1
|
Li Y, Wang X, Wang X, Qin Z, Li C, Yang J, Cao M. Electrochemical biosensor based on composite of gold nanoparticle/reduced-graphene oxide/graphitic carbon nitride and a caprolactone polymer for highly sensitive detection of CEA. Bioelectrochemistry 2025; 163:108897. [PMID: 39764934 DOI: 10.1016/j.bioelechem.2024.108897] [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: 11/04/2024] [Revised: 12/27/2024] [Accepted: 12/30/2024] [Indexed: 02/12/2025]
Abstract
Carcinoembryonic antigen (CEA) is a broad-spectrum biomarker, and its accurate detection and analysis is important for early clinical diagnosis and treatment. This study aimed to develop a highly sensitive and selective sandwich-type immunosensor based on electrochemical impedance spectroscopy (EIS) for the accurate detection of CEA. A novel composite material, gold nanoparticle/reduced-graphene oxide/graphitic carbon nitride (AuNPs/rGO/g-C3N4), was synthesized with excellent electrical conductivity and a large specific surface area to immobilize biological probes. And ab1-CEA-ab2 formed a sandwich structure of 'antibody-antigen-antibody', which ensured the high selectivity of the biosensor. Furthermore, the introduction of caprolactone polymer (DMPA-PCL) significantly amplifies the impedance signal and improves the sensitivity of the analytical method. Scanning electron microscopy, x-ray diffraction, transmission electron microscopy Fourier transform infrared spectroscopy, and ultraviolet-visible spectrophotometry were used to characterise the prepared AuNPs/rGO/g-C3N4 and DMPA-PCL. Under the optimal conditions, the sensor showed good analytical performance for the detection of CEA with a linear range of 100 fg mL-1-100 ng mL-1 and a detection limit of 83.2 fg mL-1. And the sandwich-type immunosensor showed good selectivity and stability for the recognition of CEA in real samples.
Collapse
Affiliation(s)
- Yunpeng Li
- Henan University of Chinese Medicine, Zhengzhou 450046, People's Republic of China
| | - Xia Wang
- Henan University of Chinese Medicine, Zhengzhou 450046, People's Republic of China.
| | - Xinling Wang
- Henan University of Chinese Medicine, Zhengzhou 450046, People's Republic of China.
| | - Zhe Qin
- Henan University of Chinese Medicine, Zhengzhou 450046, People's Republic of China
| | - Chong Li
- Henan University of Chinese Medicine, Zhengzhou 450046, People's Republic of China
| | - Jing Yang
- Henan University of Chinese Medicine, Zhengzhou 450046, People's Republic of China
| | - Mengmeng Cao
- Henan University of Chinese Medicine, Zhengzhou 450046, People's Republic of China
| |
Collapse
|
2
|
Chandio I, Rahujo S, Chandio ZA, Rashid MA, Rehman MU, Shar ZH, Thebo KH. Recent development in metal-organic frameworks-based electrochemical aptasensors for detection of cancer biomarkers. Bioelectrochemistry 2025; 165:109006. [PMID: 40393088 DOI: 10.1016/j.bioelechem.2025.109006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2025] [Revised: 05/09/2025] [Accepted: 05/13/2025] [Indexed: 05/22/2025]
Abstract
Aptasensors utilize aptamers as recognition elements, which offer high sensitivity, selectivity, ease of modification, outstanding stability, real-time detection, biocompatibility, cost-effectiveness, and good integration. Utilizing metal-organic framework (MOF)-based electrochemical aptasensors is a promising approach that is drawing significant attention. MOF-based aptasensors for cancer detection present a promising avenue, yet they face several challenges. The optimization of sensitivity and selectivity in the MOF-aptamer system is complex, requiring a delicate balance to distinguish the target analyte from interfering substances in physiological environments. Efficient immobilization of aptamers on MOF surfaces while maintaining their conformation and stability under physiological conditions poses a crucial challenge for robust sensing. Integrating MOFs and aptamers with transduction systems, such as electrodes in electrochemical sensors, is critical for achieving efficient signal transduction and maintaining sensor stability. Herein, the latest developments in MOF-based electrochemical aptasensors for detecting tumor markers will be discussed. Focus will be given on single metallic, bimetallic, and calcinated-based MOFs and their strengths and weaknesses will be summarized. Further, the synthesis strategy of electrochemical sensors is analyzed to meet the requirements of selectivity and sensitivity. Finally, the future perspectives for developing and applying electrochemical aptasensors are also discussed.
Collapse
Affiliation(s)
- Imamdin Chandio
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University, Beijing 100084, China; National Centre of Excellence inraphene Analytical Chemistry, University of Sindh, Jamshoro, Pakistan.
| | - Sanam Rahujo
- National Centre of Excellence inraphene Analytical Chemistry, University of Sindh, Jamshoro, Pakistan
| | - Zaheer Ahmed Chandio
- Department of Chemistry, Shaheed Benazir Bhutto University, Shaheed Benazirabad, Pakistan
| | - Md Abdur Rashid
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Al Faraa, Abha 62223, Saudi Arabi
| | - Misbah Ur Rehman
- Department of Chemistry, University of Mianwali, Mianwali 42200, Pakistan
| | - Zahid H Shar
- Dr. M.A. Kazi Institute of Chemistry, University of Sindh, Jamshoro, Pakistan
| | | |
Collapse
|
3
|
Zheng J, Tu Y, Jin H, Sun H, Shen G, Tong H. Changes of peripheral blood lymphocytes, neutrophils, CEA, TAP and ferritin in colorectal adenoma and colorectal cancer and the diagnostic performance of these makers in evaluating colorectal cancer. Arab J Gastroenterol 2025:S1687-1979(25)00056-5. [PMID: 40345957 DOI: 10.1016/j.ajg.2025.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 12/24/2024] [Accepted: 03/09/2025] [Indexed: 05/11/2025]
Abstract
BACKGROUND AND STUDY AIMS Colorectal cancer (CRC) has been recognized as a multifactorial disease arising from precursor lesions and characterized by chronic inflammation. Therefore, the inflammatory response is one of the critical indicators for the diagnosis of CRC. Timely and effective screening is an effective strategy to reduce the incidence and mortality of CRC. The primary aim of this study was to analyze the differences in white blood cell count (WBC) in patients with colorectal adenoma (CRA) and CRC. The second aim was to estimate the diagnostic performance of using a panel of serum tumor markers and WBC for CRC screen. PATIENTS AND METHODS We retrospectively reviewed a database of patients who were diagnosed with CRA and CRC. Serum tumor markers and blood routine examination data were completed before receiving any anticancer therapy. RESULTS A total of 538 participants were enrolled, including 169 health participants, 195 patients with CRA and 174 patients with CRC. Lymphocyte counts were lower in CRC than CRA and healthy participants. Neutrophil counts were higher in CRC and CRA than healthy participants. The CEA levels were higher in CRA and CRC than healthy participants, and higher in CRC than CRA. The areas of tumor TAP were larger in CRC than CRA and healthy participants. The ferritin levels were lower in CRC than CRA and healthy participants. The 8-marker panel yielded an AUC of 0.854 higher than single marker. There is no difference in the diagnostic performance of TAP, CEA, ferritin and NLR. CONCLUSIONS There are a lot of high sensitivity and specificity methods for CRC screening. However, most screening programs suffer from poor participation rates. Herein, our 8-marker panel is cost-effective and high-performance screen system for the detection of CRC and is crucial for enhancing the participation rates in current screening programs.
Collapse
Affiliation(s)
- Jialai Zheng
- Molecular Medicine Center, Shaoxing Second Hospital, Shaoxing, Zhejiang, China.
| | - Yongtao Tu
- Molecular Medicine Center, Shaoxing Second Hospital, Shaoxing, Zhejiang, China
| | - Haiyong Jin
- Departments of Laboratory Medicine, Shaoxing Second Hospital, Shaoxing, Zhejiang, China
| | - Haiyan Sun
- Departments of Laboratory Medicine, Shaoxing Second Hospital, Shaoxing, Zhejiang, China
| | - Guanqiao Shen
- Departments of Laboratory Medicine, Shaoxing Second Hospital, Shaoxing, Zhejiang, China
| | - Haijiang Tong
- Departments of Laboratory Medicine, Shaoxing Second Hospital, Shaoxing, Zhejiang, China
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Basu S, Rana N, Morgan D, Sen K. Gold Nanoparticle Incorporated Graphene Oxide as a SERS Platform for Ultratrace Antibody Free Sensing of the Cancer Biomarker CEA. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:7886-7901. [PMID: 40062598 DOI: 10.1021/acs.langmuir.5c00522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
A simple, fast, low-cost, and efficient method is designed for the synthesis of graphene oxide (GO) (20 nm) from graphite using a strong oxidant Ce(IV). GO is further modified with gold nanoparticles (AuNPs) (5-8 nm) to generate a AuGO nanocomposite (25 nm). Raman spectral analyses confirm that the synthesized AuGO has a potential selective sensing ability for the cancer biomarker carcinoembryonic antigen (CEA) in serum. Sensing assays are also carried out in the presence of high concentrations of glucose, cholesterol, and insulin using this method, which become significantly elevated in conditions of different pathophysiological disorders. Ultratrace antibody free sensing of CEA in serum is achieved using surface-enhanced Raman spectroscopy with an amazing LOD of 12.5 fg/mL. The interaction between CEA and AuGO is further established using Raman, fluorescence, circular dichroism spectroscopy, and theoretical studies. The specificity of sensing is tested by checking the response in the presence of other cancer biomarkers, such as CA 19-9, CA 125, and PSA, which do not show any signal enhancement with AuGO in Raman spectroscopy.
Collapse
Affiliation(s)
- Shalmali Basu
- Department of Chemistry, University of Calcutta, 92, APC Road, Kolkata 700009, India
| | - Nabakumar Rana
- Department of Physics, University of Calcutta, 92, APC Road, Kolkata 700009, India
| | - David Morgan
- Cardiff Catalysis Institute, School of Chemistry, Cardiff University, Park Place, Cardiff CF10 3AT, U.K
| | - Kamalika Sen
- Department of Chemistry, University of Calcutta, 92, APC Road, Kolkata 700009, India
| |
Collapse
|
7
|
Shimura T, Yin C, Ma R, Zhang A, Nagai Y, Shiratori A, Ozaki H, Yamashita S, Higashi K, Sato Y, Imaoka H, Kitajima T, Kawamura M, Koike Y, Okita Y, Yoshiyama S, Ohi M, Hayashi A, Imai H, Zhang X, Okugawa Y, Toiyama Y. The prognostic importance of the negative regulators of ferroptosis, GPX4 and HSPB1, in patients with colorectal cancer. Oncol Lett 2025; 29:144. [PMID: 39850719 PMCID: PMC11755263 DOI: 10.3892/ol.2025.14890] [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: 09/16/2024] [Accepted: 12/16/2024] [Indexed: 01/25/2025] Open
Abstract
The prognostic value of negative regulators of ferroptosis in patients with colorectal cancer (CRC) has not yet been fully elucidated. The present study performed a systematic in silico identification and selection of candidate negative regulators of ferroptosis using The Cancer Genome Atlas data cohort (n=367), followed by clinical validation through immunohistochemistry of samples from patients with CRC (n=166) and further in vitro evaluation. In silico analysis identified specific light-chain subunit of the cystine/glutamate antiporter, AIFM2, NFE2L2, FTH1, GLS2, glutathione peroxidase 4 (GPX4) and heat shock protein β-1 (HSPB1) genes as possible candidates. Furthermore, patients with high expression of GPX4 or HSPB1 exhibited significantly worse overall survival (OS) compared with those with low expression (P<0.01 for both). Immunohistochemical analysis revealed that both OS and recurrence-free survival (RFS) of patients with CRC and high GPX4 or HSPB1 expression were significantly worse compared with in patients with low expression (P<0.01 for all). Furthermore, multivariate analysis showed that high GPX4 and HSPB1 expression were independent risk factors for poor oncological outcome for OS and RFS (GPX4: RFS, P=0.03; HSPB1: OS, P=0.006 and RFS, P<0.0001). Moreover, the effects of GPX4 and HSPB1 small interfering RNAs on two CRC cell lines (DLD-1 and SW480) indicated that GPX4 and HSPB1 may exhibit important roles in attenuating the cytotoxic effect of 5-fluorouracil-based chemotherapy. In conclusion, the current study confirmed that GPX4 and HSPB1 may serve as substantial prognostic- and recurrence-predictive biomarkers in patients with CRC.
Collapse
Affiliation(s)
- Tadanobu Shimura
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Chengzeng Yin
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Ruiya Ma
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
- Department of Surgery, Tangshan Gongren Hospital, Tangshan, Hebei 063007, P.R. China
| | - Aiying Zhang
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Yuka Nagai
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Aoi Shiratori
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Hana Ozaki
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Shinji Yamashita
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Koki Higashi
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Yuki Sato
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Hiroki Imaoka
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Takahito Kitajima
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
- Department of Genomic Medicine, Mie University Hospital, Tsu, Mie 514-8507, Japan
| | - Mikio Kawamura
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Yuhki Koike
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Yoshiki Okita
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Shigeyuki Yoshiyama
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Masaki Ohi
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Akinobu Hayashi
- Department of Oncologic Pathology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Hiroshi Imai
- Department of Oncologic Pathology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Xueming Zhang
- Department of Surgery, Tangshan Gongren Hospital, Tangshan, Hebei 063007, P.R. China
| | - Yoshinaga Okugawa
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
- Department of Genomic Medicine, Mie University Hospital, Tsu, Mie 514-8507, Japan
| | - Yuji Toiyama
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| |
Collapse
|
8
|
Radhakrishnan SK, Nath D, Russ D, Merodio LB, Lad P, Daisi FK, Acharjee A. Machine learning-based identification of proteomic markers in colorectal cancer using UK Biobank data. Front Oncol 2025; 14:1505675. [PMID: 39839775 PMCID: PMC11746037 DOI: 10.3389/fonc.2024.1505675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 12/02/2024] [Indexed: 01/23/2025] Open
Abstract
Colorectal cancer is one of the leading causes of cancer-related mortality in the world. Incidence and mortality are predicted to rise globally during the next several decades. When detected early, colorectal cancer is treatable with surgery and medications. This leads to the requirement for prognostic and diagnostic biomarker development. Our study integrates machine learning models and protein network analysis to identify protein biomarkers for colorectal cancer. Our methodology leverages an extensive collection of proteome profiles from both healthy and colorectal cancer individuals. To identify a potential biomarker with high predictive ability, we used three machine learning models. To enhance the interpretability of our models, we quantify each protein's contribution to the model's predictions using SHapley Additive exPlanations values. Three classifiers-LASSO, XGBoost, and LightGBM were evaluated for predictive performance along with hyperparameter tuning of each model using grid search, with LASSO achieving the highest AUC of 75% in the UK Biobank dataset and the AUCs for LightGBM and XGBoost are 69.61% and 71.42%, respectively. Using SHapley Additive exPlanations values, TFF3, LCN2, and CEACAM5 were found to be key biomarkers associated with cell adhesion and inflammation. Protein quantitative trait loci analyze studies provided further evidence for the involvement of TFF1, CEACAM5, and SELE in colorectal cancer, with possible connections to the PI3K/Akt and MAPK signaling pathways. By offering insights into colorectal cancer diagnostics and targeted therapeutics, our findings set the stage for further biomarker validation.
