1
|
Li J, Cao X, Zhang L, Liu A, Liu S, Chen F, Li Y, Ma H, Sun W, Ouyang S, Dai L, Liu J. Anti-FDX1 Autoantibody as a Potential Biomarker for Non-Small Cell Lung Cancer Detection. Cancer Epidemiol Biomarkers Prev 2025; 34:439-447. [PMID: 39699293 DOI: 10.1158/1055-9965.epi-24-1096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 10/07/2024] [Accepted: 12/17/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Autoantibodies can be readily identified prior to biopsy and may serve as valuable biomarkers for cancer detection. Ferredoxin 1 (FDX1) is a key regulator in the process of cuproptosis and affects the prognosis of lung cancer. In this study, we investigated whether the anti-FDX1 autoantibody could serve as a novel biomarker for the detection of non-small cell lung cancer (NSCLC). METHODS A total of 1,155 plasma samples were divided into the verification and validation groups. The expression levels of the anti-FDX1 autoantibody in 414 patients with NSCLC, 327 patients with benign pulmonary nodules (BPN), and 414 normal controls (NC) were detected using ELISA. Western blotting and immunofluorescence analyses were performed to confirm the ELISA results. RESULTS Plasma anti-FDX1 autoantibody levels were significantly higher in patients with NSCLC than in patients with BPN and NCs in the verification and validation groups. The ELISA results were confirmed by Western blotting and immunofluorescence. The anti-FDX1 autoantibody distinguished NSCLC from NC and BPN with an AUC (95% confidence interval) of 0.806 (0.772-0.839) and 0.627 (0.584-0.670), respectively. CONCLUSIONS Our study demonstrated the potential benefits of the anti-FDX1 autoantibody as a novel biomarker for NSCLC detection. IMPACT These findings suggested that the anti-FDX1 autoantibody may facilitate the detection of NSCLC.
Collapse
Affiliation(s)
- Jing Li
- Henan Institute of Medical and Pharmaceutical Sciences & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Xiaobin Cao
- Henan Institute of Medical and Pharmaceutical Sciences & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
- BGI College, Zhengzhou University, Zhengzhou, China
| | - Lulu Zhang
- Henan Institute of Medical and Pharmaceutical Sciences & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Aichen Liu
- Henan Institute of Medical and Pharmaceutical Sciences & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Siyu Liu
- Henan Institute of Medical and Pharmaceutical Sciences & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Fengqi Chen
- Henan Institute of Medical and Pharmaceutical Sciences & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Yutong Li
- Henan Institute of Medical and Pharmaceutical Sciences & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
- BGI College, Zhengzhou University, Zhengzhou, China
| | - Hanke Ma
- Henan Institute of Medical and Pharmaceutical Sciences & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Wenke Sun
- Henan Institute of Medical and Pharmaceutical Sciences & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Songyun Ouyang
- Department of Respiratory and Sleep Medicine in the First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Liping Dai
- Henan Institute of Medical and Pharmaceutical Sciences & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Jingjing Liu
- Henan Institute of Medical and Pharmaceutical Sciences & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
2
|
Yu Z, Zhang H, Li S, Huo Q, Ling H, Chen K, Wang Z. Novel combined tumor autoantibody detection in serological diagnosis of gastric cancer. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2025; 18:23-29. [PMID: 39949595 PMCID: PMC11815391 DOI: 10.62347/xmaw3065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 12/29/2024] [Indexed: 02/16/2025]
Abstract
OBJECTIVE Gastric cancer (GC) is a highly prevalent malignancy, yet its early diagnosis rate is generally low. Therefore, we have established a serum-based combined detection method based on tumor autoantibodies aimed at improving the diagnostic rate of gastric cancer. METHODS Through clinical studies, we selected a series of proteins aberrantly expressed in gastric cancer patients, including RalA, Survivin, NY-ESO-1, p53, Cyclin B1, and Koc, and expressed and purified them using prokaryotic expression and nickel column chromatography. RESULTS The levels of autoantibodies in the serum of gastric cancer patients and healthy individuals were measured using enzyme-linked immunosorbent assay (ELISA), and the diagnostic value of the combined detection of tumor autoantibodies for gastric cancer was evaluated through receiver operating characteristic (ROC) curve analysis. The levels of autoantibodies against RalA, Survivin, NY-ESO-1, p53, and Cyclin B1 in the serum of gastric cancer patients were significantly higher than those in healthy individuals (P < 0.05), while the level of Koc showed no significant difference between the two groups (P > 0.05), suggesting that Koc may not be suitable for serological diagnosis of gastric cancer. ROC analysis of the combined levels of autoantibodies against RalA, Survivin, NY-ESO-1, p53, and Cyclin B1 for gastric cancer diagnosis achieved a sensitivity of 73.68% and specificity of 78.13%, with an AUC value of 0.8767. CONCLUSION The combined tumor autoantibody detection established in this study may have promising potential applications in early screening and diagnosis of gastric cancer.
Collapse
Affiliation(s)
- Zixin Yu
- School of Life Science and Engineering, Southwest University of Science and TechnologyMianyang 621000, Sichuan, China
| | - Hushan Zhang
- Zhaotong Health Vocational CollegeZhaotong 657000, Yunnan, China
| | - Sheng Li
- Sichuan Kairui Huachuang Biotechnology Co., Ltd.Mianyang 621000, Sichuan, China
| | - Qingwen Huo
- Sichuan Kairui Huachuang Biotechnology Co., Ltd.Mianyang 621000, Sichuan, China
| | - Han Ling
- Laboratory Department of Sichuan Cancer HospitalChengdu 610000, Sichuan, China
| | - Ke Chen
- School of Life Science and Engineering, Southwest University of Science and TechnologyMianyang 621000, Sichuan, China
| | - Zhiming Wang
- School of Life Science and Engineering, Southwest University of Science and TechnologyMianyang 621000, Sichuan, China
- Sichuan Kairui Huachuang Biotechnology Co., Ltd.Mianyang 621000, Sichuan, China
| |
Collapse
|
3
|
Radaic A, Kamarajan P, Cho A, Wang S, Hung G, Najarzadegan F, Wong DT, Ton‐That H, Wang C, Kapila YL. Biological biomarkers of oral cancer. Periodontol 2000 2024; 96:250-280. [PMID: 38073011 PMCID: PMC11163022 DOI: 10.1111/prd.12542] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 11/09/2023] [Indexed: 06/12/2024]
Abstract
The oral squamous cell carcinoma (OSCC) 5 year survival rate of 41% has marginally improved in the last few years, with less than a 1% improvement per year from 2005 to 2017, with higher survival rates when detected at early stages. Based on histopathological grading of oral dysplasia, it is estimated that severe dysplasia has a malignant transformation rate of 7%-50%. Despite these numbers, oral dysplasia grading does not reliably predict its clinical behavior. Thus, more accurate markers predicting oral dysplasia progression to cancer would enable better targeting of these lesions for closer follow-up, especially in the early stages of the disease. In this context, molecular biomarkers derived from genetics, proteins, and metabolites play key roles in clinical oncology. These molecular signatures can help predict the likelihood of OSCC development and/or progression and have the potential to detect the disease at an early stage and, support treatment decision-making and predict treatment responsiveness. Also, identifying reliable biomarkers for OSCC detection that can be obtained non-invasively would enhance management of OSCC. This review will discuss biomarkers for OSCC that have emerged from different biological areas, including genomics, transcriptomics, proteomics, metabolomics, immunomics, and microbiomics.
Collapse
Affiliation(s)
- Allan Radaic
- School of DentistryUniversity of California, Los Angeles (UCLA)Los AngelesCaliforniaUSA
| | - Pachiyappan Kamarajan
- School of DentistryUniversity of California, Los Angeles (UCLA)Los AngelesCaliforniaUSA
| | - Alex Cho
- School of DentistryUniversity of California, Los Angeles (UCLA)Los AngelesCaliforniaUSA
| | - Sandy Wang
- School of DentistryUniversity of California, Los Angeles (UCLA)Los AngelesCaliforniaUSA
| | - Guo‐Chin Hung
- School of DentistryUniversity of California, Los Angeles (UCLA)Los AngelesCaliforniaUSA
| | | | - David T. Wong
- School of DentistryUniversity of California, Los Angeles (UCLA)Los AngelesCaliforniaUSA
| | - Hung Ton‐That
- School of DentistryUniversity of California, Los Angeles (UCLA)Los AngelesCaliforniaUSA
| | - Cun‐Yu Wang
- School of DentistryUniversity of California, Los Angeles (UCLA)Los AngelesCaliforniaUSA
| | - Yvonne L. Kapila
- School of DentistryUniversity of California, Los Angeles (UCLA)Los AngelesCaliforniaUSA
| |
Collapse
|
4
|
Kim H, Lee JK, Kim HR, Hong YJ. Enhanced Lung Cancer Detection Using a Combined Ratio of Antigen-Autoantibody Immune Complexes against CYFRA 21-1 and p53. Cancers (Basel) 2024; 16:2661. [PMID: 39123389 PMCID: PMC11312164 DOI: 10.3390/cancers16152661] [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: 06/30/2024] [Revised: 07/20/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
The early detection of lung cancer (LC) improves patient outcomes, but current methods have limitations. Autoantibodies against tumor-associated antigens have potential as early biomarkers. This study evaluated the 9G testTM Cancer/Lung, measuring circulating complexes of two antigen-autoantibody immune complexes (AIC) against their respective free antigens (CYFRA 21-1 and p53) for LC diagnosis. We analyzed 100 LC patients and 119 healthy controls using the 9G testTM Cancer/Lung, quantifying the levels of AICs (CYFRA 21-1-Anti-CYFRA 21-1 autoantibody immune complex (CIC) and p53-Anti-p53 autoantibody immune complex (PIC)), free antigens (CYFRA 21-1 and p53), and ratios of AICs/antigens (LC index). The levels of the CICs and PICs were significantly elevated in LC compared to the controls (p < 0.0062 and p < 0.0026), while free antigens showed no significant difference. The CIC/CYFRA 21-1 and PIC/p53 ratios were also significantly higher in LC (all, p < 0.0001). The LC index, when combining both ratios, exhibited the best diagnostic performance with an area under the curve (AUC) of 0.945, exceeding individual CICs, PICs, and free antigens (AUCs ≤ 0.887). At a cut-off of 3.60, the LC index achieved 81% sensitivity and 95% specificity for LC diagnosis. It detected early-stage (Stage I-II) LC with 87.5% sensitivity, exceeding its performance in advanced stages (72.7%). The LC index showed no significant differences based on age, gender, smoking status (former, current, or never smoker), or pack years smoked. The LC index demonstrates promising potential for early LC diagnosis, exceeding conventional free antigen markers.
