Department of Translational Oncology, Institute of Oncology, Dokuz Eylül University, İzmir, Türkiye
| | - Nur Olgun
- Department of Clinical Oncology, Institute of Oncology, Dokuz Eylül University, İzmir, Türkiye
| |
Collapse
|
48
|
Mihailović J, Roganović J, Starčević I, Nikolić I, Prvulović Bunović N, Nikin Z. Diagnostic Performance of F-18 FDG PET/CT in the Detection of Recurrent Colorectal Cancer: Correlation with Biochemical Markers and Conventional Imaging Modalities. J Clin Med 2024; 13:3602. [PMID: 38930131 PMCID: PMC11204678 DOI: 10.3390/jcm13123602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/05/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Background/Objectives: Although the role of PET/CT imaging is well established in oncology, its diagnostic value in routine monitoring for recurrent colorectal cancer (CRC) is still controversial. The aim was to evaluate the diagnostic value of F-18 FDG PET/CT in detecting recurrent CRC in correlation with CEA, CA 19-9 levels, and conventional imaging modalities (CIM). Methods: Between 2009 and 2023, a retrospective study was performed including 134 CRC patients referred for PET/CT imaging on the suspicion of recurrence, based on elevated CEA and/or CA 19-9 and/or equivocal CIM findings. According to our institution's Tumor Board CRC protocol, after the initial treatment, which was dependent on the TNM stage (neoadjuvant therapy, primary resection, or adjuvant treatment), patients underwent a standard 5-year surveillance including CEA and CA 19-9 measurements, CIM, and colonoscopy, every six months. The statistics, including univariate and multivariate analyses were conducted using the IBM SPSS 20.0 statistical software. p-values < 0.05 were considered statistically significant. Results: Recurrent CRC was confirmed in 54/134 (40.3%) patients with elevated tumor markers. PET/CT showed high diagnostic performance in detecting recurrent CRC with sensitivity, specificity, PPV, NPV, and accuracy of 94.4%, 82.5%, 78.5%, 95.7%, and 87.3%, respectively. The CEA showed a high sensitivity of 98.1% but both low specificity and accuracy of 15% and 48.5%, respectively. The sensitivity, specificity, and accuracy for CA 19-9 and CIM for diagnosis of CRC recurrence were 44.4%, 67.5%, 58.2%, and 51.9%, 98.8%, 79.9%, respectively. The AUC for PET/CT, elevated CEA levels, CIM, and elevated CA 19-9 levels was 0.885 (95% CI: 0.824-0.946; p < 0.001), 0.844 (95% CI: 0.772-0.916; p < 0.001), 0.753 (95% CI: 0.612-0.844; p < 0.001), and 0.547 (95% CI: 0.442-0.652; p = 0.358), respectively. Univariate analysis showed that both PET/CT and CIM positive results were highly associated with CRC recurrence (p < 0.001 and p < 0.001, respectively). At the same time, gender, mucinous tumor type, presence of initial lymph node metastasis (N+), and presence of initial distant metastasis (M+) had no significance (p = 0.211, p = 0.158, p = 0.583, and p = 0.201, respectively). Our multivariate analysis showed that independent predictors for CRC recurrence are positive PET/CT scans (p < 0.001), positive CIM results (p = 0.001), and elevated CA 19-9 levels (p = 0.023). Although CA 19-9 was not detected as a statistically significant predictor in the univariate analysis (p = 0.358), in a multivariate analysis it was recognized as a significant predicting factor in detecting the CRC recurrence (p = 0.023). Conclusions: F-18 FDG PET/CT showed high diagnostic efficacy in CRC recurrence detection, in correlation with CEA levels, CA 19-9 levels, and CIM. This imaging modality should be routinely integrated into the post-operative follow-op in patients with elevated tumor markers.