Collapse
Affiliation(s)
| | - Dipanwita Nath
- College of Medicine and Health, School of Medical Sciences, Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Dominic Russ
- College of Medicine and Health, School of Medical Sciences, Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
- Institute of Translational Medicine, University Hospitals Birmingham National Health Service (NHS) Foundation Trust, Birmingham, United Kingdom
- Centre for Health Data Research, University of Birmingham, Birmingham, United Kingdom
| | - Laura Bravo Merodio
- College of Medicine and Health, School of Medical Sciences, Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
- Institute of Translational Medicine, University Hospitals Birmingham National Health Service (NHS) Foundation Trust, Birmingham, United Kingdom
- Centre for Health Data Research, University of Birmingham, Birmingham, United Kingdom
| | - Priyani Lad
- College of Medicine and Health, School of Medical Sciences, Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Folakemi Kola Daisi
- College of Medicine and Health, School of Medical Sciences, Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Animesh Acharjee
- College of Medicine and Health, School of Medical Sciences, Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
- Institute of Translational Medicine, University Hospitals Birmingham National Health Service (NHS) Foundation Trust, Birmingham, United Kingdom
- Centre for Health Data Research, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
9
|
Tong G, Li H, Shen Y, Tan Z, Qian H. The combined evaluation of preoperative serum CEA and postoperative tissue CEA as a prognostic factor in stages 0-IV colorectal cancer: a retrospective cohort study. Front Med (Lausanne) 2025; 11:1447041. [PMID: 39830382 PMCID: PMC11739305 DOI: 10.3389/fmed.2024.1447041] [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: 06/11/2024] [Accepted: 12/11/2024] [Indexed: 01/22/2025] Open
Abstract
Background The roles of preoperative serum carcinoembryonic antigen (sCEA) and postoperative tissue carcinoembryonic antigen (tCEA) have been extensively studied in isolation in colorectal cancer (CRC). However, the combined role of sCEA and tCEA remains inadequately described. Methods A total of 1,757 retrospective cases of stage 0-IV CRC from January 2006 to January 2016 in our institution were included. Clinicopathological features and follow-up data were collected. Stage 0 was combined with stage I. sCEA levels were classified as normal or high (>10 ng/mL), while tCEA levels were categorised into three grades (+, ++, and +++). This resulted in six combined groups (2 × 3). ANOVA and cross-tabulation were employed to analyse continuous and categorical data, respectively. Univariate and multivariate analyses were conducted using Cox regression. All data were analysed using SPSS 27 and R 4.3.1. Results Some clinicopathologic features differed significantly among the combined CEA test groups (all p < 0.05). The receiver operating characteristic (ROC) curves for sCEA, tCEA, and combined CEA exhibited significant differences in five-year OS with death as the input variable (all p < 0.05). The area under the curve (AUC) for combined CEA was the highest, indicating the value of this study. Cox regression analysis demonstrated that tumour location, T stage, differentiation, chemotherapy, TNM stage, tCEA, and combined CEA were significant in the univariate analysis; however, tCEA was not significant (p = 0.096) in the multivariate analysis among these seven variables. Five-year OS analysis revealed that sCEA, tCEA, and combined CEA were not significant in stages 0 & I-II (all p > 0.05) but were significant in stages III-IV (all p < 0.05), except for tCEA in stage IV (p = 0.24) as per K-M and univariate analysis. No significant difference was observed between sCEA and tCEA (p = 0.55, 0.095), whereas combined CEA demonstrated a significant difference (p < 0.001) in both univariate and multivariate analyses. Conclusion sCEA, tCEA, and combined CEA exhibit prognostic roles in stages III-IV of CRC, with only combined CEA serving as an independent factor in these stages.
Collapse
Affiliation(s)
- Guojun Tong
- Department of Colorectal Surgery, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Zhejiang, China
- Department of Central Laboratory, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Zhejiang, China
| | - Hui Li
- Department of General Surgery, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Zhejiang, China
| | - Yan Shen
- Department of General Surgery, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Zhejiang, China
| | - Zhenhua Tan
- Department of General Surgery, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Zhejiang, China
| | - Hai Qian
- Department of General Surgery, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Zhejiang, China
| |
Collapse
|
10
|
Yu L, Wang H, Wang F, Guo J, Xiao B, Hou Z, Lu Z, Pan Z, Zhou Y, Ye S, Wan D, Lin B, Ou Q, Fang Y. Serum biomarkers REG1A and REG3A combined with the traditional CEA represent a novel nomogram for the screening and risk stratification of colorectal cancer. Clin Transl Oncol 2025; 27:277-290. [PMID: 38965192 DOI: 10.1007/s12094-024-03566-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 06/09/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND To develop and validate a serum protein nomogram for colorectal cancer (CRC) screening. METHODS The serum protein characteristics were extracted from an independent sample containing 30 colorectal cancer and 12 polyp tissues along with their paired samples, and different serum protein expression profiles were validated using RNA microarrays. The prediction model was developed in a training cohort that included 1345 patients clinicopathologically confirmed CRC and 518 normal participants, and data were gathered from November 2011 to January 2017. The lasso logistic regression model was employed for features selection and serum nomogram building. An internal validation cohort containing 576 CRC patients and 222 normal participants was assessed. RESULTS Serum signatures containing 27 secreted proteins were significantly differentially expressed in polyps and CRC compared to paired normal tissue, and REG family proteins were selected as potential predictors. The C-index of the nomogram1 (based on Lasso logistic regression model) which contains REG1A, REG3A, CEA and age was 0.913 (95% CI, 0.899 to 0.928) and was well calibrated. Addition of CA199 to the nomogram failed to show incremental prognostic value, as shown in nomogram2 (based on logistic regression model). Application of the nomogram1 in the independent validation cohort had similar discrimination (C-index, 0.912 [95% CI, 0.890 to 0.934]) and good calibration. The decision curve (DCA) and clinical impact curve (ICI) analysis demonstrated that nomogram1 was clinically useful. CONCLUSIONS This study presents a serum nomogram that included REG1A, REG3A, CEA and age, which can be convenient for screening of colorectal cancer.
Collapse
Affiliation(s)
- Long Yu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Hao Wang
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Fulong Wang
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Jian Guo
- Senboll Biotechnology Co., Ltd., Pingshan Bio-Pharmacy Business Accelerator, Pingshan District, Shenzhen, 518000, Guangdong, China
| | - Binyi Xiao
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Zhenlin Hou
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Zhenhai Lu
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Zhizhong Pan
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Yaxian Zhou
- Senboll Biotechnology Co., Ltd., Pingshan Bio-Pharmacy Business Accelerator, Pingshan District, Shenzhen, 518000, Guangdong, China
| | - Sibin Ye
- Senboll Biotechnology Co., Ltd., Pingshan Bio-Pharmacy Business Accelerator, Pingshan District, Shenzhen, 518000, Guangdong, China
| | - Desen Wan
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Bo Lin
- Department of Thyroid Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510060, China.
| | - Qingjian Ou
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China.
| | - Yujing Fang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China.
| |
Collapse
|
11
|
Feng X, Chen J, Lian J, Dong T, Gao Y, Zhang X, Zhai Y, Zou B, Guo Y, Xu E, Cui Y, Zhang L. The glycogene alterations and potential effects in esophageal squamous cell carcinoma. Cell Mol Life Sci 2024; 81:481. [PMID: 39636330 PMCID: PMC11621258 DOI: 10.1007/s00018-024-05534-3] [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: 06/12/2024] [Revised: 11/13/2024] [Accepted: 11/23/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Aberrant glycosylation is one of the hallmarks of cancer. The profile of glycoprotein expression caused by abnormal glycosylation has been revealed, while abnormal glycogenes that may disturb the structure of glycans have not yet been identified in esophageal squamous cell carcinoma (ESCC). METHODS Genomic alterations driven by differentially expressed glycogenes in ESCC were compared with matched normal tissues by multi-omics analysis. Immunohistochemistry, MTT, colony formation, transwell assays, subcutaneous tumor formation experiments and tail vein injection were used to study the expression and the effect on the proliferation and metastasis of the differentially expressed glycogenes POFUT1 and RPN1 in ESCC. In the alkyne fucose labeling experiment, AAL lectin affinity chromatography and immunoprecipitation were used to explore the mechanism of POFUT1 in ESCC. RESULTS The expression of the POFUT1 and RPN1 glycogenes were upregulated, as determined by genomic copy number gain and proteomics analysis. The overexpression of POFUT1 or RPN1 was associated with poor prognosis in ESCC patients and affected the proliferation and metastasis of ESCC in vivo and in vitro. The overexpression of POFUT1 increased the overall fucosylation level and activated the Notch signaling pathway, which partially mediated POFUT1 induced pro-migration in ESCC. The regulation of malignant progression of ESCC by RPN1 may be related to the TNF signaling pathway, p53 signaling pathway, etc. CONCLUSIONS: Our study fills a gap in the study of abnormal glycogenes and highlights the potential role of the POFUT1/Notch axis in ESCC. Moreover, our study identifies POFUT1 and RPN1 as promising anticancer targets in ESCC.
Collapse
Affiliation(s)
- Xuefei Feng
- Department of Pathology, Basic Medical Sciences Center, Key Laboratory of Cellular Physiology of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Jinyan Chen
- Department of Pathology, Basic Medical Sciences Center, Key Laboratory of Cellular Physiology of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Jianhong Lian
- Department of Thoracic Surgery, Shanxi Cancer Hospital, Taiyuan, 030001, Shanxi, China
| | - Tianyue Dong
- Department of Pathology, Basic Medical Sciences Center, Key Laboratory of Cellular Physiology of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Yingzhen Gao
- Department of Pathology, Basic Medical Sciences Center, Key Laboratory of Cellular Physiology of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Xiaojuan Zhang
- Department of Pathology, People's Hospital of Puyang, Henan, 457005, China
| | - Yuanfang Zhai
- Department of Pathology, Basic Medical Sciences Center, Key Laboratory of Cellular Physiology of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Binbin Zou
- Department of Pathology, Basic Medical Sciences Center, Key Laboratory of Cellular Physiology of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Yanlin Guo
- Department of Pathology, Basic Medical Sciences Center, Key Laboratory of Cellular Physiology of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Enwei Xu
- Department of Pathology, Shanxi Cancer Hospital, Taiyuan, 030001, Shanxi, China
| | - Yongping Cui
- Department of Pathology, Basic Medical Sciences Center, Key Laboratory of Cellular Physiology of Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| | - Ling Zhang
- Department of Pathology, Basic Medical Sciences Center, Key Laboratory of Cellular Physiology of Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
12
|
Yoon ML, Chun H, Lee H, Seo W, Lee JY, Yoon JH. Identification and Validation of Serum Biomarkers to Improve Colorectal Cancer Diagnosis. Cancer Med 2024; 13:e70460. [PMID: 39628390 PMCID: PMC11615507 DOI: 10.1002/cam4.70460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 08/16/2024] [Accepted: 11/16/2024] [Indexed: 12/08/2024] Open
Abstract
BACKGROUND The pressing need for reliable biomarkers in colorectal cancer (CRC) diagnosis and prognosis is a major global health concern. Current diagnostic methods rely heavily on invasive procedures like colonoscopy, and existing biomarkers such as Carbohydrate Antigen 19-9 (CA19-9) and Carcinoembryonic Antigen (CEA) exhibit limitations in accuracy and specificity. AIMS This study aims to identify and validate novel biomarkers that can enhance the early detection and diagnostic precision of CRC while overcoming the shortcomings of conventional biomarkers. MATERIALS AND METHODS Leveraging advancements in genomic and proteomic technologies, gene expression datasets were obtained from the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA). We identified differentially expressed genes (DEGs) and conducted further analyses, including Gene Ontology (GO) enrichment and Protein-Protein Interaction (PPI) network construction. Five promising biomarkers-INHBA, MMP7, PSAT1, SLC7A5, and TGFBI-were selected based on their robust performance in Receiver Operating Characteristic (ROC) curve analysis. Serum concentrations of these biomarkers were measured in 200 CRC patients and 100 healthy controls. RESULTS The study revealed significantly elevated expression levels of the selected biomarkers in CRC tissues compared to normal tissues. Additionally, serum concentrations of INHBA, MMP7, PSAT1, SLC7A5, and TGFBI were notably higher in CRC patients than in healthy individuals, with Area Under the Curve (AUC) values ranging from 0.8361 to 0.9869 indicating high diagnostic accuracy. Optimal cutoff values for diagnosis ranged from 38.9 pg/mL to 280.7 pg/mL, yielding sensitivity and specificity values between 74.5% and 92.9%. DISCUSSION The findings underscore the potential of INHBA, MMP7, PSAT1, SLC7A5, and TGFBI as effective non-invasive biomarkers for CRC detection. Their elevated serum concentrations and robust discriminatory abilities highlight their promise in improving diagnostic accuracy and patient outcomes compared to traditional biomarkers. CONCLUSION The identification and validation of these novel biomarkers represent a significant advancement in CRC diagnosis and management. Further studies are required to validate their clinical applicability in larger cohorts and to elucidate their functional roles in CRC pathogenesis, ultimately enhancing diagnostic strategies and personalized treatment approaches.
Collapse
Affiliation(s)
- Minha Lea Yoon
- Clinical Trial CenterGangnam St. Peter's HospitalSeoulRepublic of Korea
| | - Hyelim Chun
- Clinical Trial CenterGangnam St. Peter's HospitalSeoulRepublic of Korea
| | - HyunJu Lee
- Clinical Trial CenterGangnam St. Peter's HospitalSeoulRepublic of Korea
| | - WooJeong Seo
- Clinical Trial CenterGangnam St. Peter's HospitalSeoulRepublic of Korea
| | - Jung Young Lee
- Clinical Trial CenterGangnam St. Peter's HospitalSeoulRepublic of Korea
| | - Jung Hwan Yoon
- Department of PathologyCollege of Medicine, The Catholic University of KoreaSeoulRepublic of Korea
| |
Collapse
|
13
|
Liao MY, Hao YJ, Luo CS, Chen CM, Feng PH, Yang HY, Yao DJ, Lee KY, Tseng FG. Development and validation of a novel combinational index of liquid biopsy biomarker for longitudinal lung cancer patient management. THE JOURNAL OF LIQUID BIOPSY 2024; 6:100167. [PMID: 40027304 PMCID: PMC11863939 DOI: 10.1016/j.jlb.2024.100167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 03/05/2025]
Abstract
Objectives Many cancer biomarkers such as the circulating tumor cells/microemboli (CTCs/CTM) have been reported significant associations with clinical outcomes. However, different biomarkers have different sensitivities and specificities for cancer types and cohort patients, and synergistic effects between certain biomarkers have also been observed, leading to the inaccurate, fluctuating, and even controversial results when multiple biomarkers are analyzed together. In this paper, a novel combinational index, P-score, was developed for monitoring and predicting the disease condition of lung cancer patients during follow-up visits. Materials and methods There were totally 13 return patients with 54 blood samples involved in this study to examine the number of CTC and CTM. Information from one group of 7 patients including 27 blood samples with published clinical data was employed to develop while those from another group of 4 patients containing 14 blood samples with unpublished clinical data were used to validate the P score in prediction. Enumerations were based on immunofluorescent staining images. Distributions of CTC/CTM and their frequencies in stratified patients were carefully examined and analyzed the ROC curve and AUC value to develop the P score and P score-based prediction model. Results and conclusion We found that the predictive power of P-score was not only comparable to the traditional cancer marker, in comparison with individual CTC/CTM, more false positives could be corrected by using P-score, thereby to improve the accuracy of analysis. From our preliminary validation tests, the prognosis and disease progression monitored longitudinally by P-score were further confirmed by clinical outcome data from physicians and its sensitivity was even better than those from individual biomarkers. We believe that this novel combinational indicator could be a promising tool to interpret clinical outcomes more accurately from multiple factors, particularly useful for the early prognosis and longitudinal monitoring in cancer patient management.