Collapse
Affiliation(s)
- Heyjin Kim
- Department of Laboratory Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea; (H.K.); (J.K.L.)
| | - Jin Kyung Lee
- Department of Laboratory Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea; (H.K.); (J.K.L.)
| | - Hye-Ryoun Kim
- Division of Pulmonology, Department of Internal Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea;
| | - Young Jun Hong
- Department of Laboratory Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea; (H.K.); (J.K.L.)
| |
Collapse
|
5
|
Mou X, Peng Z, Yin T, Sun X. Non-endoscopic Screening for Esophageal Squamous Cell Carcinoma: Recent Advances. J Gastrointest Cancer 2024; 55:118-128. [PMID: 37924487 DOI: 10.1007/s12029-023-00980-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2023] [Indexed: 11/06/2023]
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is one of the most common tumors in the gastrointestinal tract, and China has a high incidence area with a high burden on the disease. As early symptoms of ESCC are not obvious, the mortality rate is high, and it is often diagnosed in the intermediate and advanced stages. However, early screening and treatment may reduce morbidity and mortality. METHODS Screening methods are divided into endoscopic and non-endoscopic screening. RESULTS Endoscopic screening cannot be widely used because of its invasive nature and high cost. Currently, non-endoscopic screening consists primarily of tumor biomarkers and cytology, and tumor biomarkers including autoantibodies, circulating tumor cells, circulating tumor DNA, exosomes and serum metabolomics are more likely to be effective. But the efficiency of early diagnosis of esophageal cancer is low and the accuracy of screening needs to be improved. The aim of this study is to summarize advances in non-endoscopic esophageal cancer screening and strategies to provide a scientific basis and research idea for esophageal cancer prevention and control. CONCLUSIONS Non-endoscopic screening is better than endoscopic screening. And the application of tumor biomarkers is much better than other non-endoscopic screening methods.
Collapse
Affiliation(s)
- Xiao Mou
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China.
| | - Zhenglin Peng
- College of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Tao Yin
- College of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
- Department of Pathology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Xingwang Sun
- College of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
- Department of Pathology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
6
|
Milardi G, Lleo A. Tumor-Infiltrating B Lymphocytes: Promising Immunotherapeutic Targets for Primary Liver Cancer Treatment. Cancers (Basel) 2023; 15:2182. [PMID: 37046842 PMCID: PMC10093314 DOI: 10.3390/cancers15072182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/23/2023] [Accepted: 04/03/2023] [Indexed: 04/14/2023] Open
Abstract
Hepatocellular carcinoma and cholangiocarcinoma are the fourth most lethal primary cancers worldwide. Therefore, there is an urgent need for therapeutic strategies, including immune cell targeting therapies. The heterogeneity of liver cancer is partially explained by the characteristics of the tumor microenvironment (TME), where adaptive and innate immune system cells are the main components. Pioneering studies of primary liver cancers revealed that tumor-infiltrating immune cells and their dynamic interaction with cancer cells significantly impacted carcinogenesis, playing an important role in cancer immune evasion and responses to immunotherapy treatment. In particular, B cells may play a prominent role and have a controversial function in the TME. In this work, we highlight the effect of B lymphocytes as tumor infiltrates in relation to primary liver cancers and their potential prognostic value. We also present the key pathways underlying B-cell interactions within the TME, as well as the way that a comprehensive characterization of B-cell biology can be exploited to develop novel immune-based therapeutic approaches.
Collapse
Affiliation(s)
- Giulia Milardi
- Hepatobiliary Immunopathology Labaratory, IRCCS Humanitas Research Hospital, 20089 Milan, Italy
| | - Ana Lleo
- Department of Biomedical Sciences, Humanitas University, 20072 Milan, Italy
- Department of Gastroenterology, Division of Internal Medicine and Hepatology, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| |
Collapse
|
7
|
Montero-Calle A, Garranzo-Asensio M, Torrente-Rodríguez RM, Ruiz-Valdepeñas Montiel V, Poves C, Dziaková J, Sanz R, Díaz del Arco C, Pingarrón JM, Fernández-Aceñero MJ, Campuzano S, Barderas R. p53 and p63 Proteoforms Derived from Alternative Splicing Possess Differential Seroreactivity in Colorectal Cancer with Distinct Diagnostic Ability from the Canonical Proteins. Cancers (Basel) 2023; 15:2102. [PMID: 37046764 PMCID: PMC10092954 DOI: 10.3390/cancers15072102] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer and the second most frequent cause of cancer-related death worldwide. The detection in plasma samples of autoantibodies against specific tumor-associated antigens has been demonstrated to be useful for the early diagnosis of CRC by liquid biopsy. However, new studies related to the humoral immune response in cancer are needed to enable blood-based diagnosis of the disease. Here, our aim was to characterize the humoral immune response associated with the different p53 and p63 proteoforms derived from alternative splicing and previously described as aberrantly expressed in CRC. Thus, here we investigated the diagnostic ability of the twelve p53 proteoforms and the eight p63 proteoforms described to date, and their specific N-terminal and C-terminal end peptides, by means of luminescence HaloTag beads immunoassays. Full-length proteoforms or specific peptides were cloned as HaloTag fusion proteins and their seroreactivity analyzed using plasma from CRC patients at stages I-IV (n = 31), individuals with premalignant lesions (n = 31), and healthy individuals (n = 48). p53γ, Δ40p53β, Δ40p53γ, Δ133p53γ, Δ160p53γ, TAp63α, TAp63δ, ΔNp63α, and ΔNp63δ, together with the specific C-terminal end α and δ p63 peptides, were found to be more seroreactive against plasma from CRC patients and/or individuals with premalignant lesions than from healthy individuals. In addition, ROC (receiver operating characteristic) curves revealed a high diagnostic ability of those p53 and p63 proteoforms to detect CRC and premalignant individuals (AUC higher than 85%). Finally, electrochemical biosensing platforms were employed in POC-like devices to investigate their usefulness for CRC detection using selected p53 and p63 proteoforms. Our results demonstrate not only the potential of these biosensors for the simultaneous analysis of proteoforms' seroreactivity, but also their convenience and versatility for the clinical detection of CRC by liquid biopsy. In conclusion, we here show that p53 and p63 proteoforms possess differential seroreactivity in CRC patients in comparison to controls, distinctive from canonical proteins, which should improve the diagnostic panels for obtaining a blood-based biomarker signature for CRC detection.
Collapse
Affiliation(s)
- Ana Montero-Calle
- Chronic Disease Programme (UFIEC), Instituto de Salud Carlos III, 28220 Madrid, Spain; (A.M.-C.); (M.G.-A.)
| | - María Garranzo-Asensio
- Chronic Disease Programme (UFIEC), Instituto de Salud Carlos III, 28220 Madrid, Spain; (A.M.-C.); (M.G.-A.)
| | - Rebeca M. Torrente-Rodríguez
- Department of Analytical Chemistry, Faculty of Chemistry, Complutense University of Madrid, 28014 Madrid, Spain; (R.M.T.-R.); (V.R.-V.M.); (J.M.P.); (S.C.)
| | - Víctor Ruiz-Valdepeñas Montiel
- Department of Analytical Chemistry, Faculty of Chemistry, Complutense University of Madrid, 28014 Madrid, Spain; (R.M.T.-R.); (V.R.-V.M.); (J.M.P.); (S.C.)
| | - Carmen Poves
- Gastroenterology Unit, Hospital Universitario Clínico San Carlos, 28040 Madrid, Spain;
| | - Jana Dziaková
- Surgical Digestive Department, Hospital Universitario Clínico San Carlos, 28040 Madrid, Spain
| | - Rodrigo Sanz
- Surgical Digestive Department, Hospital Universitario Clínico San Carlos, 28040 Madrid, Spain
| | - Cristina Díaz del Arco
- Surgical Pathology Department, Hospital Universitario Clínico San Carlos, 28040 Madrid, Spain (M.J.F.-A.)
| | - José Manuel Pingarrón
- Department of Analytical Chemistry, Faculty of Chemistry, Complutense University of Madrid, 28014 Madrid, Spain; (R.M.T.-R.); (V.R.-V.M.); (J.M.P.); (S.C.)
| | | | - Susana Campuzano
- Department of Analytical Chemistry, Faculty of Chemistry, Complutense University of Madrid, 28014 Madrid, Spain; (R.M.T.-R.); (V.R.-V.M.); (J.M.P.); (S.C.)
| | - Rodrigo Barderas
- Chronic Disease Programme (UFIEC), Instituto de Salud Carlos III, 28220 Madrid, Spain; (A.M.-C.); (M.G.-A.)
| |
Collapse
|
8
|
Zhuang L, Huang C, Ning Z, Yang L, Zou W, Wang P, Cheng CS, Meng Z. Circulating tumor-associated autoantibodies as novel diagnostic biomarkers in pancreatic adenocarcinoma. Int J Cancer 2023; 152:1013-1024. [PMID: 36274627 DOI: 10.1002/ijc.34334] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 10/03/2022] [Accepted: 10/10/2022] [Indexed: 01/06/2023]
Abstract
To develop a superior diagnostic approach for pancreatic adenocarcinoma (PAAC), the present study prospectively included 338 PAAC patients, 294 normal healthy volunteers (NHV), 122 chronic pancreatitis (CP) patients and 100 patients with non-PAAC malignancies. In the identification phase, HuProt Human Proteome Microarray, comprising 21 065 proteins, was used to identify serum tumor-associated autoantibodies (TAAbs) candidates differentiating PAAC (n = 30) from NHV (n = 30). A PAAC-focused array containing 165 differentially expressed TAAbs identified was subsequently adopted in the validation phase (n = 712) for specificity and sensitivities. The multivariate TAAbs signature for differentiation PAAC from controls (NHV + CP) identified five candidates, namely the IgG-type TAAbs against CLDN17, KCNN3, SLAMF7, SLC22A11 and OR51F2. Multivariate logistic performance model of y = (22.893 × CA19-9 + 0.68 × CLDN17 - 4.012) showed a significant better diagnostic accuracy than that of CA19-9 and CLDN17 in differentiating PAAC from controls (NHV + CP) (AUC = 0.97, 0.92 and 0.82, respectively, P-value < .0001). We further tested the autoantigen level of CLDN17 by ELISA in 82 sera samples from PAAC (n = 42), CP (n = 24) and NHV (n = 16). Similarly, the model showed superior diagnostic performance than that of CA19-9 and CLDN17 (AUC = 0.93, 0.83 and 0.81, respectively, P-value < .0001) in differentiating PAAC from controls. In conclusion, our study is the first to characterize the circulating TAAbs signatures in PAAC. The results showed that CLDN17 combined with CA19-9 provided potentially clinical value and may serve as noninvasive novel biomarkers for PAAC diagnosis.