Collapse
Affiliation(s)
- Jasna Mihailović
- Department of Nuclear Medicine, Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000 Novi Sad, Serbia;
- Division of Nuclear Medicine, Oncology Institute of Vojvodina, Put dr Goldmana 4, 21204 Sremska Kamenica, Serbia; (J.R.); (I.S.)
| | - Jelena Roganović
- Division of Nuclear Medicine, Oncology Institute of Vojvodina, Put dr Goldmana 4, 21204 Sremska Kamenica, Serbia; (J.R.); (I.S.)
| | - Ivana Starčević
- Division of Nuclear Medicine, Oncology Institute of Vojvodina, Put dr Goldmana 4, 21204 Sremska Kamenica, Serbia; (J.R.); (I.S.)
| | - Ivan Nikolić
- Department of Internal Medicine, Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000 Novi Sad, Serbia;
- Clinic for Medical Oncology, Oncology Institute of Vojvodina, Put dr Goldmana 4, 21204 Sremska Kamenica, Serbia
| | - Nataša Prvulović Bunović
- Department of Nuclear Medicine, Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000 Novi Sad, Serbia;
- Centre for Diagnostic Imaging, Oncology Institute of Vojvodina, Put dr Goldmana 4, 21204 Sremska Kamenica, Serbia
| | - Zoran Nikin
- Department for Pathoanatomical Diagnostics, Oncology Institute of Vojvodina, Put dr Goldmana 4, 21204 Sremska Kamenica, Serbia;
| |
Collapse
|
49
|
Yaghoubi Naei V, Ivanova E, Mullally W, O'Leary CG, Ladwa R, O'Byrne K, Warkiani ME, Kulasinghe A. Characterisation of circulating tumor-associated and immune cells in patients with advanced-stage non-small cell lung cancer. Clin Transl Immunology 2024; 13:e1516. [PMID: 38835954 PMCID: PMC11147668 DOI: 10.1002/cti2.1516] [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: 02/29/2024] [Revised: 04/26/2024] [Accepted: 05/16/2024] [Indexed: 06/06/2024] Open
Abstract
Objectives Globally, non-small cell lung cancer (NSCLC) is the most prevalent form of lung cancer and the leading cause of cancer-related deaths. Tumor-associated circulating cells in NSCLC can have a wide variety of morphological and phenotypic characteristics, including epithelial, immunological or hybrid subtypes. The distinctive characteristics and potential clinical significance of these cells in patients with NSCLC are explored in this study. Methods We utilised a spiral microfluidic device to enrich large cells and cell aggregates from the peripheral blood samples of NSCLC patients. These cells were characterised through high-resolution immunofluorescent imaging and statistical analysis, correlating findings with clinical information from our patient cohort. Results We have identified varied populations of heterotypic circulating tumor cell clusters with differing immune cell composition that included a distinct class of atypical tumor-associated macrophages that exhibits unique morphology and cell size. This subtype's prevalence is positively correlated with the tumor stage, progression and metastasis. Conclusions Our study reveals a heterogeneous landscape of circulating tumor cells and their clusters, underscoring the complexity of NSCLC pathobiology. The identification of a unique subtype of atypical tumor-associatedmacrophages that simultaneously express both tumor and immune markers and whose presence correlates with late disease stages, poor clinical outcomes and metastatic risk infers the potential of these cells as biomarkers for NSCLC staging and prognosis. Future studies should focus on the role of these cells in the tumor microenvironment and their potential as therapeutic targets. Additionally, longitudinal studies tracking these cell types through disease progression could provide further insights into their roles in NSCLC evolution and response to treatment.