Collapse
Affiliation(s)
- Min-Yi Liao
- Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Yun-Jie Hao
- Department of Engineering and System Science, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Ching-Shan Luo
- International Ph.D. Program in Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan
- TMU Research Center of Thoracic Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Ching-Mei Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan
| | - Po-Hao Feng
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan
- TMU Research Center of Thoracic Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Hsin-Yu Yang
- Department of Engineering and System Science, National Tsing Hua University, Hsinchu, 30013, Taiwan
- Nano Science and Technology Program, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Da-Jeng Yao
- Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
- Department of Engineering and System Science, National Tsing Hua University, Hsinchu, 30013, Taiwan
- Institute of Nano Engineering and Micro Systems (NEMS), National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Kang-Yun Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan
- TMU Research Center of Thoracic Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Fan-Gang Tseng
- Department of Engineering and System Science, National Tsing Hua University, Hsinchu, 30013, Taiwan
- Institute of Nano Engineering and Micro Systems (NEMS), National Tsing Hua University, Hsinchu, 30013, Taiwan
- Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, 30013, Taiwan
| |
Collapse
|
14
|
Bates M, Mohamed BM, Lewis F, O'Toole S, O'Leary JJ. Biomarkers in high grade serous ovarian cancer. Biochim Biophys Acta Rev Cancer 2024; 1879:189224. [PMID: 39581234 DOI: 10.1016/j.bbcan.2024.189224] [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: 01/28/2024] [Revised: 11/15/2024] [Accepted: 11/15/2024] [Indexed: 11/26/2024]
Abstract
High-grade serous ovarian cancer (HGSC) is the most common subtype of ovarian cancer. HGSC patients typically present with advanced disease, which is often resistant to chemotherapy and recurs despite initial responses to therapy, resulting in the poor prognosis associated with this disease. There is a need to utilise biomarkers to manage the various aspects of HGSC patient care. In this review we discuss the current state of biomarkers in HGSC, focusing on the various available immunohistochemical (IHC) and blood-based biomarkers, which have been examined for their diagnostic, prognostic and theranostic potential in HGSC. These include various routine clinical IHC biomarkers such as p53, WT1, keratins, PAX8, Ki67 and p16 and clinical blood-borne markers and algorithms such as CA125, HE4, ROMA, RMI, ROCA, and others. We also discuss various components of the liquid biopsy as well as a number of novel IHC biomarkers and non-routine blood-borne biomarkers, which have been examined in various ovarian cancer studies. We also discuss the future of ovarian cancer biomarker research and highlight some of the challenges currently facing the field.
Collapse
Affiliation(s)
- Mark Bates
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland.
| | - Bashir M Mohamed
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland
| | - Faye Lewis
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland
| | - Sharon O'Toole
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland; Department of Obstetrics and Gynaecology, Trinity College Dublin, Dublin, Ireland
| | - John J O'Leary
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland; Department of Pathology, Coombe Women & Infants University Hospital, Dublin, Ireland
| |
Collapse
|
15
|
Xie H, Wei L, Wang Q, Tang S, Gan J. Grading carcinoembryonic antigen levels can enhance the effectiveness of prognostic stratification in patients with colorectal cancer: a single-centre retrospective study. BMJ Open 2024; 14:e084219. [PMID: 39477273 PMCID: PMC11529588 DOI: 10.1136/bmjopen-2024-084219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 10/02/2024] [Indexed: 11/03/2024] Open
Abstract
OBJECTIVES This study developed a refined carcinoembryonic antigen (CEA) grading system using CEA cut-off points of 5, 20 and 50 ng/mL and to explore the prognostic value of CEA grading in predicting the progression-free survival (PFS) and overall survival (OS) of colorectal cancer (CRC) patients. DESIGN A retrospective cohort study. SETTING First Affiliated Hospital of Guangxi Medical University. PARTICIPANTS 1107 CRC patients who received surgical treatment. MATERIALS AND METHODS Survival analysis was conducted using the Kaplan-Meier method and compared using the log-rank test. A Cox regression model with a 95% CI was used to evaluate the independent prognostic risk factors for CRC. Prognostic nomograms were constructed to predict the 1-5-year PFS/OS. RESULTS Elevated serum CEA levels are often indicative of recurrence and death in CRC patients. Higher CEA levels were significantly associated with more aggressive tumour phenotypes. The CEA grading system was an independent predictor of prognosis in CRC patients and effectively stratified PFS (62.0% vs 51.2% vs 33.7% vs 20.2%, p<0.001) and OS (64.7% vs 54.4% vs 36.6% vs 22.5%, p<0.001) in CRC patients. As the CEA grade increased, the risk of poor prognosis gradually increased in a gradient manner, with an approximately 10% difference in risk grade between each CEA grade. The internal validation cohort further confirmed that CEA grade remains an effective prognostic factor for the survival of CRC patients. Prognostic nomograms, which integrate individual characteristics, tumour features and CEA grading, provide a more comprehensive prognostic evaluation for CRC patients. CONCLUSIONS The CEA grading system is an independent predictor of prognosis for CRC patients and can effectively stratify PFS and OS.
Collapse
Affiliation(s)
- Hailun Xie
- Department of Gastrointestinal and Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Lishuang Wei
- Geriatric respiratory medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Qiwen Wang
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Shuangyi Tang
- Department of Pharmacy, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jialiang Gan
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
16
|
Lee S, Lee J, Kang SH. Super-resolution Multispectral Imaging Nanoimmunosensor for Simultaneous Detection of Diverse Early Cancer Biomarkers. ACS Sens 2024; 9:3652-3659. [PMID: 38960915 DOI: 10.1021/acssensors.4c00752] [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: 07/05/2024]
Abstract
In medical diagnosis, relying on only one type of biomarker is insufficient to accurately identify cancer. Blood-based multicancer early detection can help identify more than one type of cancer from a single blood sample. In this study, a super-resolution multispectral imaging nanoimmunosensor (srMINI) based on three quantum dots (QDs) of different color conjugated with streptavidin was developed for the simultaneous screening of various cancer biomarkers in blood at the single-molecule level. In the experiment, the srMINI chip was used to simultaneously detect three key cancer biomarkers: carcinoembryonic antigen (CEA), C-reactive protein (CRP), and alpha-fetoprotein (AFP). The srMINI chip exhibited 108 times higher detection sensitivity of 0.18-0.5 ag/mL (1.1-2.6 zM) for these cancer biomarkers than commercial enzyme-linked immunosorbent assay kits because of the absence of interfering signals from the substrate, establishing considerable potential for multiplex detection of cancer biomarkers in blood. Therefore, the simultaneous detection of various cancer biomarkers using the developed srMINI chip with high diagnostic precision and accuracy is expected to play a decisive role in early diagnosis or community screening as a single-molecule biosensor.
Collapse
Affiliation(s)
- Seungah Lee
- Department of Applied Chemistry and Institute of Natural Sciences, Kyung Hee University, Yongin -si, Gyeonggi-do 17104, Republic of Korea
| | - Junghwa Lee
- Department of Chemistry, Graduate School, Kyung Hee University, Yongin -si, Gyeonggi-do 17104, Republic of Korea
| | - Seong Ho Kang
- Department of Applied Chemistry and Institute of Natural Sciences, Kyung Hee University, Yongin -si, Gyeonggi-do 17104, Republic of Korea
- Department of Chemistry, Graduate School, Kyung Hee University, Yongin -si, Gyeonggi-do 17104, Republic of Korea
| |
Collapse
|
17
|
Boersma B, Poinot H, Pommier A. Stimulating the Antitumor Immune Response Using Immunocytokines: A Preclinical and Clinical Overview. Pharmaceutics 2024; 16:974. [PMID: 39204319 PMCID: PMC11357675 DOI: 10.3390/pharmaceutics16080974] [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: 06/03/2024] [Revised: 07/11/2024] [Accepted: 07/16/2024] [Indexed: 09/04/2024] Open
Abstract
Cytokines are immune modulators which can enhance the immune response and have been proven to be an effective class of immunotherapy. Nevertheless, the clinical use of cytokines in cancer treatment has faced several challenges associated with poor pharmacokinetic properties and the occurrence of adverse effects. Immunocytokines (ICKs) have emerged as a promising approach to overcome the pharmacological limitations observed with cytokines. ICKs are fusion proteins designed to deliver cytokines in the tumor microenvironment by taking advantage of the stability and specificity of immunoglobulin-based scaffolds. Several technological approaches have been developed. This review focuses on ICKs designed with the most impactful cytokines in the cancer field: IL-2, TNFα, IL-10, IL-12, IL-15, IL-21, IFNγ, GM-CSF, and IFNα. An overview of the pharmacological effects of the naked cytokines and ICKs tested for cancer therapy is detailed. A particular emphasis is given on the immunomodulatory effects of ICKs associated with their technological design. In conclusion, this review highlights active ways of development of ICKs. Their already promising results observed in clinical trials are likely to be improved with the advances in targeting technologies such as cytokine/linker engineering and the design of multispecific antibodies with tumor targeting and immunostimulatory functional properties.
Collapse
Affiliation(s)
- Bart Boersma
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland;
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland
| | - Hélène Poinot
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland;
- Translational Research Centre in Oncohaematology, University of Geneva, 1211 Geneva, Switzerland
| | - Aurélien Pommier
- UMR1240 Imagerie Moléculaire et Stratégies Théranostiques INSERM, Université Clermont Auvergne, BP 184, F-63005 Clermont-Ferrand, France
| |
Collapse
|
18
|
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
|
19
|
Magowan D, Abdulshafea M, Thompson D, Rajamoorthy SI, Owen R, Harris D, Prosser S. Blood-based biomarkers and novel technologies for the diagnosis of colorectal cancer and adenomas: a narrative review. Biomark Med 2024; 18:493-506. [PMID: 38900496 DOI: 10.1080/17520363.2024.2345583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 03/12/2024] [Indexed: 06/21/2024] Open
Abstract
Aim: Blood-based biomarkers have shown promise for diagnosing colorectal cancer (CRC) and adenomas (CRA). This review summarizes recent studies in this area. Methods: A literature search was undertaken for 01/01/2017-01/03/2023. Criteria included CRC, CRA, liquid-biopsy, blood-based tests and diagnosis. Results: 12,378 studies were reduced to 178 for data extraction. Sixty focused on proteomics, 53 on RNA species, 30 on cfDNA methylation, seven on antigens and autoantibodies and 28 on novel techniques. 169 case control and nine cohort studies. Number of participants ranged 100-54,297, mean age 58.26. CRC sensitivity and specificity ranged 9.10-100% and 20.40-100%, respectively. CRA sensitivity and specificity ranged 8.00-95.70% and 4.00-97.00%, respectively. Conclusion: Sensitive and specific blood-based tests exist for CRC and CRA. However, studies demonstrate heterogenous techniques and reporting quality. Further work should concentrate on validation and meta-analyzes.
Collapse
Affiliation(s)
- Drew Magowan
- Swansea University, Singleton Park, SA2 8PP, Swansea, UK
- Swansea Bay University Health Board, Department of General Surgery, Morriston Hospital, SA6 6NL, Swansea, UK
| | - Mansour Abdulshafea
- Swansea Bay University Health Board, Department of General Surgery, Morriston Hospital, SA6 6NL, Swansea, UK
| | - Dominic Thompson
- Swansea Bay University Health Board, Department of General Surgery, Morriston Hospital, SA6 6NL, Swansea, UK
| | - Shri-Ishvarya Rajamoorthy
- Swansea Bay University Health Board, Department of General Surgery, Morriston Hospital, SA6 6NL, Swansea, UK
| | - Rhiannon Owen
- Swansea University, Singleton Park, SA2 8PP, Swansea, UK
| | - Dean Harris
- Swansea University, Singleton Park, SA2 8PP, Swansea, UK
- Swansea Bay University Health Board, Department of General Surgery, Morriston Hospital, SA6 6NL, Swansea, UK
| | - Susan Prosser
- Swansea Bay University Health Board, Department of General Surgery, Morriston Hospital, SA6 6NL, Swansea, UK
| |
Collapse
|
20
|
Tsogka I, Mermiga E, Pagkali V, Kokkinos C, Economou A. A simplified lateral flow immunosensor for the assay of carcinoembryonic antigen in low-resource settings. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:2921-2929. [PMID: 38661387 DOI: 10.1039/d4ay00381k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Carcinoembryonic antigen (CEA) is a glycoprotein widely used as a tumor marker. In this work, a colorimetric lateral flow immunosensor is developed for rapid and low-cost quantification of CEA in human blood serum. The immunosensor consists of a glass fiber sample/conjugation pad, a nitrocellulose detection pad and a cellulose absorption pad. The detection is based on a sandwich immunoreaction: the sample/conjugation pad is modified with gold nanoparticles (GNPs)-labeled anti-CEA conjugate probes which bind to the CEA target molecules in the sample and the complexes are captured at capture anti-CEA immobilized at the test line. The color intensity of the test line, measured from a scanned image of the strip, is related to the CEA concentration in the sample. The different assay parameters are studied in detail. The linearity holds from 1.25 to 640 ng mL-1 of CEA, the instrumental and visual limits of detection are 0.45 and 0.63 ng mL-1, respectively, and the total assay time is 15 min. The specificity of the immunoassay versus other cancer biomarkers is satisfactory. The recovery in samples of human serum spiked with CEA is in the range of 81-118% and the coefficient of variation of the method is ≤10%. Results obtained with the lateral flow immunosensor correlated well with a reference radioimmunoassay method (R2 = 0.99). This immunosensor can be readily applied to CEA monitoring at the point-of-care (POC) or in resource-limited settings thanks to its low-cost and simplicity.
Collapse
Affiliation(s)
- Ioanna Tsogka
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis 157 71, Greece.
| | - Electra Mermiga
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis 157 71, Greece.
| | - Varvara Pagkali
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis 157 71, Greece.
| | - Christos Kokkinos
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis 157 71, Greece.
| | - Anastasios Economou
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis 157 71, Greece.
| |
Collapse
|
21
|
Dilek ON, Arslan Kahraman Dİ, Kahraman G. Carcinoembryonic antigen in the diagnosis, treatment, and follow-up of focal liver lesions. World J Gastrointest Surg 2024; 16:999-1007. [PMID: 38690060 PMCID: PMC11056666 DOI: 10.4240/wjgs.v16.i4.999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 02/02/2024] [Accepted: 03/13/2024] [Indexed: 04/22/2024] Open
Abstract
In this editorial review, we comment on the article published in the recent issue of the World Journal of Gastrointestinal Surgery. Carcinoembryonic antigen (CEA) is a fetal glycoprotein and can be secreted in very small amounts from healthy adults after birth. CEA is widely used not only for diagnostic tumor markers but also importantly for the management of some gastrointestinal tumors. The most common clinical use is surveillance for the monitoring of colorectal carcinoma. However, CEA can become elevated in several malign or benign characterized pathologies. Serum CEA level may vary depending on the location of the lesion, whether it metastasizes or not, and its histopathological characteristics. It has been determined that cases with high preoperative CEA have a more aggressive course and the risk of metastasis to the lymph tissue and liver increases. In this editorial review, we focused on evaluating the role of CEA in clinical practice with a holistic approach, including the diagnostic and prognostic significance of CEA in patients with focal liver lesions, the role of CEA in follow-up after definitive surgery, and also hepatic resection for metastasis, and the management of all patients with raised CEA.