Collapse
Affiliation(s)
- Liping Zhuang
- Minimally Invasive Therapy Center, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Integrative Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Changjing Huang
- Minimally Invasive Therapy Center, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Integrative Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhouyu Ning
- Minimally Invasive Therapy Center, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Integrative Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lina Yang
- Minimally Invasive Therapy Center, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Integrative Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Shandong Province, China
| | - Wenbin Zou
- Department of Gastroenterology, Digestive Endoscopy Center, Changhai Hospital, the Second Military Medical University, Shanghai, China
| | - Peng Wang
- Minimally Invasive Therapy Center, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Integrative Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chien-Shan Cheng
- Minimally Invasive Therapy Center, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Integrative Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhiqiang Meng
- Minimally Invasive Therapy Center, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Integrative Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
9
|
Chen H, Sun G, Han Z, Wang H, Li J, Ye H, Song C, Zhang J, Wang P. Anti-CXCL8 Autoantibody: A Potential Diagnostic Biomarker for Esophageal Squamous Cell Carcinoma. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58101480. [PMID: 36295640 PMCID: PMC9607113 DOI: 10.3390/medicina58101480] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 09/15/2022] [Accepted: 10/13/2022] [Indexed: 11/07/2022]
Abstract
Background and Objectives: Esophageal squamous cell carcinoma (ESCC) is one of the most common malignancies. Anti-tumor associated antigen autoantibodies (TAAbs) can be used as biomarkers for tumor detection. The aim of this study was to identify a reliable TAAb as the diagnostic marker for ESCC. Materials and Methods: The Cancer Genome Atlas (TCGA) database was used to screen candidate genes. The mRNA expression of the key gene was then verified by micro array dataset GSE44021 from the Gene Expression Omnibus (GEO) database and the diag nostic value of the corresponding autoantibody to the key gene in ESCC was detected by enzyme-linked im muno sorbent assay (ELISA). Results: CXCL8 was identified as the key gene. The dataset GSE44021 showed that CXCL8 mRNA expression was prominently over-expressed in ESCC tissues compared with normal tissues. ELISA results showed that the level of anti-CXCL8 autoantibody in ESCC patients was significantly higher than in normal controls and the receiver operating char ac teristic (ROC) curve indicated that anti-CXCL8 autoantibody could discriminate ESCC patients from normal controls, with the area under the ROC curve (AUC) for the verification cohort, and the validation cohort were 0.713 and 0.751, respectively. Conclusions: Our study illustrated that anti-CXCL8 autoantibody had good diagnostic value, and may become a candidate biomarker for ESCC.
Collapse
Affiliation(s)
- Huili Chen
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, China
| | - Guiying Sun
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, China
| | - Zhuo Han
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, China
| | - Huimin Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, China
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, China
| | - Jiaxin Li
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, China
| | - Hua Ye
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, China
| | - Chunhua Song
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, China
| | - Jianying Zhang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, China
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, China
| | - Peng Wang
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450052, China
- Correspondence: ; Tel.: +86-0371-67781453
| |
Collapse
|
10
|
Clases D, Gonzalez de Vega R. Facets of ICP-MS and their potential in the medical sciences-Part 2: nanomedicine, immunochemistry, mass cytometry, and bioassays. Anal Bioanal Chem 2022; 414:7363-7386. [PMID: 36042038 PMCID: PMC9427439 DOI: 10.1007/s00216-022-04260-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/26/2022] [Accepted: 07/29/2022] [Indexed: 11/30/2022]
Abstract
Inductively coupled-plasma mass spectrometry (ICP-MS) has transformed our knowledge on the role of trace and major elements in biology and has emerged as the most versatile technique in elemental mass spectrometry. The scope of ICP-MS has dramatically changed since its inception, and nowadays, it is a mature platform technology that is compatible with chromatographic and laser ablation (LA) systems. Over the last decades, it kept pace with various technological advances and was inspired by interdisciplinary approaches which endorsed new areas of applications. While the first part of this review was dedicated to fundamentals in ICP-MS, its hyphenated techniques and the application in biomonitoring, isotope ratio analysis, elemental speciation analysis, and elemental bioimaging, this second part will introduce relatively current directions in ICP-MS and their potential to provide novel perspectives in the medical sciences. In this context, current directions for the characterisation of novel nanomaterials which are considered for biomedical applications like drug delivery and imaging platforms will be discussed while considering different facets of ICP-MS including single event analysis and dedicated hyphenated techniques. Subsequently, immunochemistry techniques will be reviewed in their capability to expand the scope of ICP-MS enabling analysis of a large range of biomolecules alongside elements. These methods inspired mass cytometry and imaging mass cytometry and have the potential to transform diagnostics and treatment by offering new paradigms for personalised medicine. Finally, the interlacing of immunochemistry methods, single event analysis, and functional nanomaterials has opened new horizons to design novel bioassays which promise potential as assets for clinical applications and larger screening programs and will be discussed in their capabilities to detect low-level proteins and nucleic acids.
Collapse
Affiliation(s)
- David Clases
- Nano Mirco LAB, Institute of Chemistry, University of Graz, Graz, Austria.
| | | |
Collapse
|
11
|
Wong MCS, Deng Y, Huang J, Bai Y, Wang HHX, Yuan J, Zhang L, Yip HC, Chiu PWY. Performance of screening tests for esophageal squamous cell carcinoma: a systematic review and meta-analysis. Gastrointest Endosc 2022; 96:197-207.e34. [PMID: 35413332 DOI: 10.1016/j.gie.2022.04.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 04/04/2022] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS This systematic review and meta-analysis aims to compare the pooled diagnostic accuracy of the currently available esophageal squamous cell carcinoma (ESCC) screening tests. METHODS A comprehensive literature search of Embase and Medline (up to October 31, 2020) was performed to identify eligible studies. We pooled sensitivity, specificity, positive likelihood ratio, negative likelihood ratio, and diagnostic odds ratio for ESCC screening tools using a bivariate random-effects model. The summary receiver operating characteristic curves with area under the curve (AUC) were plotted for each screening test. RESULTS We included 161 studies conducted in 81 research articles involving 32,209 subjects. The pooled sensitivity, specificity, and AUC of the major screening tools were respectively as follows: endoscopy (peroral endoscopy): .94 (95% confidence interval [CI], .87-.97), .92 (95% CI, .87-.95), and .97 (95% CI, .96-.99); endoscopy (transnasal endoscopy): .85 (95% CI, .70-.93), .96 (95% CI, .91-.98), and .97 (95% CI, .95-.98); microRNA: .77 (95% CI, .75-.80), .78 (95% CI, .75-.80), and .85 (95% CI, .81-.87); autoantibody: .45 (95% CI, .36-.53), .91 (95% CI, .89-.93), and .84 (95% CI, .81-.87); and cytology: .82 (95% CI, .60-.93), .97 (95% CI, .88-.99), and .97 (95% CI, .95-.98). There was high heterogeneity. CONCLUSIONS The diagnostic accuracy seemed to be comparable between cytology and endoscopy, whereas autoantibody and microRNAs bear potential as future noninvasive screening tools for ESCC. To reduce ESCC-related death in high-risk populations, it is important to develop a more accurate and less-invasive screening test.
Collapse
Affiliation(s)
- Martin C S Wong
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong SAR, China; School of Public Health, Peking Union Medical College and The Chinese Academy of Medical Sciences, Beijing, China; Department of Global Health, School of Public Health, Peking University, Beijing, China
| | - Yunyang Deng
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Junjie Huang
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yijun Bai
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Harry H X Wang
- School of Public Health, Sun Yat-Sen University, Guangzhou, China; General Practice and Primary Care, Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| | - Jinqiu Yuan
- Clinical Research Centre, Scientific Research Centre, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Lin Zhang
- School of Public Health, Peking Union Medical College and The Chinese Academy of Medical Sciences, Beijing, China; Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Hon Chi Yip
- Department of Surgery, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Philip Wai Yan Chiu
- Department of Surgery, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
12
|
Brindl N, Boekhoff H, Bauer AS, Gaida MM, Dang HT, Kaiser J, Hoheisel JD, Felix K. Use of Autoreactive Antibodies in Blood of Patients with Pancreatic Intraductal Papillary Mucinous Neoplasms (IPMN) for Grade Distinction and Detection of Malignancy. Cancers (Basel) 2022; 14:cancers14153562. [PMID: 35892825 PMCID: PMC9332220 DOI: 10.3390/cancers14153562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/17/2022] [Accepted: 07/20/2022] [Indexed: 11/16/2022] Open
Abstract
(1) Background: A reliable non-invasive distinction between low- and high-risk pancreatic intraductal papillary mucinous neoplasms (IPMN) is needed to effectively detect IPMN with malignant potential. This would improve preventative care and reduce the risk of developing pancreatic cancer and overtreatment. The present study aimed at exploring the presence of autoreactive antibodies in the blood of patients with IPMN of various grades of dysplasia. (2) Methods: A single-center cohort was studied composed of 378 serum samples from patients with low-grade IPMN (n = 91), high-grade IPMN (n = 66), IPMN with associated invasive cancer (n = 30), pancreatic ductal adenocarcinoma (PDAC) stages T1 (n = 24) and T2 (n = 113), and healthy controls (n = 54). A 249 full-length recombinant human protein microarray was used for profiling the serum samples. (3) Results: 14 proteins were identified as potential biomarkers for grade distinction in IPMN, yielding high specificity but mediocre sensitivity. (4) Conclusions: The identified autoantibodies are potential biomarkers that may assist in the detection of malignancy in IPMN patients.
Collapse
Affiliation(s)
- Niall Brindl
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, 69120 Heidelberg, Germany;
- Division of Functional Genome Analysis, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (H.B.); (A.S.B.); (J.D.H.)