Collapse
Affiliation(s)
- Vahid Yaghoubi Naei
- School of Biomedical EngineeringUniversity of Technology SydneySydneyNSWAustralia
- Frazer Institute, Faculty of MedicineThe University of QueenslandBrisbaneQLDAustralia
| | - Ekaterina Ivanova
- Cancer and Ageing Research Program, Centre for Genomics and Personalised HealthQueensland University of TechnologyWoolloongabbaQLDAustralia
| | | | | | - Rahul Ladwa
- Frazer Institute, Faculty of MedicineThe University of QueenslandBrisbaneQLDAustralia
- The Princess Alexandra HospitalBrisbaneQLDAustralia
| | - Ken O'Byrne
- The Princess Alexandra HospitalBrisbaneQLDAustralia
| | - Majid E Warkiani
- School of Biomedical EngineeringUniversity of Technology SydneySydneyNSWAustralia
| | - Arutha Kulasinghe
- Frazer Institute, Faculty of MedicineThe University of QueenslandBrisbaneQLDAustralia
| |
Collapse
|
50
|
Mohammadian Rad N, Sosef O, Seegers J, Koolen LJER, Hoofwijk JJWA, Woodruff HC, Hoofwijk TAGM, Sosef M, Lambin P. Prognostic models for colorectal cancer recurrence using carcinoembryonic antigen measurements. Front Oncol 2024; 14:1368120. [PMID: 38873251 PMCID: PMC11169634 DOI: 10.3389/fonc.2024.1368120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 05/06/2024] [Indexed: 06/15/2024] Open
Abstract
Objective Colorectal cancer (CRC) is one of the most prevalent cancers worldwide. A considerable percentage of patients who undergo surgery with curative intent will experience cancer recurrence. Early identification of individuals with a higher risk of recurrence is crucial for healthcare professionals to intervene promptly and devise appropriate treatment strategies. In this study, we developed prognostic models for CRC recurrence using machine learning models on a limited number of CEA measurements. Method A dataset of 1927 patients diagnosed with Stage I-III CRC and referred to Zuyderland Hospital for surgery between 2008 and 2016 was utilized. Machine learning models were trained using this comprehensive dataset, which included demographic details, clinicopathological factors, and serial measurements of Carcinoembryonic Antigen (CEA). In this study, the predictive performance of these models was assessed, and the key prognostic factors influencing colorectal cancer (CRC) recurrence were pinpointed. Result Among the evaluated models, the gradient boosting classifier demonstrated superior performance, achieving an Area Under the Curve (AUC) score of 0.81 and a balanced accuracy rate of 0.73. Recurrence prediction was shown to be feasible with an AUC of 0.71 when using only five post-operative CEA measurements. Furthermore, key factors influencing recurrence were identified and elucidated. Conclusion This study shows the transformative role of machine learning in recurrence prediction for CRC, particularly by investigating the minimum number of CEA measurements required for effective recurrence prediction. This approach not only contributes to the optimization of clinical workflows but also facilitates the development of more effective, individualized treatment plans, thereby laying the groundwork for future advancements in this area. Future directions involve validating these models in larger and more diverse cohorts. Building on these efforts, our ultimate goal is to develop a risk-based follow-up strategy that can improve patient outcomes and enhance healthcare efficiency.
Collapse
Affiliation(s)
- Nastaran Mohammadian Rad
- The D-Lab, Department of Precision Medicine, GROW – Research Institute for Oncology & Reproduction, Maastricht University, Maastricht, Netherlands
| | - Odin Sosef
- Department of Surgery, Zuyderland Medisch Centrum, Sittard-Geleen, Netherlands
| | - Jord Seegers
- Department of Surgery, Zuyderland Medisch Centrum, Sittard-Geleen, Netherlands
| | | | | | - Henry C. Woodruff
- The D-Lab, Department of Precision Medicine, GROW – Research Institute for Oncology & Reproduction, Maastricht University, Maastricht, Netherlands
- Department of Radiology and Nuclear Medicine, GROW – Research Institute for Oncology & Reproduction, Maastricht University Medical Center, Maastricht, Netherlands
| | | | - Meindert Sosef
- Department of Surgery, Zuyderland Medisch Centrum, Sittard-Geleen, Netherlands
| | - Philippe Lambin
- The D-Lab, Department of Precision Medicine, GROW – Research Institute for Oncology & Reproduction, Maastricht University, Maastricht, Netherlands
| |
Collapse
|