Collapse
Affiliation(s)
- Osman Nuri Dilek
- Department of Surgery, İzmir Katip Celebi University, School of Medicine, İzmir 35150, Turkey
| | | | - Gökhan Kahraman
- Department of Radiology, Suluova State Hospital, Amasya 5500, Turkey
| |
Collapse
|
22
|
Seckinger A, Buatois V, Moine V, Daubeuf B, Richard F, Chatel L, Viandier A, Bosson N, Rousset E, Masternak K, Salgado-Pires S, Batista C, Mougin C, Juan-Bégeot F, Poitevin Y, Hose D. Targeting CEACAM5-positive solid tumors using NILK-2401, a novel CEACAM5xCD47 κλ bispecific antibody. Front Immunol 2024; 15:1378813. [PMID: 38720892 PMCID: PMC11076849 DOI: 10.3389/fimmu.2024.1378813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 04/10/2024] [Indexed: 05/12/2024] Open
Abstract
Background Blocking the CD47 "don't eat me"-signal on tumor cells with monoclonal antibodies or fusion proteins has shown limited clinical activity in hematologic malignancies and solid tumors thus far. Main side effects are associated with non-tumor targeted binding to CD47 particularly on blood cells. Methods We present here the generation and preclinical development of NILK-2401, a CEACAM5×CD47 bispecific antibody (BsAb) composed of a common heavy chain and two different light chains, one kappa and one lambda, determining specificity (so-called κλ body format). Results NILK-2401 is a fully human BsAb binding the CEACAM5 N-terminal domain on tumor cells by its lambda light chain arm with an affinity of ≈4 nM and CD47 with its kappa chain arm with an intendedly low affinity of ≈500 nM to enabling tumor-specific blockade of the CD47-SIRPα interaction. For increased activity, NILK-2401 features a functional IgG1 Fc-part. NILK-2401 eliminates CEACAM5-positive tumor cell lines (3/3 colorectal, 2/2 gastric, 2/2 lung) with EC50 for antibody-dependent cellular phagocytosis and antibody-dependent cellular cytotoxicity ranging from 0.38 to 25.84 nM and 0.04 to 0.25 nM, respectively. NILK-2401 binds neither CD47-positive/CEACAM5-negative cell lines nor primary epithelial cells. No erythrophagocytosis or platelet activation is observed. Quantification of the pre-existing NILK-2401-reactive T-cell repertoire in the blood of 14 healthy donors with diverse HLA molecules shows a low immunogenic potential. In vivo, NILK-2401 significantly delayed tumor growth in a NOD-SCID colon cancer model and a syngeneic mouse model using human CD47/human SIRPα transgenic mice and prolonged survival. In cynomolgus monkeys, single doses of 0.5 and 20 mg/kg were well tolerated; PK linked to anti-CD47 and Fc-binding seemed to be more than dose-proportional for Cmax and AUC0-inf. Data were validated in human FcRn TG32 mice. Combination of a CEACAM5-targeting T-cell engager (NILK-2301) with NILK-2401 can either boost NILK-2301 activity (Emax) up to 2.5-fold or allows reaching equal NILK-2301 activity at >600-fold (LS174T) to >3,000-fold (MKN-45) lower doses. Conclusion NILK-2401 combines promising preclinical activity with limited potential side effects due to the tumor-targeted blockade of CD47 and low immunogenicity and is planned to enter clinical testing.
Collapse
Affiliation(s)
- Anja Seckinger
- LamKap Bio beta AG, Pfäffikon SZ, Switzerland
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel (VUB), Jette, Belgium
| | | | - Valéry Moine
- Light Chain Bioscience (LCB), Plan-les-Ouates, Switzerland
| | - Bruno Daubeuf
- Light Chain Bioscience (LCB), Plan-les-Ouates, Switzerland
| | | | | | | | - Nicolas Bosson
- Light Chain Bioscience (LCB), Plan-les-Ouates, Switzerland
| | | | | | | | | | | | | | - Yves Poitevin
- Light Chain Bioscience (LCB), Plan-les-Ouates, Switzerland
| | - Dirk Hose
- LamKap Bio beta AG, Pfäffikon SZ, Switzerland
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel (VUB), Jette, Belgium
| |
Collapse
|
23
|
Li X, Hu M, Wang Z, Liu M, Chen Y. Prevalence of diverse colorectal polyps and risk factors for colorectal carcinoma in situ and neoplastic polyps. J Transl Med 2024; 22:361. [PMID: 38632639 PMCID: PMC11022362 DOI: 10.1186/s12967-024-05111-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 03/20/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Most colorectal cancers originate from precancerous polyps. This study aimed to determine the prevalence of colorectal polyps with diverse pathological morphologies and to explore the risk factors for colorectal carcinoma in situ (CCS) and neoplastic polyps. METHODS Inpatients admitted from January 2018 to May 2023 were screened through the hospital information system. Polyps were classified according to pathological morphology. The prevalence of polyps was described by frequency and 95% confidence interval. Univariate and multivariate logistic regression analyses were used to explore the risk factors for CCS and neoplastic polyps. RESULTS In total, 2329 individuals with 3550 polyps were recruited. Among all patients, 76.99% had neoplastic polyps and 44.31% had advanced adenomas. Tubular adenoma had the highest prevalence at 60.15%, and the prevalence of CCS was 3.86%. Patients with a colorectal polyp diameter ≥ 1.0 cm or number ≥ 3 were 8.07 times or 1.98 times more likely to develop CCS than were those with a diameter < 1.0 cm or number < 3, respectively (OR 8.07, 95%CI 4.48-14.55, p < 0.0001; and OR 1.98, 95%CI 1.27-3.09, p = 0.002). The risk of CCS with schistosome egg deposition was also significantly increased (OR 2.70, 95%CI 1.05-6.98). The higher the levels of carbohydrate antigen (CA) 724 (OR 1.01, 95%CI 1.00-1.02) and CA211 (OR 1.16, 95%CI 1.03-1.32) in patients with colorectal polyps were, the greater the risk of CCS. When colorectal neoplastic polyps were analyzed, we discovered that for each 1-year increase in age, the risk of neoplastic polyps increased by 3% (OR 1.03, 95%CI 1.02-1.04), p < 0.0001. Patients with a polyp diameter ≥ 1.0 cm had a 2.11-fold greater risk of neoplastic polyps compared to diameter < 1.0 cm patients (OR 3.11, 95%CI 2.48-3.92), p < 0.0001. In addition, multiple polyps and CA199 levels are risk factors for neoplastic polyps. CONCLUSION More than 3/4 of colorectal polyp patients have neoplastic polyps. Patients are more inclined to develop CCS and neoplastic polyps if they have large polyps (> 1.0 cm) or multifocal polyps. The levels of the tumor markers CA724 and CA211 show some potential usefulness for predicting CCS and may be exploited for early identification of high-risk populations.
Collapse
Affiliation(s)
- Xiaojuan Li
- Department of Gastroenterology, Minhang Hospital, Fudan University, Shanghai, 201199, China
| | - Mengting Hu
- Department of General Medicine, Minhang Hospital, Fudan University, Shanghai, 201199, China
| | - Zhangjun Wang
- Proprietary Trading Department, Huaan Securities Co., Ltd, Shanghai, 200120, China
| | - Mei Liu
- Department of General Medicine, Minhang Hospital, Fudan University, Shanghai, 201199, China.
| | - Ying Chen
- Department of Gastroenterology, Minhang Hospital, Fudan University, Shanghai, 201199, China.
| |
Collapse
|
24
|
Ren F, Fei Q, Qiu K, Zhang Y, Zhang H, Sun L. Liquid biopsy techniques and lung cancer: diagnosis, monitoring and evaluation. J Exp Clin Cancer Res 2024; 43:96. [PMID: 38561776 PMCID: PMC10985944 DOI: 10.1186/s13046-024-03026-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 03/24/2024] [Indexed: 04/04/2024] Open
Abstract
Lung cancer stands as the most prevalent form of cancer globally, posing a significant threat to human well-being. Due to the lack of effective and accurate early diagnostic methods, many patients are diagnosed with advanced lung cancer. Although surgical resection is still a potential means of eradicating lung cancer, patients with advanced lung cancer usually miss the best chance for surgical treatment, and even after surgical resection patients may still experience tumor recurrence. Additionally, chemotherapy, the mainstay of treatment for patients with advanced lung cancer, has the potential to be chemo-resistant, resulting in poor clinical outcomes. The emergence of liquid biopsies has garnered considerable attention owing to their noninvasive nature and the ability for continuous sampling. Technological advancements have propelled circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), extracellular vesicles (EVs), tumor metabolites, tumor-educated platelets (TEPs), and tumor-associated antigens (TAA) to the forefront as key liquid biopsy biomarkers, demonstrating intriguing and encouraging results for early diagnosis and prognostic evaluation of lung cancer. This review provides an overview of molecular biomarkers and assays utilized in liquid biopsies for lung cancer, encompassing CTCs, ctDNA, non-coding RNA (ncRNA), EVs, tumor metabolites, TAAs and TEPs. Furthermore, we expound on the practical applications of liquid biopsies, including early diagnosis, treatment response monitoring, prognostic evaluation, and recurrence monitoring in the context of lung cancer.
Collapse
Affiliation(s)
- Fei Ren
- Department of Geriatrics, The First Hospital of China Medical University, Shen Yang, 110000, China
| | - Qian Fei
- Department of Oncology, Shengjing Hospital of China Medical University, Shen Yang, 110000, China
| | - Kun Qiu
- Thoracic Surgery, The First Hospital of China Medical University, Shen Yang, 110000, China
| | - Yuanjie Zhang
- Thoracic Surgery, The First Hospital of China Medical University, Shen Yang, 110000, China
| | - Heyang Zhang
- Department of Hematology, The First Hospital of China Medical University, Shen Yang, 110000, China.
| | - Lei Sun
- Thoracic Surgery, The First Hospital of China Medical University, Shen Yang, 110000, China.
| |
Collapse
|
25
|
Galeș LN, Păun MA, Anghel RM, Trifănescu OG. Cancer Screening: Present Recommendations, the Development of Multi-Cancer Early Development Tests, and the Prospect of Universal Cancer Screening. Cancers (Basel) 2024; 16:1191. [PMID: 38539525 PMCID: PMC10969110 DOI: 10.3390/cancers16061191] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/08/2024] [Accepted: 03/15/2024] [Indexed: 11/11/2024] Open
Abstract
Cancer continues to pose a considerable challenge to global health. In the search for innovative strategies to combat this complex enemy, the concept of universal cancer screening has emerged as a promising avenue for early detection and prevention. In contrast to targeted approaches that focus on specific populations or high-risk individuals, universal screening seeks to cast a wide net to detect incipient malignancies in different demographic groups. This paradigm shift in cancer care underscores the importance of comprehensive screening programs that go beyond conventional boundaries. As our understanding of the complex molecular and genetic basis of cancer deepens, the need to develop comprehensive screening methods becomes increasingly apparent. In this article, we look at the rationale and potential benefits of universal cancer screening.
Collapse
Affiliation(s)
- Laurenția Nicoleta Galeș
- Department of Oncology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (L.N.G.); (R.M.A.); (O.G.T.)
- Department of Medical Oncology II, Prof. Dr. Al. Trestioreanu Institute of Oncology, 022328 Bucharest, Romania
| | - Mihai-Andrei Păun
- Department of Radiotherapy II, Prof. Dr. Al. Trestioreanu Institute of Oncology, 022328 Bucharest, Romania
| | - Rodica Maricela Anghel
- Department of Oncology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (L.N.G.); (R.M.A.); (O.G.T.)
- Department of Radiotherapy II, Prof. Dr. Al. Trestioreanu Institute of Oncology, 022328 Bucharest, Romania
| | - Oana Gabriela Trifănescu
- Department of Oncology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (L.N.G.); (R.M.A.); (O.G.T.)
- Department of Radiotherapy II, Prof. Dr. Al. Trestioreanu Institute of Oncology, 022328 Bucharest, Romania
| |
Collapse
|
26
|
Mehta D, Kaur S, Nagaiah TC. Realizing the label-free sensitive detection of carcinoembryogenic antigen (CEA) in blood serum via a MNC-decorated flexible immunosensor. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:1473-1479. [PMID: 38404261 DOI: 10.1039/d3ay02073h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
A label-free electrochemical immunosensor utilising nitrogen-rich mesoporous carbon (MNC) as the substrate material was developed for the sensitive quantification of carcinoembryonic antigen (CEA). The synergic interactions between MNC and AbCEA also eliminated the need for coupling agents such as EDC/NHS. The novel immunosensor demonstrated a wide detection range from 500 fM (9.04 pg mL-1) to 50 nM (1 μg mL-1) and a low detection limit (LOD) of 500 fM. Moreover, the immunosensor showed sensitivities of 12.27 mA nM-1 cm-2 and 0.066 mA nM-1 cm-2 for detecting CEA in the linear ranges 10 pM to 1 nM and 2 nM to 50 nM, respectively, while maintaining long-term storage stability of 6 weeks. Analysis of real serum sample analysis yielded highly accurate results with recovery rates ranging from 99.3% to 103.7%. Furthermore, the developed paper-based screen-printed electrode exhibited a similar detection range, suggesting its potential for use in point-of-care detection devices in future applications.
Collapse
Affiliation(s)
- Daisy Mehta
- Department of Chemistry, Indian Institute of Technology Ropar, Rupnagar, Punjab 140001, India.
| | - Sukhjot Kaur
- Department of Chemistry, Indian Institute of Technology Ropar, Rupnagar, Punjab 140001, India.
| | - Tharamani C Nagaiah
- Department of Chemistry, Indian Institute of Technology Ropar, Rupnagar, Punjab 140001, India.