- Correspondence: (N.B.); (K.F.); Tel.: +49-163-638-1860 (N.B.)
| | - Henning Boekhoff
- Division of Functional Genome Analysis, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (H.B.); (A.S.B.); (J.D.H.)
| | - Andrea S. Bauer
- Division of Functional Genome Analysis, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (H.B.); (A.S.B.); (J.D.H.)
| | - Matthias M. Gaida
- Institute of Pathology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany;
- Research Center for Immunotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- TRON, Translational Oncology at the University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Hien T. Dang
- Department of Surgery, Thomas Jefferson University, Philadelphia, PA 19144, USA;
| | - Jörg Kaiser
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, 69120 Heidelberg, Germany;
| | - Jörg D. Hoheisel
- Division of Functional Genome Analysis, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (H.B.); (A.S.B.); (J.D.H.)
| | - Klaus Felix
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, 69120 Heidelberg, Germany;
- Correspondence: (N.B.); (K.F.); Tel.: +49-163-638-1860 (N.B.)
| |
Collapse
|
13
|
Li W, Sun Y, Xu N. Application Value Analysis of Seven-Autoantibody Panel in the Lung Cancer Screening in Yunnan. COMPUTATIONAL INTELLIGENCE AND NEUROSCIENCE 2022; 2022:2368155. [PMID: 35832243 PMCID: PMC9273362 DOI: 10.1155/2022/2368155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 12/24/2022]
Abstract
Objective To study the application value of seven-autoantibody (anti-CAGE, anti-GAGE7, anti-GBU4-5, anti-MAGE A1, anti-P53, and anti-PGP9.5, anti-SOX2) in lung cancer (LC) screening in Yunnan. Methods The clinical data of 329 lung cancer patients and 202 nonlung cancer controls in the First People's Hospital of Yunnan Province from November 2018 to April 2022 were retrospectively analyzed. The detection results of anti-CAGE, anti-GAGE7, anti-GBU4-5, anti-MAGE A1, anti-P53, anti-PGP9.5, and anti-SOX2 were collected. The receiver operating curve (ROC) was used to analyze the value of these seven-autoantibody in the detection of LC alone and in combination, and the area under the curve (AUC), sensitivity, specificity, and cut-off values were calculated. Results The levels of anti-CAGE, anti-GAGE7, anti-GBU4-5, anti-MAGE A1, anti-P53, anti-PGP9.5 and anti-SOX2 in LC patients were significantly higher than those in the controls, and the differences were statistically significant (P < 0.001). The AUC of anti-CAGE, anti-GAGE7, anti-GBU4-5, anti-MAGE A1, anti-P53, anti-PGP9.5, and anti-SOX2 for the diagnosis of LC were 0.586 (95% CI: 0.537-0.634, P < 0.001), 0.620 (95% CI: 0.572-0.667, P < 0.001), 0.570 (95% CI: 0.521-0.619, P=0.007), 0.612 (95% CI: 0.563-0.660, P < 0.007) 0.001), 0.561 (95% CI: 0.510-0.611, P=0.019), 0.667 (95% CI: 0.619-0.715, P < 0.001), 0.587 (95% CI: 0.538-0.636, P < 0.001), respectively. The AUC of combined detection of LC was 0.719 (95% CI: 0.676-0.761, P < 0.001). The positive rate of the combined detection of seven-autoantibody in the LC group was 48.02% (158/329), which was significantly higher than that of the control group (13.86% (28/202)), the difference was statistically significant (χ 2 = 64.183, P < 0.001). Conclusion The individual detection and combined detection of the seven-autoantibody have a certain value in the diagnosis of LC in Yunnan, and it can provide a certain reference for clinical LC screening.
Collapse
Affiliation(s)
- Wenrun Li
- Department of Medical Laboratory, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Yi Sun
- Department of Medical Laboratory, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Ning Xu
- Department of Medical Laboratory, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| |
Collapse
|
14
|
B-Cell-Based Immunotherapy: A Promising New Alternative. Vaccines (Basel) 2022; 10:vaccines10060879. [PMID: 35746487 PMCID: PMC9227543 DOI: 10.3390/vaccines10060879] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/23/2022] [Accepted: 05/23/2022] [Indexed: 12/04/2022] Open
Abstract
The field of immunotherapy has undergone radical conceptual changes over the last decade. There are various examples of immunotherapy, including the use of monoclonal antibodies, cancer vaccines, tumor-infecting viruses, cytokines, adjuvants, and autologous T cells carrying chimeric antigen receptors (CARs) that can bind cancer-specific antigens known as adoptive immunotherapy. While a lot has been achieved in the field of T-cell immunotherapy, only a fraction of patients (20%) see lasting benefits from this mode of treatment, which is why there is a critical need to turn our attention to other immune cells. B cells have been shown to play both anti- and pro-tumorigenic roles in tumor tissue. In this review, we shed light on the dual nature of B cells in the tumor microenvironment. Furthermore, we discussed the different factors affecting the biology and function of B cells in tumors. In the third section, we described B-cell-based immunotherapies and their clinical applications and challenges. These current studies provide a springboard for carrying out future mechanistic studies to help us unleash the full potential of B cells in immunotherapy.
Collapse
|
15
|
Yan Y, Yeon SY, Qian C, You S, Yang W. On the Road to Accurate Protein Biomarkers in Prostate Cancer Diagnosis and Prognosis: Current Status and Future Advances. Int J Mol Sci 2021; 22:13537. [PMID: 34948334 PMCID: PMC8703658 DOI: 10.3390/ijms222413537] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 12/14/2021] [Indexed: 12/11/2022] Open
Abstract
Prostate cancer (PC) is a leading cause of morbidity and mortality among men worldwide. Molecular biomarkers work in conjunction with existing clinicopathologic tools to help physicians decide who to biopsy, re-biopsy, treat, or re-treat. The past decade has witnessed the commercialization of multiple PC protein biomarkers with improved performance, remarkable progress in proteomic technologies for global discovery and targeted validation of novel protein biomarkers from clinical specimens, and the emergence of novel, promising PC protein biomarkers. In this review, we summarize these advances and discuss the challenges and potential solutions for identifying and validating clinically useful protein biomarkers in PC diagnosis and prognosis. The identification of multi-protein biomarkers with high sensitivity and specificity, as well as their integration with clinicopathologic parameters, imaging, and other molecular biomarkers, bodes well for optimal personalized management of PC patients.
Collapse
Affiliation(s)
- Yiwu Yan
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (Y.Y.); (S.Y.Y.); (C.Q.); (S.Y.)
| | - Su Yeon Yeon
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (Y.Y.); (S.Y.Y.); (C.Q.); (S.Y.)
| | - Chen Qian
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (Y.Y.); (S.Y.Y.); (C.Q.); (S.Y.)
| | - Sungyong You
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (Y.Y.); (S.Y.Y.); (C.Q.); (S.Y.)
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Wei Yang
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (Y.Y.); (S.Y.Y.); (C.Q.); (S.Y.)
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
16
|
Sena P, Mancini S, Bertacchini J, Carnevale G, Pedroni M, Roncucci L. Autoimmunity Profiles as Prognostic Indicators in Patients with Colorectal Cancer versus Those with Cancer at Other Sites: A Prospective Study. Cancers (Basel) 2021; 13:3239. [PMID: 34209517 PMCID: PMC8269181 DOI: 10.3390/cancers13133239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/19/2021] [Accepted: 06/25/2021] [Indexed: 11/26/2022] Open
Abstract
Colorectal cancer represents a paradigmatic model of inflammatory carcinogenesis accompanied by the production of several kinds of tumor-associated autoantibodies (TAABs). The specific aim of this study is to define the clinical impact of the presence of non-specific circulating TAABs in a cohort of cancer patients and to establish whether significant differences were present between colorectal cancer and cancers at other sites. For this aim a prospective study was developed and a five-year survival analysis performed. Indirect immunofluorescence on rat tissues for non-organ specific autoantibodies (NOSAs: liver-kidney-stomach), on rat colon substrates (colon-related autoantibodies, CAAs) and on HEp-2 cell lines was performed. NOSA positivity was more frequent in patients with colorectal cancer than in those with cancer at other sites. Survival analysis demonstrated a significantly worse prognosis in cancer patients positive for TAABs. CAA positivity is a predictor of survival, independently from the presence of comorbidities, and HEp-2 reactivity was a strong predictor of survival in a stepwise Cox-regression model, including stage at diagnosis. Overall overproduction of TAABs is associated with advanced oncological disease, the presence of metastasis, and poorer prognosis of cancer patients.
Collapse
Affiliation(s)
- Paola Sena
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy; (P.S.); (J.B.); (G.C.)
| | - Stefano Mancini
- Department of Internal Medicine and Rehabilitation, Santa Maria Bianca Hospital, AUSL Modena, Via A. Fogazzaro 6, 41037 Mirandola, Italy;
| | - Jessika Bertacchini
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy; (P.S.); (J.B.); (G.C.)
| | - Gianluca Carnevale
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy; (P.S.); (J.B.); (G.C.)
| | - Monica Pedroni
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy;
| | - Luca Roncucci
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy;
| |
Collapse
|
17
|
Adeniyi O, Sicwetsha S, Adesina A, Mashazi P. Immunoassay detection of tumor-associated autoantibodies using protein G bioconjugated to nanomagnet-silica decorated with Au@Pd nanoparticles. Talanta 2021; 226:122127. [PMID: 33676681 DOI: 10.1016/j.talanta.2021.122127] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 01/06/2021] [Accepted: 01/12/2021] [Indexed: 11/17/2022]
Abstract
A colorimetric immunosensor was developed for the detection of tumor-associated anti-p53 autoantibodies (anti-p53aAbs). The immunosensor platform was prepared by immobilizing human-protein (p53Ag) onto a high binding 96-well plate. The immunoassay was based on the immunometric sandwich protocol, and protein G functionalized nanomagnet-silica nanoparticles decorated with Au@Pd (Fe3O4@SiO2-NH2-Au@Pd0.30NPs-protG) was used as the detection nanobioprobe. The Fe3O4@SiO2-NH2-Au@Pd0.30NPs-protG exhibited a high binding affinity for the captured anti-p53aAbs and high catalytic performance towards the oxidation of 3,3',5,5'-tetramethylbenzidine (TMB). The oxidation of TMB resulted in significant color change and a UV-vis absorption signal. The detection was achieved by measuring the changes in UV-Vis absorption as the concentrations of anti-p53aAbs changed. The apparent binding affinity (KD) between the p53aAbs and Fe3O4@SiO2-NH2-Au@Pd0.30NPs-protG was 35.2 ng mL-1. The plot of change in the absorption intensity against the logarithm of anti-p53aAbs was linear within 1.0-500.0 ng mL-1 with a correlation coefficient (R2) of 0.98. The detection limit (LoD) using 3σ was calculated to be 15 pg mL-1, which is lower than the conventional HRP-label based colorimetric immunoassay. The real sample detection was investigated using the serum recovery method. The recovery of the anti-p53aAbs ranges from 98.5% to 105.7%, demonstrating its potential for practical applications.
Collapse
Affiliation(s)
- Omotayo Adeniyi
- Department of Chemistry, P.O. Box 94, Makhanda, 6140, South Africa
| | | | - Abiola Adesina
- Department of Chemistry, P.O. Box 94, Makhanda, 6140, South Africa
| | - Philani Mashazi
- Department of Chemistry, P.O. Box 94, Makhanda, 6140, South Africa; Institute for Nanotechnology Innovation Rhodes University, P.O. Box 94, Makhanda, 6140, South Africa.
| |
Collapse
|
18
|
de Graaf JF, Huberts M, Fouchier RAM, van den Hoogen BG. Determinants of the efficacy of viro-immunotherapy: A review. Cytokine Growth Factor Rev 2020; 56:124-132. [PMID: 32919831 DOI: 10.1016/j.cytogfr.2020.07.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 07/02/2020] [Indexed: 10/23/2022]
Abstract
Oncolytic virus immunotherapy is rapidly gaining interest in the field of immunotherapy against cancer. The minimal toxicity upon treatment and the dual activity of direct oncolysis and immune activation make therapy with oncolytic viruses (OVs) an interesting treatment modality. The safety and efficacy of several OVs have been assessed in clinical trials and, so far, the Food and Drug Administration (FDA) has approved one OV. Unfortunately, most treatments with OVs have shown suboptimal responses in clinical trials, while they appeared more promising in preclinical studies, with tumours reducing after immune cell influx. In several clinical trials with OVs, parameters such as virus replication, virus-specific antibodies, systemic immune responses, immune cell influx into tumours and tumour-specific antibodies have been studied as predictors or correlates of therapy efficacy. In this review, these studies are summarized to improve our understanding of the determinants of the efficacy of OV therapies in humans and to provide insights for future developments in the viro-immunotherapy treatment field.
Collapse
Affiliation(s)
- J F de Graaf
- Viroscience Department, Erasmus Medical Centrum, Rotterdam, The Netherlands
| | - M Huberts
- Viroscience Department, Erasmus Medical Centrum, Rotterdam, The Netherlands
| | - R A M Fouchier
- Viroscience Department, Erasmus Medical Centrum, Rotterdam, The Netherlands
| | - B G van den Hoogen
- Viroscience Department, Erasmus Medical Centrum, Rotterdam, The Netherlands.
| |
Collapse
|
19
|
Profiling of Naturally Occurring Antibodies to the Thomsen-Friedenreich Antigen in Health and Cancer: The Diversity and Clinical Potential. BIOMED RESEARCH INTERNATIONAL 2020; 2020:9747040. [PMID: 32280709 PMCID: PMC7128052 DOI: 10.1155/2020/9747040] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 03/17/2020] [Indexed: 02/07/2023]
Abstract
The Thomsen-Friedenreich (TF) antigen is expressed in a majority of human tumors due to aberrant glycosylation in cancer cells. There is strong evidence that humoral immune response to TF represents an effective mechanism for the elimination of cancer cells that express TF-positive glycoconjugates. The presence of naturally occurring antibodies to tumor-associated TF and cancer-specific changes in their levels, isotype distribution and interrelation, avidity, and glycosylation profile make these Abs a convenient and ubiquitous marker for cancer diagnostics and prognostics. In this review, we attempt to summarize the latest data on the potential of TF-specific Abs for cancer diagnostics and prognostics.
Collapse
|
20
|
Longitudinal serum autoantibody repertoire profiling identifies surgery-associated biomarkers in lung adenocarcinoma. EBioMedicine 2020; 53:102674. [PMID: 32113159 PMCID: PMC7047177 DOI: 10.1016/j.ebiom.2020.102674] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 01/30/2020] [Accepted: 01/30/2020] [Indexed: 12/18/2022] Open
Abstract
Longitudinal sera were globally analyzed for identification of surgery-associated serum biomarker for the first time. Autoantibody repertories are stable for a single individual at different time points but highly variable among individuals. Surgery-associated serum biomarkers are prevalent in lung adenocarcinoma patients.
Background Autoantibodies against tumor associated antigens are highly related to cancer progression. Autoantibodies could serve as indicators of tumor burden, and have the potential to monitor the response of treatment and tumor recurrence. However, how the autoantibody repertoire changes in response to cancer treatment are largely unknown. Methods Sera of five lung adenocarcinoma patients before and after surgery, were collected longitudinally. These sera were analyzed on a human proteome microarray of 20,240 recombinant proteins to acquire dynamic autoantibody repertoire in response to surgery, as well as to identify the antigens with decreased antibody response after tumor excision or surgery, named as surgery-associated antigens. The identified candidate antigens were then used to construct focused microarray and validated by longitudinal sera collected from a variety of time points of the same patient and a larger cohort of 45 sera from lung adenocarcinoma patients. Findings The autoantibody profiles are highly variable among patients. Meanwhile, the autoantibody profiles of the sera from the same patient were surprisingly stable for at least 3 months after surgery. Six surgery-associated antigens were identified and validated. All the five patients have at least one surgery-associated antigen, demonstrating this type of biomarkers is prevalent, while specific antigens are poorly shared among individuals. The prevalence of each antigen is 2%–14% according to the test with a larger cohort. Interpretation To our knowledge, this is the first study of dynamically profiling of autoantibody repertoires before/after surgery of cancer patients. The high prevalence of surgery-associated antigens implies the possible broad application for monitoring of tumor recurrence in population, while the low prevalence of specific antigens allows personalized medicine. After the accumulation and analysis of more longitudinal samples, the surgery-associated serum biomarkers, combined as a panel, may be applied to alarm the recurrence of tumor in a personalized manner. Funding Research supported by grants from National Key Research and Development Program of China Grant (No. 2016YFA0500600), National Natural Science Foundation of China (No. 31970130, 31600672, 31670831, and 31370813), Open Foundation of Key Laboratory of Systems Biomedicine (No. KLSB2017QN-01), Science and Technology Commission of Shanghai Municipality Medical Guidance Science &Technology Support Project (16411966100), Shanghai Municipal Education Commission-Gaofeng Clinical Medicine Grant Support (20172005), Shanghai Municipal Commission of Health and Family Planning Outstanding Academic Leaders Training Program (2017BR055) and National Natural Science Foundation of China (81871882).
Collapse
|
21
|
Zhao F, Cao M, Jiang XH, Xie K, Ye SR, Yie SM. A specific autoantibody against a novel tumour-association antigen derived from human DNA-topoiomerase I is a potential biomarker for early diagnosis and favourable prognosis in patients with colorectal carcinoma. Biomarkers 2020; 25:149-156. [PMID: 31922440 DOI: 10.1080/1354750x.2020.1714734] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Context: We previously reported a novel tumour associated antigen (TTA) with molecular weight around 48 kDa and identified the novel TTA as a fragment derived from human DNA-topoiomerase I (TOP1). We termed the novel TAA as TOPO48 and termed autoantibody against the TAA as anti-TOPO48 autoantibody.Objective: To explore the clinical significance of anti-TOPO48 autoantibody in patients with colorectal carcinoma (CRC).Materials and methods: Serum levels of the autoantibody in patients with CRC or benign tumours and healthy volunteers were measured with a specific ELISA.Results: CRC patients at early stage had higher frequency of positive levels of the autoantibody and CRC patients with positive autoantibody levels had higher overall survival rate than those with negative autoantibody levels.Conclusion: The autoantibody is a potential biomarker for early diagnosis and favourable prognosis of CRC.
Collapse
Affiliation(s)
- Feng Zhao
- State Key Laboratory of Biotherapy and Cancer, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, PR China
| | - Mei Cao
- Core Laboratory, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospitals, Chengdu, PR China
| | - Xiao-Hui Jiang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Chengdu, PR China
| | - Ke Xie
- Department of Oncology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospitals, Chengdu, PR China
| | - Shang-Rong Ye
- Chengdu Cancer Bioengineering Research Institute, Chengdu, PR China
| | - Shang-Mian Yie
- Chengdu Cancer Bioengineering Research Institute, Chengdu, PR China
| |
Collapse
|
22
|
Stable thin films of human P53 antigen on gold surface for the detection of tumour associated anti-P53 autoantibodies. Electrochim Acta 2020. [DOI: 10.1016/j.electacta.2019.135272] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
23
|
He X, Jiang XH, Yie KYX, Chen J, Zhang JB, Yie SM. An autoantibody against a 48-Kd fragment of human DNA-topoiomerase I in breast cancer: Implication for diagnosis and prognosis, and antibody-dependent cellular cytotoxicity in vitro. Cell Immunol 2019; 347:104007. [PMID: 31732123 DOI: 10.1016/j.cellimm.2019.104007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 09/25/2019] [Accepted: 10/31/2019] [Indexed: 12/18/2022]
Abstract
Previously, we reported a novel tumor-associated antigen (TAA) derived from human DNA-topoiomerase I (TOP 1). In the present study, we demonstrated that the autoantibody against the TAA could be a potential biomarker in the early diagnosis and favorable prognosis of patients with breast cancer (BC). To understand the survival benefits in BC patients, we investigated whether the autoantibody could induce antibody-dependent cellular cytotoxicity activities (ADCC) against breast cancer cells in vitro. We found that the autoantibody exhibited significant ADCC activities that destroyed breast cancer MCF-7 and MDA-MB-231cells with peripheral blood mononuclear cells (PBMCs). The ADCC activities of the autoantibody were significantly correlated with the number of natural killer (NK) cells, NKT cells, and CD4+/CD8+ T cells. Accordingly, our findings showed that the autoantibody not only represented an early index of immune response to the TAA, but also was involved in host immune defense mechanisms that initiated the destruction of cancer cells.