| |
Collapse
|
27
|
Dubey AK, Kaur I, Madaan R, Raheja S, Bala R, Garg M, Kumar S, Lather V, Mittal V, Pandita D, Gundamaraju R, Singla RK, Sharma R. Unlocking the potential of oncology biomarkers: advancements in clinical theranostics. Drug Metab Pers Ther 2024; 39:5-20. [PMID: 38469723 DOI: 10.1515/dmpt-2023-0056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 01/11/2024] [Indexed: 03/13/2024]
Abstract
INTRODUCTION Cancer biomarkers have revolutionized the field of oncology by providing valuable insights into tumor changes and aiding in screening, diagnosis, prognosis, treatment prediction, and risk assessment. The emergence of "omic" technologies has enabled biomarkers to become reliable and accurate predictors of outcomes during cancer treatment. CONTENT In this review, we highlight the clinical utility of biomarkers in cancer identification and motivate researchers to establish a personalized/precision approach in oncology. By extending a multidisciplinary technology-based approach, biomarkers offer an alternative to traditional techniques, fulfilling the goal of cancer therapeutics to find a needle in a haystack. SUMMARY AND OUTLOOK We target different forms of cancer to establish a dynamic role of biomarkers in understanding the spectrum of malignancies and their biochemical and molecular characterization, emphasizing their prospective contribution to cancer screening. Biomarkers offer a promising avenue for the early detection of human cancers and the exploration of novel technologies to predict disease severity, facilitating maximum survival and minimum mortality rates. This review provides a comprehensive overview of the potential of biomarkers in oncology and highlights their prospects in advancing cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Ankit Kumar Dubey
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, 34753 Sichuan University , Chengdu, P.R. China
- iGlobal Research and Publishing Foundation, New Delhi, India
| | - Ishnoor Kaur
- Chitkara College of Pharmacy, 154025 Chitkara University Punjab , Rajpura, India
| | - Reecha Madaan
- Chitkara College of Pharmacy, 154025 Chitkara University Punjab , Rajpura, India
| | - Shikha Raheja
- Jan Nayak Ch. Devi Lal Memorial College of Pharmacy, Sirsa, Haryana, India
| | - Rajni Bala
- Chitkara College of Pharmacy, 154025 Chitkara University Punjab , Rajpura, India
| | - Manoj Garg
- Amity Institute of Molecular Medicine & Stem Cell Research, 77282 Amity University, Sector-125 , Noida, India
| | - Suresh Kumar
- Department of Pharmaceutical Sciences and Drug Research, 429174 Punjabi University Patiala , Patiala, India
| | - Viney Lather
- Amity Institute of Pharmacy, 77282 Amity University , Noida, India
| | - Vineet Mittal
- Department of Pharmaceutical Sciences, 29062 Maharshi Dayanand University , Rohtak, Haryana, India
| | - Deepti Pandita
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University, PushpVihar, 633274 Govt. of NCT of Delhi , New Delhi, India
- Centre for Advanced Formulation and Technology (CAFT), Delhi Pharmaceutical Sciences and Research University, PushpVihar, Govt. of NCT of Delhi, New Delhi, India
| | - Rohit Gundamaraju
- ER Stress and Mucosal Immunology Lab, School of Health Sciences, 8785 University of Tasmania , Launceston, Tasmania, Australia
- School of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Rajeev K Singla
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, 34753 Sichuan University , Chengdu, P.R. China
- School of Pharmaceutical Sciences, 34753 Lovely Professional University , Phagwara, Punjab, India
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, 80095 Banaras Hindu University , Varanasi, Uttar Pradesh, India
| |
Collapse
|
28
|
Seow-En I, Koh YX, Zhao Y, Ang BH, Tan IEH, Chok AY, Tan EJKW, Au MKH. Predictive modeling algorithms for liver metastasis in colorectal cancer: A systematic review of the current literature. Ann Hepatobiliary Pancreat Surg 2024; 28:14-24. [PMID: 38129965 PMCID: PMC10896689 DOI: 10.14701/ahbps.23-078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 08/16/2023] [Indexed: 12/23/2023] Open
Abstract
This study aims to assess the quality and performance of predictive models for colorectal cancer liver metastasis (CRCLM). A systematic review was performed to identify relevant studies from various databases. Studies that described or validated predictive models for CRCLM were included. The methodological quality of the predictive models was assessed. Model performance was evaluated by the reported area under the receiver operating characteristic curve (AUC). Of the 117 articles screened, seven studies comprising 14 predictive models were included. The distribution of included predictive models was as follows: radiomics (n = 3), logistic regression (n = 3), Cox regression (n = 2), nomogram (n = 3), support vector machine (SVM, n = 2), random forest (n = 2), and convolutional neural network (CNN, n = 2). Age, sex, carcinoembryonic antigen, and tumor staging (T and N stage) were the most frequently used clinicopathological predictors for CRCLM. The mean AUCs ranged from 0.697 to 0.870, with 86% of the models demonstrating clear discriminative ability (AUC > 0.70). A hybrid approach combining clinical and radiomic features with SVM provided the best performance, achieving an AUC of 0.870. The overall risk of bias was identified as high in 71% of the included studies. This review highlights the potential of predictive modeling to accurately predict the occurrence of CRCLM. Integrating clinicopathological and radiomic features with machine learning algorithms demonstrates superior predictive capabilities.
Collapse
Affiliation(s)
- Isaac Seow-En
- Department of Colorectal Surgery, Singapore General Hospital and National Cancer Centre Singapore, Singapore
| | - Ye Xin Koh
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital and National Cancer Centre Singapore, Singapore
- Duke-National University of Singapore Medical School, Singapore
- Liver Transplant Service, SingHealth Duke-National University of Singapore Transplant Centre, Singapore
| | - Yun Zhao
- Department of Colorectal Surgery, Singapore General Hospital and National Cancer Centre Singapore, Singapore
- Group Finance Analytics, Singapore Health Services, Singapore
| | - Boon Hwee Ang
- Group Finance Analytics, Singapore Health Services, Singapore
| | | | - Aik Yong Chok
- Department of Colorectal Surgery, Singapore General Hospital and National Cancer Centre Singapore, Singapore
| | - Emile John Kwong Wei Tan
- Department of Colorectal Surgery, Singapore General Hospital and National Cancer Centre Singapore, Singapore
| | - Marianne Kit Har Au
- Group Finance Analytics, Singapore Health Services, Singapore
- Finance, SingHealth Community Hospitals, Singapore
| |
Collapse
|
29
|
West-Szymanski DC, Zhang Z, Cui XL, Kowitwanich K, Gao L, Deng Z, Dougherty U, Williams C, Merkle S, Moore M, He C, Bissonnette M, Zhang W. Machine learning identifies cell-free DNA 5-hydroxymethylation biomarkers that detect occult colorectal cancer in PLCO Screening Trial subjects. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.25.581955. [PMID: 38464122 PMCID: PMC10925134 DOI: 10.1101/2024.02.25.581955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Background Colorectal cancer (CRC) is a leading cause of cancer-related mortality, and CRC detection through screening improves survival rates. A promising avenue to improve patient screening compliance is the development of minimally-invasive liquid biopsy assays that target CRC biomarkers on circulating cell-free DNA (cfDNA) in peripheral plasma. In this report, we identify cfDNA biomarker candidate genes bearing the epigenetic mark 5-hydroxymethylcytosine (5hmC) that diagnose occult CRC up to 36 months prior to clinical diagnosis using the Prostate, Lung, Colorectal and Ovarian (PLCO) Cancer Screening Trial samples. Methods Archived PLCO Trial plasma samples containing cfDNA were obtained from the National Cancer Institute (NCI) biorepositories. Study subjects included those who were diagnosed with CRC within 36 months of blood collection (i.e., case, n = 201) and those who were not diagnosed with any cancer during an average of 16.3 years of follow-up (i.e., controls, n = 402). Following the extraction of 3 - 8 ng cfDNA from less than 300 microliters plasma, we employed the sensitive 5hmC-Seal chemical labeling approach, followed by next-generation sequencing (NGS). We then conducted association studies and machine-learning modeling to analyze the genome-wide 5hmC profiles within training and validation groups that were randomly selected at a 2:1 ratio. Results Despite the technical challenges associated with the PLCO samples (e.g., limited plasma volumes, low cfDNA amounts, and long archival times), robust genome-wide 5hmC profiles were successfully obtained from these samples. Association analyses using the Cox proportional hazards models suggested several epigenetic pathways relevant to CRC development distinguishing cases from controls. A weighted Cox model, comprised of 32-associated gene bodies, showed predictive detection value for CRC as early as 24-36 months prior to overt tumor presentation, and a trend for increased predictive power was observed for blood samples collected closer to CRC diagnosis. Notably, the 5hmC-based predictive model showed comparable performance regardless of sex and self-reported race/ethnicity, and significantly outperformed risk factors such as age and obesity according to BMI (body mass index). Additionally, further improvement of predictive performance was achieved by combining the 5hmC-based model and risk factors for CRC. Conclusions An assay of 5hmC epigenetic signals on cfDNA revealed candidate biomarkers with the potential to predict CRC occurrence despite the absence of clinical symptoms or the availability of effective predictors. Developing a minimally-invasive clinical assay that detects 5hmC-modified biomarkers holds promise for improving early CRC detection and ultimately patient survival through higher compliance screening and earlier intervention. Future investigation to expand this strategy to prospectively collected samples is warranted.
Collapse
|
30
|
Williams L, Li L, Yazaki PJ, Wong P, Hong T, Poku EK, Hui S, Ghimire H, Shively JE, Kujawski M. Comparison of IL-2-antibody to IL-2-Fc with or without stereotactic radiation therapy in CEA immunocompetent mice with CEA positive tumors. Cancer Med 2024; 13:e6909. [PMID: 38317590 PMCID: PMC10905250 DOI: 10.1002/cam4.6909] [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: 06/02/2023] [Revised: 10/31/2023] [Accepted: 12/04/2023] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND The potent immune effects of interleukin-2 (IL-2) for cancer therapy can be increased by genetic fusion of IL-2 to the Fc domain of an antibody (IL-2-Fc) or tumor targeted by genetic fusion to a whole antibody known as an immunocytokine (ICK). METHODS An anti-CEA ICK (M5A-IL-2) was compared to an IL-2-Fc fusion protein using tumor therapy and PET imaging in CEA transgenic immunocompetent mice bearing CEA positive colon or breast tumors. Combination with stereotactic radiation therapy (SRT) was performed with either ICK or IL-2-Fc. RESULTS ICK and IL-2-Fc had comparable antitumor effects in both tumor models, although ICK had higher tumor uptake and slower blood clearance than an IL-2-Fc. Analysis of IFNγ+ /CD8+ and FoxP3+ /CD4+ T cells revealed higher levels of IFNγ-producing CD8+ T cells in ICK treated mice versus more efficient Treg elimination in IL-2-Fc treated mice. No significant or lasting toxicity was detected for either agent. Combination therapies with SRT revealed comparable efficacy and induction of immune memory for both ICK and IL-2-Fc when mice were rechallenged post-therapy. CONCLUSIONS IL-2-Fc had comparable antitumor efficacy to CEA-targeted M5A-IL-2 ICK, while both fusion proteins induced immune memory when combined with SRT. Differences in the therapeutic mechanisms of both agents were observed.
Collapse
Affiliation(s)
- Lindsay Williams
- Department of Immunology and Theranostics, Riggs Diabetes, Metabolism, and Research InstituteBeckman Research Institute of City of HopeDuarteCaliforniaUSA
| | - Lin Li
- Department of Immunology and Theranostics, Riggs Diabetes, Metabolism, and Research InstituteBeckman Research Institute of City of HopeDuarteCaliforniaUSA
| | - Paul J. Yazaki
- Department of Immunology and Theranostics, Riggs Diabetes, Metabolism, and Research InstituteBeckman Research Institute of City of HopeDuarteCaliforniaUSA
| | - Patty Wong
- Department of Immunology and Theranostics, Riggs Diabetes, Metabolism, and Research InstituteBeckman Research Institute of City of HopeDuarteCaliforniaUSA
| | - Teresa Hong
- Department of Immunology and Theranostics, Riggs Diabetes, Metabolism, and Research InstituteBeckman Research Institute of City of HopeDuarteCaliforniaUSA
| | - Erasmus K. Poku
- RadiopharmacyCity of HopeDuarteCaliforniaUSA
- Beckman Research Institute of City of HopeDuarteCaliforniaUSA
| | - Susanta Hui
- Department of Radiation OncologyCity of HopeDuarteCaliforniaUSA
- City of Hope Medical CenterDuarteCaliforniaUSA
| | - Hemendra Ghimire
- Department of Radiation OncologyCity of HopeDuarteCaliforniaUSA
- City of Hope Medical CenterDuarteCaliforniaUSA
| | - John E. Shively
- Department of Immunology and Theranostics, Riggs Diabetes, Metabolism, and Research InstituteBeckman Research Institute of City of HopeDuarteCaliforniaUSA
| | - Maciej Kujawski
- Department of Immunology and Theranostics, Riggs Diabetes, Metabolism, and Research InstituteBeckman Research Institute of City of HopeDuarteCaliforniaUSA
| |
Collapse
|
31
|
Ahmadipour M, Bhattacharya A, Sarafbidabad M, Syuhada Sazali E, Krishna Ghoshal S, Satgunam M, Singh R, Rezaei Ardani M, Missaoui N, Kahri H, Pal U, Ling Pang A. CA19-9 and CEA biosensors in pancreatic cancer. Clin Chim Acta 2024; 554:117788. [PMID: 38246211 DOI: 10.1016/j.cca.2024.117788] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 01/15/2024] [Accepted: 01/15/2024] [Indexed: 01/23/2024]
Abstract
Cancer is a complex pathophysiological condition causing millions of deaths each year. Early diagnosis is essential especially for pancreatic cancer. Existing diagnostic tools rely on circulating biomarkers such as Carbohydrate Antigen 19-9 (CA19-9) and Carcinoembryonic Antigen (CEA). Unfortunately, these markers are nonspecific and may be increased in a variety of disorders. Accordingly, diagnosis of pancreatic cancer generally involves more invasive approaches such as biopsy as well as imaging studies. Recent advances in biosensor technology have allowed the development of precise diagnostic tools having enhanced analytical sensitivity and specificity. Herein we examine these advances in the detection of cancer in general and in pancreatic cancer specifically. Furthermore, we highlight novel technologies in the measurement of CA19-9 and CEA and explore their future application in the early detection of pancreatic cancer.
Collapse
Affiliation(s)
- Mohsen Ahmadipour
- Institute of Power Engineering, Universiti Tenaga Nasional, 43650 Serdang, Selangor, Malaysia.
| | - Anish Bhattacharya
- Advanced Optical Materials Research Group, Department of Physics, Faculty of Science, Universiti Teknologi Malaysia, 81310 Skudai, Johor, Malaysia; Ibnu Sina Institute of Laser Centre, Universiti Teknologi Malaysia, 81310 Skudai, Johor, Malaysia
| | - Mohsen Sarafbidabad
- Biomedical Engineering Department, Faculty of Engineering, University of Isfahan, Isfahan, Iran
| | - Ezza Syuhada Sazali
- Advanced Optical Materials Research Group, Department of Physics, Faculty of Science, Universiti Teknologi Malaysia, 81310 Skudai, Johor, Malaysia; Ibnu Sina Institute of Laser Centre, Universiti Teknologi Malaysia, 81310 Skudai, Johor, Malaysia
| | - Sib Krishna Ghoshal
- Advanced Optical Materials Research Group, Department of Physics, Faculty of Science, Universiti Teknologi Malaysia, 81310 Skudai, Johor, Malaysia; Ibnu Sina Institute of Laser Centre, Universiti Teknologi Malaysia, 81310 Skudai, Johor, Malaysia
| | - Meenaloshini Satgunam
- Institute of Power Engineering, Universiti Tenaga Nasional, 43650 Serdang, Selangor, Malaysia; Department of Mechanical Engineering, Universiti Tenaga Nasional, 43650 Serdang, Selangor, Malaysia
| | - Ramesh Singh
- Institute of Power Engineering, Universiti Tenaga Nasional, 43650 Serdang, Selangor, Malaysia; Center of Advanced Manufacturing and Materials Processing (AMMP), Faculty of Engineering, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Mohammad Rezaei Ardani
- School of Materials and Mineral Resources Engineering, Universiti Sains Malaysia, Engineering Campus, 14300 Nibong Tebal, Pulau Pinang, Malaysia
| | - Nadhem Missaoui
- Laboratory of Interfaces and Advanced Materials, Faculty of Sciences, University of Monastir, Monastir, Tunisia
| | - Hamza Kahri
- Laboratory of Interfaces and Advanced Materials, Faculty of Sciences, University of Monastir, Monastir, Tunisia
| | - Ujjwal Pal
- Department of Analytical and Structural Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad, India
| | - Ai Ling Pang
- Department of Chemical Science, Faculty of Science, Universiti Tunku Abdul Rahman, 31900 Kampar, Perak, Malaysia
| |
Collapse
|
32
|
Li C, Xue G, Wu R, Zhang J, Cheng Y, Huang G, Xu H, Song Q, Cheng R, Shen Z, Xue C. Lighting up Lipidic Nanoflares with Self-Powered and Multivalent 3D DNA Rolling Motors for High-Efficiency MicroRNA Sensing in Serum and Living Cells. ACS APPLIED MATERIALS & INTERFACES 2024; 16:281-291. [PMID: 38156775 DOI: 10.1021/acsami.3c14718] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Intelligent DNA nanomachines are powerful and versatile molecular tools for bioimaging and biodiagnostic applications; however, they are generally constrained by complicated synthetic processes and poor reaction efficiencies. In this study, we developed a simple and efficient molecular machine by coupling a self-powered rolling motor with a lipidic nanoflare (termed RMNF), enabling high-contrast, robust, and rapid probing of cancer-associated microRNA (miRNA) in serum and living cells. The lipidic nanoflare is a cholesterol-based lipidic micelle decorated with hairpin-shaped tracks that can be facilely synthesized by stirring in buffered solution, whereas the 3D rolling motor (3D RM) is a rigidified tetrahedral DNA scaffold equipped with four single-stranded "legs" each silenced by a locking strand. Once exposed to the target miRNA, the 3D RM can be activated, followed by self-powered precession based on catalyzed hairpin assembly (CHA) and lighting up of the lipidic nanoflare. Notably, the multivalent 3D RM that moves using four DNA legs, which allows the motor to continuously and acceleratedly interreact with DNA tracks rather than dissociate from the surface of the nanoflare, yielded a limit of detection (LOD) of 500 fM at 37 °C within 1.5 h. Through the nick-hidden and rigidified structure design, RMNF exhibits high biostability and a low false-positive signal under complex physiological settings. The final application of RMNF for miRNA detection in clinical samples and living cells demonstrates its considerable potential for biomedical imaging and clinical diagnosis.