Collapse
Affiliation(s)
- Xu He
- Core Laboratory, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Xiao-Hui Jiang
- Key Laboratory of Birth Defects and Related Diseases for Women and Children (Sichuan University), Ministry of Education, Human Sperm Bank, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Kevin Yi-Xiao Yie
- Chengdu Cancer Bioengineering Research Institute, Chengdu, Sichuan, China
| | - Jie Chen
- Core Laboratory, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Jian-Bo Zhang
- Core Laboratory, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Shang-Mian Yie
- Chengdu Cancer Bioengineering Research Institute, Chengdu, Sichuan, China.
| |
Collapse
|
24
|
Xu L, Lee JR, Hao S, Ling XB, Brooks JD, Wang SX, Gambhir SS. Improved detection of prostate cancer using a magneto-nanosensor assay for serum circulating autoantibodies. PLoS One 2019; 14:e0221051. [PMID: 31404106 PMCID: PMC6690541 DOI: 10.1371/journal.pone.0221051] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 07/29/2019] [Indexed: 12/22/2022] Open
Abstract
Purpose To develop a magneto-nanosensor (MNS) based multiplex assay to measure protein and autoantibody biomarkers from human serum for prostate cancer (CaP) diagnosis. Materials and methods A 4-panel MNS autoantibody assay and a MNS protein assay were developed and optimized in our labs. Using these assays, serum concentration of six biomarkers including prostate-specific antigen (PSA) protein, free/total PSA ratio, as well as four autoantibodies against Parkinson disease 7 (PARK7), TAR DNA-binding protein 43 (TARDBP), Talin 1 (TLN1), and Caldesmon 1 (CALD1) and were analyzed. Human serum samples from 99 patients (50 with non-cancer and 49 with clinically localized CaP) were evaluated. Results The MNS assay showed excellent performance characteristics and no cross-reactivity. All autoantibody assays showed a statistically significant difference between CaP and non-cancer samples except for PARK7. The most significant difference was the combination of the four autoantibodies as a panel in addition to the free/total PSA ratio. This combination had the highest area under the curve (AUC)– 0.916 in ROC analysis. Conclusions Our results suggest that this autoantibody panel along with PSA and free PSA have potential to segregate patients without cancer from those with prostate cancer with higher sensitivity and specificity than PSA alone.
Collapse
Affiliation(s)
- Lingyun Xu
- Department of Radiology, Molecular Imaging Program at Stanford, Bio-X Program, Stanford University School of Medicine, Stanford, California, United States of America
| | - Jung-Rok Lee
- Division of Mechanical and Biomedical Engineering, Ewha Womans University, Seoul, South Korea
| | - Shiying Hao
- Clinical and Translational Research Program, Betty Irene Moore Children's Heart Center, Lucile Packard Children’s Hospital, Palo Alto, California, United States of America
- Departments of Surgery, Stanford University, Stanford, California, United States of America
| | - Xuefeng Bruce Ling
- Clinical and Translational Research Program, Betty Irene Moore Children's Heart Center, Lucile Packard Children’s Hospital, Palo Alto, California, United States of America
- Departments of Surgery, Stanford University, Stanford, California, United States of America
| | - James D. Brooks
- Department of Urology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Shan X. Wang
- Department of Materials Science & Engineering, Stanford University, Stanford, California, United States of America
- Department of Electrical Engineering, Stanford University, Stanford, California, United States of America
- Department of Radiology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Sanjiv Sam Gambhir
- Department of Radiology, Molecular Imaging Program at Stanford, Bio-X Program, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Electrical Engineering, Stanford University, Stanford, California, United States of America
- Department of Bioengineering, Stanford University, Stanford, California, United States of America
- * E-mail:
| |
Collapse
|
25
|
Yadav S, Kashaninejad N, Masud MK, Yamauchi Y, Nguyen NT, Shiddiky MJ. Autoantibodies as diagnostic and prognostic cancer biomarker: Detection techniques and approaches. Biosens Bioelectron 2019; 139:111315. [DOI: 10.1016/j.bios.2019.111315] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/07/2019] [Accepted: 05/08/2019] [Indexed: 01/25/2023]
|
26
|
Zhou Y, Cui J, Du H. Autoantibody-targeted TAAs in pancreatic cancer: A comprehensive analysis. Pancreatology 2019; 19:760-768. [PMID: 31255446 DOI: 10.1016/j.pan.2019.06.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 05/23/2019] [Accepted: 06/16/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Pancreatic cancer is one of the leading causes of cancer mortality and lacks efficient biomarkers for early diagnosis. In the early stages of pancreatic cancer, humoral immunity can respond to a certain amount of tumor-associated antigens (TAAs) with the production of corresponding autoantibodies. Such autoantibody-targeted TAAs (autoTAAs) are highly likely to indicate early events during pancreatic carcinogenesis. Herein, we performed a comprehensive analysis of these autoTAAs to explore their physiological function and their involvement and prognostic value in pancreatic cancer. METHODS We first searched the literature to identify the autoTAAs. A PPI network of these autoTAAs was constructed, and core network modules were extracted by Cytoscape software. GO annotation and KEGG pathway analysis were performed to analyze the main physiological functions of these autoTAAs. The prognostic value of autoTAAs in pancreatic cancer was analyzed by using RNA-seq data generated by TCGA. RESULTS The PPI network including 98 autoTAAs was constructed, and 2 subgroups were extracted as core modules. GO and KEGG analysis revealed that key functions and pathways of these autoTAAs were significantly enriched in nucleotide repair, protein synthesis, and cancer-associated events. MSH2, EZR, PGK1, VCL and ANXA2 have prognostic value in pancreatic cancer, and high mRNA expression of these 5 proteins is associated with unfavorable prognosis in pancreatic cancer. CONCLUSIONS AutoTAAs may be associated with early events in the carcinogenesis of pancreatic cancer. MSH2, EZR, PGK1, VCL and ANXA2 predict poor prognosis in pancreatic cancer. Some autoTAAs also have prognostic value in other cancers.
Collapse
Affiliation(s)
- Yabin Zhou
- 112 Lab, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Jiawen Cui
- 112 Lab, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Hongwu Du
- 112 Lab, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, China.
| |
Collapse
|
27
|
Hettegger P, Huber J, Paßecker K, Soldo R, Kegler U, Nöhammer C, Weinhäusel A. High similarity of IgG antibody profiles in blood and saliva opens opportunities for saliva based serology. PLoS One 2019; 14:e0218456. [PMID: 31220138 PMCID: PMC6586443 DOI: 10.1371/journal.pone.0218456] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 06/03/2019] [Indexed: 12/16/2022] Open
Abstract
Saliva based diagnostics is a rapidly evolving field due to the large diagnostic potential and simple sample collection. Currently only few individual molecules were investigated for their diagnostic capabilities in saliva. A systematic comparison of IgG antibody profiles in saliva and plasma is still missing in scientific literature. Our hypothesis is that IgG profiles in plasma and saliva are highly similar for each individual. As a consequence, one could implement practically any plasma based IgG assay (classical serology) as saliva based assay. In other words, the IgG antibodies found in blood are also accessible from saliva. We confirm our hypothesis by comparing IgG reactivities towards protein and peptide antigens. We isolated saliva IgG with high purity and demonstrate that plasma IgG reactivities (classical serology) can be inferred from saliva. As a showcase we perform Hepatitis B virus antibody (plasma-)titer determination from saliva. Additionally we show that plasma and saliva IgG profiles of 20 individuals are highly similar for 256 peptide antigens and match (unsupervised) with high probabilities. Finally, we argue for generalisation to the complete IgG antibody profile. The presented findings could contribute greatly to the development of saliva based diagnostic methods of numerous antibody based tests.
Collapse
Affiliation(s)
- Peter Hettegger
- Austrian Institute of Technology, Center for Health and Bioresources, Molecular Diagnostics, Vienna, Austria
| | - Jasmin Huber
- Austrian Institute of Technology, Center for Health and Bioresources, Molecular Diagnostics, Vienna, Austria
| | - Katharina Paßecker
- Austrian Institute of Technology, Center for Health and Bioresources, Molecular Diagnostics, Vienna, Austria
| | - Regina Soldo
- Austrian Institute of Technology, Center for Health and Bioresources, Molecular Diagnostics, Vienna, Austria
| | - Ulrike Kegler
- Austrian Institute of Technology, Center for Health and Bioresources, Molecular Diagnostics, Vienna, Austria
| | - Christa Nöhammer
- Austrian Institute of Technology, Center for Health and Bioresources, Molecular Diagnostics, Vienna, Austria
| | - Andreas Weinhäusel
- Austrian Institute of Technology, Center for Health and Bioresources, Molecular Diagnostics, Vienna, Austria
- * E-mail:
| |
Collapse
|
28
|
Khayeka-Wandabwa C, Ma X, Cao X, Nunna V, Pathak JL, Bernhardt R, Cai P, Bureik M. Plasma membrane localization of CYP4Z1 and CYP19A1 and the detection of anti-CYP19A1 autoantibodies in humans. Int Immunopharmacol 2019; 73:64-71. [PMID: 31082724 DOI: 10.1016/j.intimp.2019.05.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 04/14/2019] [Accepted: 05/02/2019] [Indexed: 12/15/2022]
Abstract
It is thought that autoantibody (aAb) production can be caused by (aberrant) protein targeting to the plasma surface of cells. We recently demonstrated the presence of the human cytochrome P450 enzyme CYP4Z1 on the plasma membrane of MCF-7 breast cancer cells and the detection of high titers of anti-CYP4Z1 aAbs in breast cancer patients, but not in healthy controls. In the present study we show that cells of the normal breast cell line MCF-10A do not display CYP4Z1 on their surface. By contrast, we detected CYP19A1 (aromatase) on the plasma membrane of both cell lines. Interestingly, the presence of CYPs on the cell surface did not correlate with their relative expression levels in these cell lines. Indirect ELISA experiments demonstrated the presence of anti-CYP19A1 aAbs in female breast cancer patient sera as well as in male and female controls, respectively; aAb titers in all three groups varied considerably and overall, the results obtained for each group were not significantly different from those of either of the other two groups. Based on these data we propose the hypothesis that CYP translocation to the plasma membrane, but not the intracellular expression level, is the crucial precondition for the generation of anti-CYP aAbs.
Collapse
Affiliation(s)
- Christopher Khayeka-Wandabwa
- School of Pharmaceutical Science and Technology (SPST), Health Science Platform, Tianjin University, Tianjin, China
| | - Xiaoshuang Ma
- School of Pharmaceutical Science and Technology (SPST), Health Science Platform, Tianjin University, Tianjin, China
| | - Xiaolin Cao
- School of Pharmaceutical Science and Technology (SPST), Health Science Platform, Tianjin University, Tianjin, China
| | - Venkatrao Nunna
- School of Pharmaceutical Science and Technology (SPST), Health Science Platform, Tianjin University, Tianjin, China
| | - Janak L Pathak
- School of Pharmaceutical Science and Technology (SPST), Health Science Platform, Tianjin University, Tianjin, China
| | - Rita Bernhardt
- Institute of Biochemistry, Saarland University, Saarbruecken, Germany
| | - Pengcheng Cai
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Matthias Bureik
- School of Pharmaceutical Science and Technology (SPST), Health Science Platform, Tianjin University, Tianjin, China.
| |
Collapse
|
29
|
Xu YW, Chen H, Guo HP, Yang SH, Luo YH, Liu CT, Huang XY, Tang XM, Hong CQ, Li EM, Xu LY, Peng YH. Combined detection of serum autoantibodies as diagnostic biomarkers in esophagogastric junction adenocarcinoma. Gastric Cancer 2019; 22:546-557. [PMID: 30426295 PMCID: PMC6476828 DOI: 10.1007/s10120-018-0894-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 10/25/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND We previously found that autoantibodies against a panel of six tumor-associated antigens (p53, NY-ESO-1, MMP-7, Hsp70, PRDX6 and Bmi-1) may aid in early detection of esophageal squamous cell carcinoma. Here we aimed to evaluate the diagnostic value of this autoantibody panel in esophagogastric junction adenocarcinoma (EJA) patients. METHODS Serum autoantibody levels were measured by enzyme-linked immunosorbent assay in a training cohort and a validation cohort. We used receiver-operating characteristics (ROC) to calculate diagnostic accuracy. RESULTS We recruited 169 normal controls and 122 EJA patients to the training cohort, and 80 normal controls and 70 EJA patients to the validation cohort. Detection of the autoantibody panel demonstrated an area under the curve (AUC) of 0.818, sensitivity 59.0% and specificity 90.5% in training cohort, and AUC 0.815, sensitivity 61.4% and specificity 90.0% in validation cohort in the diagnosis of EJA. Measurement of the autoantibody panel could distinguish early stage EJA patients from normal controls (AUC 0.786 and 0.786, sensitivity 50.0% and 56.0%, and specificity 90.5% and 90.0%, for training and validation cohorts, respectively). Moreover, a restricted panel consisting of autoantibodies against p53, NY-ESO-1 and Bmi-1 exhibited similar diagnostic performance for EJA (AUC 0.814 and 0.823, sensitivity 53.5% and 60.0%, and specificity 90.5% and 93.7%, for training and validation cohorts, respectively) and early stage EJA (AUC 0.744 and 0.773, sensitivity 55.6% and 52.0%, and specificity 90.5% and 93.7%, for training and validation cohorts, respectively). CONCLUSIONS Autoantibodies against an optimized TAA panel as serum biomarkers appear to help identify the present of early stage EJA.
Collapse
Affiliation(s)
- Yi-Wei Xu
- Department of Clinical Laboratory Medicine, The Cancer Hospital of Shantou University Medical College, Shantou, 515041, People's Republic of China
- Guangdong Esophageal Cancer Research Institute, Shantou University Medical College, Shantou, 515041, People's Republic of China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, 515041, People's Republic of China
| | - Hao Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Hai-Peng Guo
- Department of Head and Neck Surgery, The Cancer Hospital of Shantou University Medical College, Shantou, 515041, People's Republic of China
| | - Shi-Han Yang
- Department of Dermatology and Venereology, Shantou Central Hospital, Shantou, 515041, People's Republic of China
| | - Yu-Hao Luo
- Department of Clinical Laboratory Medicine, The Cancer Hospital of Shantou University Medical College, Shantou, 515041, People's Republic of China
| | - Can-Tong Liu
- Department of Clinical Laboratory Medicine, The Cancer Hospital of Shantou University Medical College, Shantou, 515041, People's Republic of China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, 515041, People's Republic of China
| | - Xin-Yi Huang
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, 515041, People's Republic of China
| | - Xue-Miao Tang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Chao-Qun Hong
- Department of Oncological Laboratory Research, The Cancer Hospital of Shantou University Medical College, Shantou, 515041, People's Republic of China
| | - En-Min Li
- Guangdong Esophageal Cancer Research Institute, Shantou University Medical College, Shantou, 515041, People's Republic of China.
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, 515041, People's Republic of China.
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, 515041, People's Republic of China.
| | - Li-Yan Xu
- Guangdong Esophageal Cancer Research Institute, Shantou University Medical College, Shantou, 515041, People's Republic of China.
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, 515041, People's Republic of China.
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou, 515041, People's Republic of China.
| | - Yu-Hui Peng
- Department of Clinical Laboratory Medicine, The Cancer Hospital of Shantou University Medical College, Shantou, 515041, People's Republic of China.
- Guangdong Esophageal Cancer Research Institute, Shantou University Medical College, Shantou, 515041, People's Republic of China.
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, 515041, People's Republic of China.
| |
Collapse
|
30
|
Rajappa S, Sharma S, Prasad K. Unmet Clinical Need in the Management of Locally Advanced Unresectable Lung Cancer: Treatment Strategies to Improve Patient Outcomes. Adv Ther 2019; 36:563-578. [PMID: 30693419 DOI: 10.1007/s12325-019-0876-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Indexed: 12/19/2022]
Abstract
Stage III locally advanced non-small cell lung cancer (LA NSCLC) comprises the most heterogeneous group of patients, accounts for one-third of patients with lung cancer, and is unresectable at presentation. Multiple treatment approaches have evolved over the past few decades focusing on timing of chemoradiation (concurrent vs. sequential) and sequencing of therapy (induction vs. consolidation). Concurrent chemoradiation (CCRT) emerged as the standard of care for the majority of the patients worldwide. Despite improvements in median and overall survival (OS) using the concurrent approach, the rate of distant failure remains high. Consolidation with chemotherapy or targeted agents, adding more radiation dose, or induction chemotherapy did not improve OS. With continued research on defining optimal radiation doses and schedules and integrating novel systemic agents, immunotherapy consolidation has renewed optimism. Synergistic use of radiation and immunotherapy can prevent micrometastatic disease and reduce local failure and may have an abscopal effect in addition to survival benefits. The PACIFIC study reported an absolute progression-free survival benefit of 11.2 months with durvalumab consolidation after standard CCRT compared with placebo. The OS data with durvalumab consolidation are encouraging. Durvalumab is the only approved immunotherapy for unresectable stage III LA NSCLC. Improved survival confirms the definitive role of durvalumab as an effective adjuvant therapy after CCRT with no new safety signals. However, the potential mechanisms driving interaction between immunotherapy and chemoradiotherapy require definitive investigation. These mechanisms may help define the timing of immunotherapy initiation as neoadjuvant, adjuvant, or consolidation and maintenance therapy after progression. FUNDING: AstraZeneca Pharma India Limited.
Collapse
Affiliation(s)
- Senthil Rajappa
- Basavatarakam Indo-American Cancer Hospital and Research Institute, Hyderabad, India.
| | | | | |
Collapse
|
31
|
Music M, Prassas I, Diamandis EP. Optimizing cancer immunotherapy: Is it time for personalized predictive biomarkers? Crit Rev Clin Lab Sci 2018; 55:466-479. [PMID: 30277835 DOI: 10.1080/10408363.2018.1499706] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cancer immunotherapy, a treatment that selectively augments a patient's anti-tumor immune response, is a breakthrough advancement in personalized medicine. A subset of cancer patients undergoing immunotherapy have displayed robust and long-lasting therapeutic responses. Currently, the spotlight is on the use of blocking antibodies against the T-cell checkpoint molecules, cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and programed cell death-1 (PD-1)/programed death-ligand 1 (PD-L1), which have been effectively used to combat many cancers types. Despite the overall enthusiasm, immune checkpoint blockade inhibitors suffer from significant limitations such as high cost, serious toxicity in a substantial proportion of patients, and a response rate as low as 10%-40% in some clinical trials. Consequently, there is an urgent and unmet medical need for companion biomarkers that could both predict the response of individual patients to these therapies, and provide the means for precise monitoring of their therapeutic outcome. In this era of precision medicine, predictive biomarkers are a hot commodity because they can effectively separate responders from non-responders, and spare non-responders from serious therapy-related toxicity. Emerging predictive biomarkers for immune checkpoint blockade are: PD-L1 expression, increased amounts of tumor-infiltrating lymphocytes, increased mutational load and mismatch repair deficiency. Other well-studied biomarkers include inflammatory infiltrate, absolute lymphocyte count and lactate dehydrogenase levels. We review recent progress on predictive cancer biomarkers in immunotherapy, with a special emphasis on serum autoantibodies that have the potential to be personalized for optimal clinical outcomes.
Collapse
Affiliation(s)
- Milena Music
- a Department of Laboratory Medicine and Pathobiology , University of Toronto , Toronto , Canada
| | - Ioannis Prassas
- b Department of Pathology and Laboratory Medicine , Mount Sinai Hospital , Toronto , Canada
| | - Eleftherios P Diamandis
- a Department of Laboratory Medicine and Pathobiology , University of Toronto , Toronto , Canada.,b Department of Pathology and Laboratory Medicine , Mount Sinai Hospital , Toronto , Canada.,c Department of Clinical Biochemistry , University Health Network , Toronto , Canada.,d Lunenfeld-Tanenbaum Research Institute , Mount Sinai Hospital , Toronto , Canada
| |
Collapse
|
32
|
Mohsenzadegan M, Saebi F, Yazdani M, Abolhasani M, Saemi N, Jahanbani F, Farajollahi MM. Autoantibody against new gene expressed in prostate protein is traceable in prostate cancer patients. Biomark Med 2018; 12:1125-1138. [DOI: 10.2217/bmm-2018-0069] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Aim: We assessed an autoantibody against new gene expressed in prostate (NGEP) protein for prostate cancer (PCa) that may better diagnosis and prognosis approaches in the patients with PCa. Methods: Autoantibodies against NGEP were measured in sera of PCa patients by ELISA. Results: The autoantibody against NGEP is present in a significantly higher proportion in the sera of PCa patients as compared with healthy controls (p < 0.001). An inverse significant correlation was found between seropositive patients and Gleason score (p < 0.05) and serum prostate-specific antigen (recombinant NGEP; p < 0.05). Conclusion: The data showed that measurement of autoantibody against NGEP as a novel prostate-specific antigen in sera can be used as a potential biomarker to discriminate well-differentiated PCa patients from normal subjects.