Collapse
Affiliation(s)
- Chan Li
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, PR China
| | - Guohui Xue
- Department of Clinical Laboratory, Jiujiang No. 1 People's Hospital, Jiujiang, Jiangxi 332000, PR China
| | - Rong Wu
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, PR China
| | - Jing Zhang
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, PR China
| | - Yinghao Cheng
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, PR China
| | - Guoqiao Huang
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, PR China
| | - Huo Xu
- College of Materials and Chemical Engineering, Minjiang University, Fuzhou, Fujian 350108, PR China
| | - Qiufeng Song
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, PR China
| | - Ruize Cheng
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, PR China
| | - Zhifa Shen
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, PR China
| | - Chang Xue
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, PR China
| |
Collapse
|
33
|
Huang S, Chang S, Liao T, Yang M. Detection and clinical significance of CEACAM5 methylation in colorectal cancer patients. Cancer Sci 2024; 115:270-282. [PMID: 37942534 PMCID: PMC10823287 DOI: 10.1111/cas.16012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 10/19/2023] [Accepted: 10/25/2023] [Indexed: 11/10/2023] Open
Abstract
Colorectal cancer (CRC) is a globally common cancer, and the serum carcinoembryonic antigen (sCEA) is widely applied as a diagnostic and prognostic tumor marker in CRC. This study aimed to elucidate the mechanism of CEA expression and corresponding clinical features to improve prognostic assessments. In CRC cells, hypomethylation of the CEACAM5 promoter enhanced CEA expression in HCT116 and HT29 cells with 5-aza-2'-deoxycytidine (5-Aza-dC) treatment. Our clinical data indicated that 64.7% (101/156) of CRC patients had an sCEA level above the normal range, and 76.2% (77/101) of those patients showed a lower average CpG methylation level of the CEACAM5 promoter. The methylation analysis showed that both CRC cell lines and patient samples shared the same critical methylation CpG regions at -200 to -500 and -1000 to -1400 bp of the CEACAM5 promoter. Patients with hypermethylation of the CEACAM5 promoter showed features of a BRAF mutation, TGFB2 mutation, microsatellite instability-high, and preference for right-sided colorectal cancer and peritoneal seeding presentation that had a similar clinical character to the consensus molecular subtype 1 (CMS1) of colorectal cancer. Additionally, hypermethylation of the CEACAM5 promoter combined with evaluated sCEA demonstrated the worst survival among the patients. Therefore, the methylation status of the CEACAM5 promoter also served as an effective biomarker for assessing disease prognosis. Results indicated that DNA methylation is a major regulatory mechanism for CEA expression in colorectal cancer. Moreover, our data also highlighted that patients in a subgroup who escaped from inactivation by DNA methylation had distinct clinical and pathological features and the worst survival.
Collapse
Affiliation(s)
- Sheng‐Chieh Huang
- Institute of Clinical MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
- Division of Colorectal Surgery, Department of SurgeryTaipei Veterans General HospitalTaipeiTaiwan
- Faculty of Medicine, School of MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Shih‐Ching Chang
- Division of Colorectal Surgery, Department of SurgeryTaipei Veterans General HospitalTaipeiTaiwan
- Faculty of Medicine, School of MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Tsai‐Tsen Liao
- Graduate Institute of Medical Sciences, College of MedicineTaipei Medical UniversityTaipeiTaiwan
- Cell Physiology and Molecular Image Research Center, Wan Fang HospitalTaipei Medical UniversityTaipeiTaiwan
- Research Center of Cancer Translational MedicineTaipei Medical UniversityTaipeiTaiwan
- Cancer Research CenterTaipei Medical University HospitalTaipeiTaiwan
| | - Muh‐Hwa Yang
- Institute of Clinical MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
- Cancer and Immunology Research CenterNational Yang Ming Chiao UniversityTaipeiTaiwan
- Department of OncologyTaipei Veterans General HospitalTaipeiTaiwan
| |
Collapse
|
34
|
Song Q, Zheng Y, Zhong G, Wang S, He C, Li M. Application of Nanoparticles in the Diagnosis and Treatment of Colorectal Cancer. Anticancer Agents Med Chem 2024; 24:1305-1326. [PMID: 39129164 PMCID: PMC11497148 DOI: 10.2174/0118715206323900240807110122] [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: 05/15/2024] [Revised: 06/26/2024] [Accepted: 07/12/2024] [Indexed: 08/13/2024]
Abstract
Colorectal cancer is a common malignant tumor with high morbidity and mortality rates, imposing a huge burden on both patients and the healthcare system. Traditional treatments such as surgery, chemotherapy and radiotherapy have limitations, so finding more effective diagnostic and therapeutic tools is critical to improving the survival and quality of life of colorectal cancer patients. While current tumor targeting research mainly focuses on exploring the function and mechanism of molecular targets and screening for excellent drug targets, it is crucial to test the efficacy and mechanism of tumor cell therapy that targets these molecular targets. Selecting the appropriate drug carrier is a key step in effectively targeting tumor cells. In recent years, nanoparticles have gained significant interest as gene carriers in the field of colorectal cancer diagnosis and treatment due to their low toxicity and high protective properties. Nanoparticles, synthesized from natural or polymeric materials, are NM-sized particles that offer advantages such as low toxicity, slow release, and protection of target genes during delivery. By modifying nanoparticles, they can be targeted towards specific cells for efficient and safe targeting of tumor cells. Numerous studies have demonstrated the safety, efficiency, and specificity of nanoparticles in targeting tumor cells, making them a promising gene carrier for experimental and clinical studies. This paper aims to review the current application of nanoparticles in colorectal cancer diagnosis and treatment to provide insights for targeted therapy for colorectal cancer while also highlighting future prospects for nanoparticle development.
Collapse
Affiliation(s)
- Qiuyu Song
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yifeng Zheng
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Guoqiang Zhong
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shanping Wang
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chengcheng He
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mingsong Li
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
35
|
Tang F, Huang CW, Tang ZH, Lu SL, Bai T, Huang Q, Li XZ, Zhang B, Wu FX. Prognostic role of serum carcinoembryonic antigen in patients receiving liver resection for colorectal cancer liver metastasis: A meta-analysis. World J Gastrointest Surg 2023; 15:2890-2906. [PMID: 38222018 PMCID: PMC10784827 DOI: 10.4240/wjgs.v15.i12.2890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 11/03/2023] [Accepted: 11/28/2023] [Indexed: 12/27/2023] Open
Abstract
BACKGROUND Carcinoembryonic antigen (CEA) is a broad-spectrum tumor marker for differential diagnosis, monitoring, and response assessment of a variety of malignancies. AIM To evaluate 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 cut-off date of February 27, 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 11143 CRCLM patients. The results showed that a high pre-LR serum CEA level was correlated with poor overall survival (OS) [hazard ratio (HR) = 1.61, 95% confidence interval (CI): 1.49-1.75, P < 0.001] and recurrence-free survival (HR = 1.27, 95%CI: 1.11-1.45, P < 0.001) in CRCLM patients. A high post-LR serum CEA level predicted poor OS (HR = 2.66, 95%CI: 2.10-3.38, P < 0.001). A comparison by treatment modality, analysis modality, patient source, and cutoff-value showed that overall, high preoperative and postoperative serum CEA levels remained correlated with a poor prognosis. CONCLUSION This study concluded that high pre-LR and post-LR serum CEA levels were significantly correlated with a poor prognosis in CRCLM patients.
Collapse
Affiliation(s)
- Fan Tang
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Cheng-Wen Huang
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Zhi-Hong Tang
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Shao-Long Lu
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Tao Bai
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Qing Huang
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Xing-Zhi Li
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Bin Zhang
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Fei-Xiang Wu
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment, Ministry of Education, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
36
|
Bhagat A, Lyerly HK, Morse MA, Hartman ZC. CEA vaccines. Hum Vaccin Immunother 2023; 19:2291857. [PMID: 38087989 PMCID: PMC10732609 DOI: 10.1080/21645515.2023.2291857] [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: 09/13/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023] Open
Abstract
Carcinoembryonic antigen (CEA) is a glycosylated cell surface oncofetal protein involved in adhesion, proliferation, and migration that is highly upregulated in multiple carcinomas and has long been a promising target for cancer vaccination. This review summarizes the progress to date in the development of CEA vaccines, examining both pre-clinical and clinical studies across a variety of vaccine platforms that in aggregate, begin to reveal some critical insights. These studies demonstrate the ability of CEA vaccines to break immunologic tolerance and elicit CEA-specific immunity, which associates with improved clinical outcomes in select individuals. Approaches that have combined replicating viral vectors, with heterologous boosting and different adjuvant strategies have been particularly promising but, these early clinical trial results will require confirmatory studies. Collectively, these studies suggest that clinical efficacy likely depends upon harnessing a potent vaccine combination in an appropriate clinical setting to fully realize the potential of CEA vaccination.
Collapse
Affiliation(s)
- Anchit Bhagat
- Department of Surgery, Division of Surgical Sciences, Duke University, Durham, NC, USA
| | - Herbert K. Lyerly
- Department of Surgery, Division of Surgical Sciences, Duke University, Durham, NC, USA
- Department of Pathology, Duke University, Durham, NC, USA
- Department of Integrative Immunobiology, Duke University, Durham, NC, USA
| | - Michael A. Morse
- Department of Surgery, Division of Surgical Sciences, Duke University, Durham, NC, USA
- Department of Medicine, Duke University, Durham, NC, USA
| | - Zachary C. Hartman
- Department of Surgery, Division of Surgical Sciences, Duke University, Durham, NC, USA
- Department of Pathology, Duke University, Durham, NC, USA
- Department of Integrative Immunobiology, Duke University, Durham, NC, USA
| |
Collapse
|
37
|
Huang P, Meng L, Pang J, Huang H, Ma J, He L, Amani P. Development of a high-performance label-free electrochemical immunosensor for early cancer diagnosis using anti-CEA/Ag-MOF/GO/GCE nanocomposite. ENVIRONMENTAL RESEARCH 2023; 238:117178. [PMID: 37734580 DOI: 10.1016/j.envres.2023.117178] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/07/2023] [Accepted: 09/19/2023] [Indexed: 09/23/2023]
Abstract
In order to detect carcinoembryonic antigen (CEA) as a tumor marker in lung cancer for early cancer diagnosis, this study aimed to develop a label-free electrochemical immunosensor based on the immobilization of an Anti-CEA antibody on a metal-organic framework (MOF)-graphene oxide nanocomposite modified glassy carbon electrode (Anti-CEA/Ag-MOF/GO/GCE). Ag-MOF/GO nanocomposite was prepared on the GCE surface using the ultrasonic irradiation method, and Anti-CEA antibody was subsequently immobilized on the surface. Analysis of the crystal structure and morphology of the modified electrode using FE-SEM and XRD revealed that the correct combination of GO nanosheets and Ag-MOF nanoparticles produced a high surface area to trap the antibodies. Electrochemical tests utilizing the CV and DPV methods revealed that the immunosensor's sensitivity, stability, and selectivity were improved by Anti-CEA/Ag-MOF/GO/GCE. Results showed that, with a detection limit of 0.005 ng/mL, the change in the reduction peak current was inversely correlated with the logarithm concentration of CEA in the range of 10-3 to 5000 ng/mL. The suggested CEA immunosensor's applicability in a human serum sample was investigated, and findings of analytical studies via standard addition technique for both ELISA and DPV assays revealed that significant agreement existed between the outcomes of the two assays. Additionally, the recoveries ranged from 99.00% to 99.25%, and all relative standard deviations (RSDs) for the sample detections were below 5.01%, indicating satisfactory accuracy in results measured with the proposed CEA immunosensor, indicating that the prepared CEA immunosensor in this study can be used in clinical applications and human fluids.
Collapse
Affiliation(s)
- Peng Huang
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi, 533000, China
| | - Lingzhang Meng
- Center for Systemic Inflammation Research (CSIR), School of Preclinical Medicine, Youjiang Medical University for Nationalities, Guangxi, 533000, China
| | - Jun Pang
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi, 533000, China
| | - Haiting Huang
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi, 533000, China
| | - Jing Ma
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi, 533000, China
| | - Linlin He
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi, 533000, China
| | - Parnian Amani
- Department of Molecular and Cell Biology, Faculty of Basic Sciences, University of Mazandaran, Babolsar, Iran.
| |
Collapse
|
38
|
Tao C, Rouhi J. A biosensor based on graphene oxide nanocomposite for determination of carcinoembryonic antigen in colorectal cancer biomarker. ENVIRONMENTAL RESEARCH 2023; 238:117113. [PMID: 37696325 DOI: 10.1016/j.envres.2023.117113] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/06/2023] [Accepted: 09/09/2023] [Indexed: 09/13/2023]
Abstract
Colorectal cancer is still a major global health concern, and early detection and accurate biomarker analyses are critical to its successful management. This paper describes the design and testing of a new biosensor based on a graphene oxide (GO) nanocomposite for the exact measurement of carcinoembryonic antigen (CEA), a well-known biomarker for colorectal cancer. The current study attempted to create a highly sensitive immunosensor for sensitive measurement of CEA based on a polypropylene-imine-dendrimer (PPI) and GO nanocomposite on GCE (PPI/GO/GCE). The PPI/GO nanocomposite served as an appropriate biocompatible nanostructure with a large surface area for immobilizing carcinoembryonic antigen (anti-CEA) and bovine serum albumin (BSA) molecules (BSA/anti-CEA/PPI/GO/GCE), thereby promoting the selectivity of electrochemical immunosensors, according to structural and electrochemical studies. Results showed that the BSA/anti-CEA/PPI/GO/GCE as a selective, sensitive, and stable immunosensor revealed a wide linear response from 0.001 to 2000 ng/mL, and a limit of detection of 0.3 pg/mL, which indicated comparable or better performance towards the CEA immunosensors in recent reports in the literature. This was due to the synergetic effect of the GO nanosheets and PPI with porous structure and more conductivity. Analytical results showed values of RSD (4.49%-5.04%) and recovery (90.00%-99.98%) are suitable for effective and accurate practical assessments in CEA in clinical samples. The capacity of the BSA/anti-CEA/PPI/GO/GCE to determine CEA in human blood was studied.
Collapse
Affiliation(s)
- Chenyu Tao
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, 071000, Hebei, China.
| | - Jalal Rouhi
- Faculty of Physics, University of Tabriz, Tabriz, 51566, Iran.
| |
Collapse
|
39
|
Shan Y, Yin X, Zhao N, Wang J, Yang S. Comparison of serum tumor markers combined with dual-source CT in the diagnosis of lung cancer. Minerva Med 2023; 114:795-801. [PMID: 33764713 DOI: 10.23736/s0026-4806.21.07124-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND The purpose of this study is to explore the clinical value of serum tumor markers combined with dual-source CT scanning in the diagnosis of lung cancer. METHODS One hundred-two patients with lung cancer (malignant tumor group), 50 patients with benign lesions (benign control group) and 50 healthy patients (normal control group) were selected as the research objects. The levels of serum carcinoembryonic antigen (CEA), cytokeratin 19 fragment antigen 21-1 (CYFRA21-1) and gastrin releasing peptide precursor 31-98 (Pro-GRP31-98) were detected using the electrochemiluminescence and enzyme-linked immunoassay in three groups of people. Simultaneously, Siemens' second-generation dual-source CT (Siemens AG, Munich, Germany) is used to scan the lungs of patients with lung cancer and benign lesions. Retrospective statistical analysis is utilized to explore the clinical value of serum tumor markers combined with dual-source CT examination in the diagnosis of lung cancer. RESULTS The levels of serum CEA, CYFRA21-1 and Pro-GRP31-98 in lung cancer patients were significantly higher than those in the benign control group and normal control group, and the difference was statistically significant (P<0.01). There was no statistical difference between the benign control group and the normal control group (P>0.05). The levels of serum CEA, CYFRA21-1, Pro-GRP31-98 had no significant correlation with the age, sex, and tumor site of lung cancer patients (P>0.05). However, the levels of serum CEA, CYFRA21-1, Pro-GRP31-98 have an obvious correlation with tumor size, clinical stage, histological type, combined pleural effusion, recurrence and metastasis after treatment (P<0.05). CT scans of 102 lung cancer patients showed 35 cases of central type, 67 cases of peripheral type, 71 cases with tumor diameter >5cm, 31 cases of ≤5cm and 67 cases of lymph node metastasis. The sensitivities of CEA, CYFRA21-1, Pro-GRP31-98, and dual-source CT in the diagnosis of lung cancer were 64.70%, 60.11%, 56.86% and 77.45%, and the accuracy was 75.00%, 72.37%, 69.74% and 82.89%, respectively. Compared with a single examination, the sensitivity and accuracy of combined examination for the diagnosis of lung cancer were significantly improved, which were 95.10% and 92.76%, respectively. CONCLUSIONS Joint examinations can effectively improve the rate of lung cancer diagnosis.
Collapse
Affiliation(s)
- Yuqing Shan
- Department of Imaging, People's Hospital of Rizhao, Rizhao, China
| | - Xiangyuan Yin
- Department of Imaging, People's Hospital of Rizhao, Rizhao, China
| | - Na Zhao
- Department of Imaging, People's Hospital of Rizhao, Rizhao, China
| | - Jie Wang
- Department of Imaging, Lanshan People's Hospital of Rizhao, Rizhao, China
| | - Shouxiang Yang
- Department of Imaging, People's Hospital of Rizhao, Rizhao, China -
| |
Collapse
|
40
|
Xie Z, Zhang Q, Wang X, Chen Y, Deng Y, Lin H, Wu J, Huang X, Xu Z, Chi P. Development and validation of a novel radiomics nomogram for prediction of early recurrence in colorectal cancer. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2023; 49:107118. [PMID: 37844471 DOI: 10.1016/j.ejso.2023.107118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/25/2023] [Accepted: 10/10/2023] [Indexed: 10/18/2023]
Abstract
BACKGROUND Early recurrence (ER) is a significant concern following curative resection of advanced colorectal cancer (CRC) and is linked to poor long-term survival. Reliable prediction of ER is challenging, necessitating the development of a novel radiomics-based nomogram for CRC patients. METHODS We enrolled 405 patients, with 298 in the training set and 107 in the external test set. Radiomic features were extracted from preoperative venous-phase computed tomography (CT) images. A radiomics signature was created using univariate logistic regression analyses and the least absolute shrinkage and selection operator algorithm. Clinical factors were integrated into the analyses to develop a comprehensive predictive tool in a multivariate logistic regression model, resulting in a radiomics nomogram. Subsequently, the calibration, discrimination, and clinical usefulness of the nomogram were evaluated. RESULTS The radiomics signature, consisting of four selected CT features, was significantly associated with ER in both the training and test datasets (P < 0.05). Independent predictors of ER included TNM stage, carcinoembryonic antigen level and differentiation grade were identified. The radiomics nomogram, incorporating all these predictors, exhibited good predictive ability in both the training set with an area under the curve (AUC) of 0.82 (95 % confidence interval (CI), 0.74-0.90) and the test set with an AUC of 0.85 (95 % CI, 0.72-0.99), surpassing the performance of any single candidate factor alone. Furthermore, additional analysis demonstrated that the nomogram was clinically useful. CONCLUSIONS We have developed a radiomics-based nomogram that effectively predicts early recurrence in CRC patients, enhancing the potential for timely intervention and improved outcomes.
Collapse
Affiliation(s)
- Zhongdong Xie
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Qingwei Zhang
- Division of Gastroenterology and Hepatology, Key Laboratory of Digestive Diseases, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| | - Xiaojie Wang
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Yongchun Chen
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yu Deng
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Hanbin Lin
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Jiashu Wu
- Department of Science and Technology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xinming Huang
- Department of Radiology, Union Hospital, Fujian Medical University, Fuzhou, China.
| | - Zongbin Xu
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, China.
| | - Pan Chi
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
41
|
Florescu DN, Boldeanu MV, Șerban RE, Florescu LM, Serbanescu MS, Ionescu M, Streba L, Constantin C, Vere CC. Correlation of the Pro-Inflammatory Cytokines IL-1β, IL-6, and TNF-α, Inflammatory Markers, and Tumor Markers with the Diagnosis and Prognosis of Colorectal Cancer. Life (Basel) 2023; 13:2261. [PMID: 38137862 PMCID: PMC10744550 DOI: 10.3390/life13122261] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/19/2023] [Accepted: 11/25/2023] [Indexed: 12/24/2023] Open
Abstract
Colorectal cancer (CRC) remains one of the most important global health problems, being in the top 3 neoplasms in terms of the number of cases worldwide. Although CRC develops predominantly from the adenoma-adenocarcinoma sequence through APC gene mutations, in recent years, studies have demonstrated the role of chronic inflammation in this neoplasia pathogenesis. Cytokines are important components of chronic inflammation, being some of the host regulators in response to inflammation. The pro-inflammatory cytokines IL-1β, IL-6, and TNF-α are involved in tumor cell proliferation, angiogenesis, and metastasis and seem to strengthen each other's mode of action, these being stimulated by the same mediators. In our study, we collected data on 68 patients with CRC and 20 healthy patients from the Gastroenterology Department of Craiova County Emergency Clinical Hospital, who were assessed between January 2022 and February 2023. The main purpose of this study was to investigate the correlation between increased plasma levels of the cytokines and the extent of the tumor, lymph nodes, and metastasis-(TNM stage), as well as the patients' prognoses. We also compared the plasma levels of cytokines and acute inflammatory markers, namely, the erythrocyte sedimentation rate (ESR), c-reactive protein (CRP), and fibrinogen, along with the tumor markers, carcinoembryonic antigen (CEA) and carbohydrate antigen 19.9 (CA 19.9), in CRC patients. We showed that all the pro-inflammatory cytokines studied had higher levels in patients with CRC in comparison with the control group. We also showed that the acute inflammatory markers of erythrocyte sedimentation rate, C-reactive protein, and fibrinogen, and the tumor markers of CEA and CA 19.9 can be useful in diagnosis and prognosis in patients with CRC. Considering the association between pro-inflammatory cytokines and CRC, the development of new targeted therapies against IL-1β, IL-6, and TNF-α can improve patient care and the CRC survival rate.
Collapse
Affiliation(s)
- Dan Nicolae Florescu
- Department of Gastroenterology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (D.N.F.); (C.C.V.)
- Research Center of Gastroenterology and Hepatology, University of Medicine and Pharmacy of Craiova, 200638 Craiova, Romania
| | - Mihail-Virgil Boldeanu
- Department of Immunology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | - Robert-Emmanuel Șerban
- Department of Gastroenterology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (D.N.F.); (C.C.V.)
- Research Center of Gastroenterology and Hepatology, University of Medicine and Pharmacy of Craiova, 200638 Craiova, Romania
| | - Lucian Mihai Florescu
- Department of Radiology and Medical Imaging, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (L.M.F.); (C.C.)
| | - Mircea-Sebastian Serbanescu
- Department of Medical Informatics and Biostatistics, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | - Mihaela Ionescu
- Department of Medical Informatics and Biostatistics, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | - Liliana Streba
- Department of Oncology, University of Medicine and Pharmacy Craiova, 2 Petru Rares Str., 200349 Craiova, Romania;
| | - Cristian Constantin
- Department of Radiology and Medical Imaging, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (L.M.F.); (C.C.)
| | - Cristin Constantin Vere
- Department of Gastroenterology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (D.N.F.); (C.C.V.)
- Research Center of Gastroenterology and Hepatology, University of Medicine and Pharmacy of Craiova, 200638 Craiova, Romania
| |
Collapse
|
42
|
Kumarasamy G, Mohd Salim NH, Mohd Afandi NS, Hazlami Habib MA, Mat Amin ND, Ismail MN, Musa M. Glycoproteomics-based liquid biopsy: translational outlook for colorectal cancer clinical management in Southeast Asia. Future Oncol 2023; 19:2313-2332. [PMID: 37937446 DOI: 10.2217/fon-2023-0704] [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: 11/09/2023] Open
Abstract
Colorectal cancer (CRC) signifies a significant healthcare challenge in Southeast Asia. Despite advancements in screening approaches and treatment modalities, significant medical gaps remain, ranging from prevention and early diagnosis to determining targeted therapy and establishing personalized approaches to managing CRC. There is a need to expand more validated biomarkers in clinical practice. An advanced technique incorporating high-throughput mass spectrometry as a liquid biopsy to unravel a repertoire of glycoproteins and glycans would potentially drive the development of clinical tools for CRC screening, diagnosis and monitoring, and it can be further adapted to the existing standard-of-care procedure. Therefore this review offers a perspective on glycoproteomics-driven liquid biopsy and its potential integration into the clinical care of CRC in the southeast Asia region.
Collapse
Affiliation(s)
- Gaayathri Kumarasamy
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Pulau Pinang, 11800, Malaysia
| | - Nurul Hakimah Mohd Salim
- Department of Pathology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, 16150, Malaysia
| | - Nur Syafiqah Mohd Afandi
- Analytical Biochemistry Research Centre, Universiti Sains Malaysia, Bayan Lepas, Pulau Pinang, 11900, Malaysia
| | - Mohd Afiq Hazlami Habib
- Analytical Biochemistry Research Centre, Universiti Sains Malaysia, Bayan Lepas, Pulau Pinang, 11900, Malaysia
| | - Nor Datiakma Mat Amin
- Analytical Biochemistry Research Centre, Universiti Sains Malaysia, Bayan Lepas, Pulau Pinang, 11900, Malaysia
- Nature Products Division, Forest Research Institute Malaysia, Kepong, Selangor, 52109, Malaysia
| | - Mohd Nazri Ismail
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Pulau Pinang, 11800, Malaysia
- Analytical Biochemistry Research Centre, Universiti Sains Malaysia, Bayan Lepas, Pulau Pinang, 11900, Malaysia
| | - Marahaini Musa
- Human Genome Centre, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, 16150, Malaysia
| |
Collapse
|
43
|
Kobayashi S, Hiwasa T, Kitamura K, Kano M, Hoshino T, Hirano S, Hashimoto M, Seimiya M, Shimada H, Nomura F, Matsubara H, Matsushita K. Combinational antibody detection approach increases the clinical validity of colorectal cancer screening. J Clin Lab Anal 2023; 37:e24978. [PMID: 37964630 PMCID: PMC10749486 DOI: 10.1002/jcla.24978] [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: 05/23/2023] [Revised: 09/27/2023] [Accepted: 10/15/2023] [Indexed: 11/16/2023] Open
Abstract
BACKGROUND At different stages of the disease, biomarkers can help to determine disease progression and recurrence and provide a personalized indicator of therapeutic effectiveness. The serological identification of antigens by recombinant cDNA expression cloning (SEREX) has identified five SEREX antigens. RESULTS Compared with healthy donors, anti-FIRΔexon2 and anti-SOHLH antibodies (Abs) in the sera of patients with colorectal cancer (CRC) were markedly higher. Furthermore, no correlation was noted between five SEREX antigens and the three tumor markers (CEA, CA19-9, and anti-p53 Abs), indicating that anti-FIRΔexon2 Abs are an independent candidate marker for patients with CRC. Generally, the levels of anti-FIRΔexon2 Abs combined with clinically available tumor markers were determined to be significantly higher compared with CEA, CA19-9. Moreover, in early-stage CRC, the levels of anti-FIRΔexon2 Abs combined with existing tumor markers were higher than those of CEA, CA19-9. CONCLUSION Due to the highly heterogeneous nature of CRC, a single tumor marker is unlikely to become a standalone diagnostic test due to its commonly insufficient sensitivity and/or specificity. Using a combination antibody detection approach of tumor markers for CRC diagnosis has the potential to be an effective approach. Therefore, the use of serum protein biomarker candidates holds promise for the development of inexpensive, noninvasive, and inexpensive tests for the detection of CRC.
Collapse
Affiliation(s)
- Sohei Kobayashi
- Department of Laboratory Medicine & Division of Clinical GeneticsChiba University HospitalChibaJapan
- Department of Medical Technology & Sciences, School of Health Sciences at NaritaInternational University of Health and WelfareChibaJapan
| | - Takaki Hiwasa
- Department of Neurological Surgery, Graduate School of MedicineChiba UniversityChibaJapan
| | - Kouichi Kitamura
- Department of Laboratory Medicine & Division of Clinical GeneticsChiba University HospitalChibaJapan
| | - Masayuki Kano
- Department of Frontier Surgery, Graduate School of MedicineChiba UniversityChibaJapan
| | - Tyuji Hoshino
- Department of Physical Chemistry, Graduate School of Pharmaceutical SciencesChiba UniversityChibaJapan
| | - Sho Hirano
- Department of Medical Technology & Sciences, School of Health Sciences at NaritaInternational University of Health and WelfareChibaJapan
| | - Mayuko Hashimoto
- Department of Medical Technology & Sciences, School of Health Sciences at NaritaInternational University of Health and WelfareChibaJapan
| | - Masanori Seimiya
- Department of Medical Technology & Sciences, School of Health Sciences at NaritaInternational University of Health and WelfareChibaJapan
| | - Hideaki Shimada
- Department of Gastroenterological Surgery, Graduate School of MedicineToho UniversityTokyoJapan
| | - Fumio Nomura
- Department of Laboratory Medicine & Division of Clinical GeneticsChiba University HospitalChibaJapan
| | - Hisahiro Matsubara
- Department of Frontier Surgery, Graduate School of MedicineChiba UniversityChibaJapan
| | - Kazuyuki Matsushita
- Department of Laboratory Medicine & Division of Clinical GeneticsChiba University HospitalChibaJapan
| |
Collapse
|
44
|
Mohammed OA. From strings to signals: Unraveling the impact of miRNAs on diagnosis, and progression of colorectal cancer. Pathol Res Pract 2023; 251:154857. [PMID: 37804545 DOI: 10.1016/j.prp.2023.154857] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 09/21/2023] [Accepted: 10/03/2023] [Indexed: 10/09/2023]
Abstract
Colorectal cancer (CRC) stands as the third most prevalent ailment globally and represents the primary cause of mortality associated with cancer. Significant advancements have been made in the clinical management of patients with CRC, encompassing the development of more streamlined methodologies and a diverse array of biomarkers utilized for prognostic, diagnostic, and predictive objectives. MicroRNAs (miRNAs, miRs) play a key role in the development of CRC by modulating the expression of their target genes, which govern a number of metabolic and cellular processes. They are related to malignant traits such as enhanced invasive and proliferative capacity, evasion of apoptosis, cell cycle aberration, and promotion of angiogenesis through dysregulation in their function. This review's objectives were to examine miRNA biogenesis, provide an updated list of oncogenic and tumor suppressor miRNAs, and discuss the likely causes of miRNA dysregulation in CRC. Additionally, we discuss the diagnostic and predictive functions of miRNAs in CRC and summarize their biological significance and clinical potential.
Collapse
Affiliation(s)
- Osama A Mohammed
- Department of Clinical Pharmacology, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia.
| |
Collapse
|
45
|
Cheng Y, Xue G, Lan L, Xu H, Cheng R, Song Q, Li C, Zhang J, Huang G, Shen Z, Xue C. Construction of a 3D rigidified DNA nanodevice for anti-interference and reinforced biosensing by turning nuclease into a catalyst. Biosens Bioelectron 2023; 237:115501. [PMID: 37392492 DOI: 10.1016/j.bios.2023.115501] [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: 04/06/2023] [Revised: 06/13/2023] [Accepted: 06/25/2023] [Indexed: 07/03/2023]
Abstract
The practical application of DNA biosensors is impeded by numerous limitations in complicated physiological environments, particularly the susceptibility of common DNA components to nuclease degradation, which has been recognized as a major barrier in DNA nanotechnology. In contrast, the present study presents an anti-interference and reinforced biosensing strategy based on a 3D DNA-rigidified nanodevice (3D RND) by converting a nuclease into a catalyst. 3D RND is a well-known tetrahedral DNA scaffold containing four faces, four vertices, and six double-stranded edges. The scaffold was rebuilt to serve as a biosensor by embedding a recognition region and two palindromic tails on one edge. In the absence of a target, the rigidified nanodevice exhibited enhanced nuclease resistance, resulting in a low false-positive signal. 3D RNDs have been proven to be compatible with 10% serum for at least 8 h. Once exposed to the target miRNA, the system can be unlocked and converted into common DNAs from a high-defense state, followed by polymerase- and nuclease-co-driven conformational downgrading to achieve amplified and reinforced biosensing. The signal response can be improved by approximately 700% within 2 h at room temperature, and the limit of detection (LOD) is approximately 10-fold lower under biomimetic conditions. The final application to serum miRNA-mediated clinical diagnosis of colorectal cancer (CRC) patients revealed that 3D RND is a reliable approach to collecting clinical information for differentiating patients from healthy individuals. This study provides novel insights into the development of anti-interference and reinforced DNA biosensors.
Collapse
Affiliation(s)
- Yinghao Cheng
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, 325000, PR China; College of Materials and Chemical Engineering, Minjiang University, Fuzhou, Fujian, 350108, PR China
| | - Guohui Xue
- Department of Clinical Laboratory, Jiujiang NO.1 People's Hospital, Jiujiang, Jiangxi, 332000, PR China
| | - Linwen Lan
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, 325000, PR China
| | - Huo Xu
- College of Materials and Chemical Engineering, Minjiang University, Fuzhou, Fujian, 350108, PR China
| | - Ruize Cheng
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, 325000, PR China
| | - Qiufeng Song
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, 325000, PR China
| | - Chan Li
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, 325000, PR China
| | - Jing Zhang
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, 325000, PR China
| | - Guoqiao Huang
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, 325000, PR China
| | - Zhifa Shen
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, 325000, PR China.
| | - Chang Xue
- Wenzhou Key Laboratory of Cancer Pathogenesis and Translation, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, 325000, PR China.
| |
Collapse
|
46
|
Williams L, Li L, Yazaki PJ, Wong P, Miller A, Hong T, Poku EK, Bhattacharya S, Shively JE, Kujawski M. Generation of IL-2-Fc-antibody conjugates by click chemistry. Biotechnol J 2023; 18:e2300115. [PMID: 37300381 DOI: 10.1002/biot.202300115] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/10/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023]
Abstract
BACKGROUND Immunocytokines (ICKs) are antibody directed cytokines produced by genetic fusion of an antibody to a cytokine. METHODS We now show that antibodies conjugated by click chemistry to interleukin-2 (IL-2)-Fc form fully active conjugates, and in one example, equivalent activity to a genetically produced ICK. RESULTS An IL-2-Fc fusion protein was optimized for click chemistry at hinge cysteines using protein stabilizing IL-2 mutations at Lys35 and Cys125 and Fc hinge mutations at Cys142 and Cys148. The IL-2-Fc fusion protein with K35E and C125S mutations with 3 intact hinge cysteines, designated as IL-2-Fc Par, was selected based on its minimal tendency to aggregate. IL-2-Fc-antibody clicked conjugates retained high IL-2 activity and bound target antigens comparable to parent antibodies. An IL-2-Fc-anti-CEA click conjugate showed comparable anti-tumor activity to an anti-CEA-IL-2 ICK in immunocompetent CEA transgenic mice bearing CEA positive orthotopic breast tumors. Significant increases in IFNγ+ /CD8+ and decreases in FoxP3+ /CD4+ T-cells were found for the clicked conjugate and ICK therapies, suggesting a common mechanism of tumor reduction. CONCLUSION The production of antibody targeted IL-2 therapy via a click chemistry approach is feasible with comparable activity to genetically produced ICKs with the added advantage of multiplexing with other monoclonal antibodies.
Collapse
Affiliation(s)
- Lindsay Williams
- Department of Immunology and Theranostics, Riggs Diabetes, Metabolism, and Research Institute, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Lin Li
- Department of Immunology and Theranostics, Riggs Diabetes, Metabolism, and Research Institute, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Paul J Yazaki
- Department of Immunology and Theranostics, Riggs Diabetes, Metabolism, and Research Institute, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Patty Wong
- Department of Immunology and Theranostics, Riggs Diabetes, Metabolism, and Research Institute, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Aaron Miller
- Department of Immunology and Theranostics, Riggs Diabetes, Metabolism, and Research Institute, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Teresa Hong
- Department of Immunology and Theranostics, Riggs Diabetes, Metabolism, and Research Institute, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Erasmus K Poku
- Radiopharmacy, City of Hope, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Supriyo Bhattacharya
- Department of Computational and Quantitative Medicine, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - John E Shively
- Department of Immunology and Theranostics, Riggs Diabetes, Metabolism, and Research Institute, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Maciej Kujawski
- Department of Immunology and Theranostics, Riggs Diabetes, Metabolism, and Research Institute, Beckman Research Institute of City of Hope, Duarte, California, USA
| |
Collapse
|
47
|
Mohajershojai T, Spangler D, Chopra S, Frejd FY, Yazaki PJ, Nestor M. Enhanced Therapeutic Effects of 177Lu-DOTA-M5A in Combination with Heat Shock Protein 90 Inhibitor Onalespib in Colorectal Cancer Xenografts. Cancers (Basel) 2023; 15:4239. [PMID: 37686514 PMCID: PMC10486833 DOI: 10.3390/cancers15174239] [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: 07/21/2023] [Revised: 08/16/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023] Open
Abstract
Carcinoembryonic antigen (CEA) has emerged as an attractive target for theranostic applications in colorectal cancers (CRCs). In the present study, the humanized anti-CEA antibody hT84.66-M5A (M5A) was labeled with 177Lu for potential CRC therapy. Moreover, the novel combination of 177Lu-DOTA-M5A with the heat shock protein 90 inhibitor onalespib, suggested to mediate radiosensitizing properties, was assessed in vivo for the first time. M5A antibody uptake and therapeutic effects, alone or in combination with onalespib, were assessed in human CRC xenografts and visualized using SPECT/CT imaging. Although both 177Lu-DOTA-M5A and onalespib monotherapies effectively reduced tumor growth rates, the combination therapy demonstrated the most substantial impact, achieving a fourfold reduction in tumor growth compared to the control group. Median survival increased by 33% compared to 177Lu-DOTA-M5A alone, and tripled compared to control and onalespib groups. Importantly, combination therapy yielded comparable or superior effects to the double dose of 177Lu-DOTA-M5A monotherapy. 177Lu-DOTA-M5A increased apoptotic cell levels, indicating its potential to induce tumor cell death. These findings show promise for 177Lu-DOTA-M5A as a CRC therapeutic agent, and its combination with onalespib could significantly enhance treatment efficacy. Further in vivo studies are warranted to validate these findings fully and explore the treatment's potential for clinical use.
Collapse
Affiliation(s)
- Tabassom Mohajershojai
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.M.); (S.C.); (F.Y.F.)
| | - Douglas Spangler
- Department of Public Health and Caring Sciences, Uppsala University, 751 22 Uppsala, Sweden;
| | - Saloni Chopra
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.M.); (S.C.); (F.Y.F.)
| | - Fredrik Y. Frejd
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.M.); (S.C.); (F.Y.F.)
| | - Paul J. Yazaki
- Department of Immunology & Theranostics, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA;
| | - Marika Nestor
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; (T.M.); (S.C.); (F.Y.F.)
| |
Collapse
|
48
|
Silva MLS. Capitalizing glycomic changes for improved biomarker-based cancer diagnostics. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:366-395. [PMID: 37455827 PMCID: PMC10344901 DOI: 10.37349/etat.2023.00140] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 02/24/2023] [Indexed: 07/18/2023] Open
Abstract
Cancer serum biomarkers are valuable or even indispensable for cancer diagnostics and/or monitoring and, currently, many cancer serum markers are routinely used in the clinic. Most of those markers are glycoproteins, carrying cancer-specific glycan structures that can provide extra-information for cancer monitoring. Nonetheless, in the majority of cases, this differential feature is not exploited and the corresponding analytical assays detect only the protein amount, disregarding the analysis of the aberrant glycoform. Two exceptions to this trend are the biomarkers α-fetoprotein (AFP) and cancer antigen 19-9 (CA19-9), which are clinically monitored for their cancer-related glycan changes, and only the AFP assay includes quantification of both the protein amount and the altered glycoform. This narrative review demonstrates, through several examples, the advantages of the combined quantification of protein cancer biomarkers and the respective glycoform analysis, which enable to yield the maximum information and overcome the weaknesses of each individual analysis. This strategy allows to achieve higher sensitivity and specificity in the detection of cancer, enhancing the diagnostic power of biomarker-based cancer detection tests.
Collapse
Affiliation(s)
- Maria Luísa S. Silva
- Unidade de Aprendizagem ao Longo da Vida, Universidade Aberta, 1269-001 Lisboa, Portugal
| |
Collapse
|
49
|
Park S, Cho E, Chueng STD, Yoon JS, Lee T, Lee JH. Aptameric Fluorescent Biosensors for Liver Cancer Diagnosis. BIOSENSORS 2023; 13:617. [PMID: 37366982 DOI: 10.3390/bios13060617] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/01/2023] [Accepted: 06/02/2023] [Indexed: 06/28/2023]
Abstract
Liver cancer is a prevalent global health concern with a poor 5-year survival rate upon diagnosis. Current diagnostic techniques using the combination of ultrasound, CT scans, MRI, and biopsy have the limitation of detecting detectable liver cancer when the tumor has already progressed to a certain size, often leading to late-stage diagnoses and grim clinical treatment outcomes. To this end, there has been tremendous interest in developing highly sensitive and selective biosensors to analyze related cancer biomarkers in the early stage diagnosis and prescribe appropriate treatment options. Among the various approaches, aptamers are an ideal recognition element as they can specifically bind to target molecules with high affinity. Furthermore, using aptamers, in conjunction with fluorescent moieties, enables the development of highly sensitive biosensors by taking full advantage of structural and functional flexibility. This review will provide a summary and detailed discussion on recent aptamer-based fluorescence biosensors for liver cancer diagnosis. Specifically, the review focuses on two promising detection strategies: (i) Förster resonance energy transfer (FRET) and (ii) metal-enhanced fluorescence for detecting and characterizing protein and miRNA cancer biomarkers.
Collapse
Affiliation(s)
- Seonga Park
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
| | - Euni Cho
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
- Department of Information Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
| | | | - June-Sun Yoon
- Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Taek Lee
- Department of Chemical Engineering, Kwangwoon University, Seoul 01897, Republic of Korea
| | - Jin-Ho Lee
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
- Department of Information Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| |
Collapse
|
50
|
Anzinger I, Nagel D, De Toni EN, Ofner A, Philipp AB, Holdt LM, Teupser D, Kolligs FT, Herbst A. Cell-free circulating ALU repeats in serum have a prognostic value for colorectal cancer patients. Cancer Biomark 2023:CBM210536. [PMID: 37302022 DOI: 10.3233/cbm-210536] [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: 06/12/2023]
Abstract
BACKGROUND Carcinoembryonic antigen (CEA) is the only established serum biomarker for colorectal cancer (CRC). To facilitate therapy decisions and improve the overall survival of CRC patients, prognostic biomarkers are required. OBJECTIVE We studied the prognostic value of five different cell free circulating DNA (fcDNA) fragments. The potential markers were ALU115, ALU247, LINE1-79, LINE1-300 and ND1-mt. METHODS The copy numbers of the DNA fragments were measured in the peripheral blood serum of 268 CRC patients using qPCR, the results were compared to common and previously described markers. RESULTS We found that ALU115 and ALU247 fcDNA levels correlate significantly with several clinicopathological parameters. An increased amount of ALU115 and ALU247 fcDNA fragments coincides with methylation of HPP1 (P< 0.001; P< 0.01), which proved to be a prognostic marker itself in former studies and also with increased CEA level (P< 0.001). ALU115 and ALU247 can define patients with poor survival in UICC stage IV (Alu115: HR = 2.9; 95% Cl 1.8-4.8, P< 0.001; Alu247: HR = 2.2; 95% Cl 1.3-3.6; P= 0.001). Combining ALU115 and HPP1, the prognostic value in UICC stage IV is highly significant (P< 0.001). CONCLUSIONS This study shows that an increased level of ALU fcDNA is an independent prognostic biomarker for advanced colorectal cancer disease.
Collapse
Affiliation(s)
- Isabel Anzinger
- Department of Urology, St. Elisabeth Hospital, Straubing, Germany
| | - Dorothea Nagel
- Institute of Laboratory Medicine, Faculty of Medicine, Ludwig Maximilians University, Munich, Germany
| | - Enrico N De Toni
- Medical Department 2, Faculty of Medicine, Ludwig Maximilians University, Munich, Germany
| | - Andrea Ofner
- Medical Department 2, Faculty of Medicine, Ludwig Maximilians University, Munich, Germany
| | - Alexander B Philipp
- Medical Department 2, Faculty of Medicine, Ludwig Maximilians University, Munich, Germany
| | - Lesca M Holdt
- Institute of Laboratory Medicine, Faculty of Medicine, Ludwig Maximilians University, Munich, Germany
| | - Daniel Teupser
- Institute of Laboratory Medicine, Faculty of Medicine, Ludwig Maximilians University, Munich, Germany
| | | | - Andreas Herbst
- Medical Department 2, Faculty of Medicine, Ludwig Maximilians University, Munich, Germany
| |
Collapse
|