Collapse
Affiliation(s)
- Monireh Mohsenzadegan
- Cellular & Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Laboratory Science, Faculty of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Saebi
- Cellular & Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mehdi Yazdani
- Hasheminejad Kidney Center Laboratory, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Abolhasani
- Hasheminejad Kidney Center Laboratory, Iran University of Medical Sciences, Tehran, Iran
- Oncopathology Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Nazanin Saemi
- Department of Medical Biotechnology, Faculty of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Jahanbani
- Department of Genetic, Standford University School of Medicine, CA, USA
| | - Mohammad M Farajollahi
- Cellular & Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Biotechnology, Faculty of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
33
|
The Thomsen-Friedenreich Antigen-Specific Antibody Signatures in Patients with Breast Cancer. BIOMED RESEARCH INTERNATIONAL 2018; 2018:9579828. [PMID: 30105268 PMCID: PMC6076901 DOI: 10.1155/2018/9579828] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 07/03/2018] [Indexed: 01/23/2023]
Abstract
Alterations in the glycosylation of serum total immunoglobulins show these antibodies to have a diagnostic potential for cancer but the disease-related Abs to the tumor-associated antigens, including glycans, have still poorly been investigated in this respect. We analysed serum samples from patients with breast carcinoma (n = 196) and controls (n = 64) for the level of Thomsen-Friedenreich (TF) antigen-specific antibody isotypes, their sialylation, interrelationships, and the avidity by using ELISA with the synthetic TF-polyacrylamide conjugate as an antigen and the sialic acid-specific Sambucus nigra agglutinin (SNA) and ammonium thiocyanate as a chaotrope. An increased sialylation of IgG and IgM, but a lower SNA reactivity of IgA TF antibodies, and a higher level and avidity of the TF-specific IgA were found in cancer patients. Other cancer-related signatures were the highly significant increase of the IgG/IgA ratio and the very low SNA/IgA index in cancer, including patients with an early stage of the disease. These changes showed a good diagnostic potential with about 80% accuracy. Thus, the level of naturally occurring anti-TF antigen antibodies, their sialylation profile, isotype distribution, and avidity displayed cancer-specific changes that could serve as novel noninvasive Ab-based biomarkers for early breast cancer.
Collapse
|
34
|
Zhang JB, Cao M, Chen J, Ye SR, Xie K, He X, Ma XL, Zhang J, Yie SM. Serum anti-TOPO48 autoantibody as a biomarker for early diagnosis and prognosis in patients with esophageal squamous cell carcinoma. Clin Res Hepatol Gastroenterol 2018; 42:276-284. [PMID: 29170084 DOI: 10.1016/j.clinre.2017.09.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Revised: 09/03/2017] [Accepted: 09/21/2017] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND AIM We previously reported a novel tumor associated antigen (TTA) with molecular weight around 48kDa that is a fragment derived from human DNA-topoiomerase I (TOP1). We termed the novel TAA as TOPO48 and termed autoantibody against the TAA as anti-TOPO48 autoantibody. The aim of this study is to further investigate the clinical applications of the autoantibody in patients with esophageal squamous cell carcinomas (ESCC). METHODS Serum levels of the anti-TOPO48 autoantibody in 112 ESCC patients, 112 age- and gender-matched healthy controls and 75 patients with esophageal benign tumors were determined by using a specific anti-TOPO48 autoantibody ELISA. Then, we statistically evaluated its clinical significance. RESULTS We found that serum anti-TOPO48 autoantibody levels in ESCC patients were significantly higher than that in healthy controls and benign tumor patients (P=0.001). The percentage of sera with a positive level of anti-TOPO48 autoantibody in early stages was significantly higher than that in advanced stages of the cancer patients when the maximum level of healthy control sera was taken as a cut-off value (P=0.001). The area under ROC curve was 0.863 (95% CI=0.797-0.928) for healthy controls vs. early stage ESCC. In addition, patients with positive anti-TOPO48 autoantibody had significantly higher survival rate and longer survival time than that with negative anti-TOPO48 autoantibody in cancer patients (P=0.038, 0.025 and 0.047 for all stages, early stage and advanced stage, respectively). CONCLUSIONS Our results suggest that anti-TOPO48 autoantibody may be a potentially useful biomarker for early diagnosis and prognosis of ESCC.
Collapse
Affiliation(s)
- Jian-Bo Zhang
- Core Laboratory, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospitals, 610072 Chengdu, Sichuan, PR, China
| | - Mei Cao
- Core Laboratory, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospitals, 610072 Chengdu, Sichuan, PR, China
| | - Jie Chen
- Core Laboratory, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospitals, 610072 Chengdu, Sichuan, PR, China
| | - Shang-Rong Ye
- Chengdu Cancer Bioengineering Research Institute, 610075 Chengdu, Sichuan PR, China
| | - Ke Xie
- Department of Oncology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospitals, 610072 Chengdu, Sichuan, PR, China
| | - Xu He
- Core Laboratory, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospitals, 610072 Chengdu, Sichuan, PR, China
| | - Xiao-Li Ma
- Chengdu Cancer Bioengineering Research Institute, 610075 Chengdu, Sichuan PR, China
| | - Jia Zhang
- Chengdu Cancer Bioengineering Research Institute, 610075 Chengdu, Sichuan PR, China
| | - Shang-Mian Yie
- Core Laboratory, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospitals, 610072 Chengdu, Sichuan, PR, China; Chengdu Cancer Bioengineering Research Institute, 610075 Chengdu, Sichuan PR, China.
| |
Collapse
|
35
|
Evans RL, Pottala JV, Nagata S, Egland KA. Longitudinal autoantibody responses against tumor-associated antigens decrease in breast cancer patients according to treatment modality. BMC Cancer 2018; 18:119. [PMID: 29386014 PMCID: PMC5793406 DOI: 10.1186/s12885-018-4022-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 01/23/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Metastatic breast cancer (BCa) is most often diagnosed months after completion of treatment of the primary tumor when a patient reports physical symptoms. Besides a physical examination, no other alternative recurrence screening method is recommended for routine follow-up care. Detection of autoantibodies against tumor-associated antigens (TAAs) has demonstrated promise for distinguishing healthy women from patients diagnosed with primary BCa. However, it is unknown what changes occur to patient autoantibody levels during and after treatment. METHODS Three serial blood draws were collected from 200 BCa patients: before treatment, 6 and 12 months after surgery. Patients were categorized according to treatment regimen, including surgery, chemotherapy, radiation, trastuzumab and hormonal therapies. The longitudinal samples were assayed for autoantibody responses against 32 conformation-carrying TAAs using a Luminex multiplex bead assay. RESULTS The treatment modality groups that had the greatest decrease in autoantibody response levels were radiation + hormonal therapy; radiation + chemotherapy; and radiation + hormonal therapy + chemotherapy. For these three treatment groups, autoantibody responses against 9 TAAs (A1AT, ANGPTL4, CAPC, CST2, DKK1, GFRA1, GRN, LGALS3 and LRP10) were significantly reduced at 12 months after surgery compared to before treatment. One TAA, GRP78, had a significantly increased autoantibody response after 12 months. CONCLUSIONS Single treatment regimens alone did not significantly alter autoantibodies levels against the studied TAAs. Radiation treatment was the common denominator of the three most affected groups for significant changes in autoantibody response levels.
Collapse
Affiliation(s)
- Rick L Evans
- Cancer Biology Research Center, Sanford Research, Sioux Falls, SD, USA
| | - James V Pottala
- Sanford School of Medicine, University of South Dakota, 2301 East 60th Street North, Sioux Falls, SD, 57104, USA
| | - Satoshi Nagata
- Center for Drug Design Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki-City, Osaka, 5670085, Japan
| | - Kristi A Egland
- Cancer Biology Research Center, Sanford Research, Sioux Falls, SD, USA. .,Sanford School of Medicine, University of South Dakota, 2301 East 60th Street North, Sioux Falls, SD, 57104, USA.
| |
Collapse
|
36
|
Yang Q, Roehrl MH, Wang JY. Proteomic profiling of antibody-inducing immunogens in tumor tissue identifies PSMA1, LAP3, ANXA3, and maspin as colon cancer markers. Oncotarget 2017; 9:3996-4019. [PMID: 29423100 PMCID: PMC5790517 DOI: 10.18632/oncotarget.23583] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 06/27/2017] [Indexed: 12/13/2022] Open
Abstract
We hypothesized that cancer tissue immunogens - antigens capable of inducing specific antibody production in patients - are promising targets for development of precision diagnostics and humoral immunotherapies. We developed an innovative immuno-proteomic strategy and identified new immunogenic markers of colon cancer. Proteins from cancers and matched normal tissues were separated by 2D gel electrophoresis and blotted with serum antibodies from the same patients. Antibody-reactive proteins were sequenced by mass spectrometry and validated by Western blotting and immunohistochemistry. 170 serum antibody-reactive proteins were identified only in cancerous but not matched normal. Among these, proteasome subunit alpha type 1 (PSA1), leucine aminopeptidase 3 (LAP3), annexin A3 (ANXA3), and maspin (serpin B5) were reproducibly found in tissues from three patients. Differential expression patterns were confirmed in samples from eight patients with various stages of colon adenocarcinoma and liver metastases. These tumor-resident proteins and/or their associated serum antibodies may be promising markers for colon cancer screening and early diagnosis. Furthermore, tumor tissue-specific antibodies could potentially be exploited as immunotherapeutic targets against cancer. More generally, proteomic profiling of antibody-inducing cancer-associated immunogens represents a powerful generic method for uncovering the tumor antigen-ome, i.e., the totality of immunogenic tumor-associated proteins.
Collapse
Affiliation(s)
- Qian Yang
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.,Ontario Cancer Institute, University Health Network, Toronto, Canada
| | - Michael H Roehrl
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.,Ontario Cancer Institute, University Health Network, Toronto, Canada.,Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Julia Y Wang
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.,Ontario Cancer Institute, University Health Network, Toronto, Canada.,Curandis, Boston, MA, USA
| |
Collapse
|
37
|
Chen H, Werner S, Butt J, Zörnig I, Knebel P, Michel A, Eichmüller SB, Jäger D, Waterboer T, Pawlita M, Brenner H. Prospective evaluation of 64 serum autoantibodies as biomarkers for early detection of colorectal cancer in a true screening setting. Oncotarget 2017; 7:16420-32. [PMID: 26909861 PMCID: PMC4941325 DOI: 10.18632/oncotarget.7500] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 02/05/2016] [Indexed: 02/07/2023] Open
Abstract
Novel blood-based screening tests are strongly desirable for early detection of colorectal cancer (CRC). We aimed to identify and evaluate autoantibodies against tumor-associated antigens as biomarkers for early detection of CRC. 380 clinically identified CRC patients and samples of participants with selected findings from a cohort of screening colonoscopy participants in 2005–2013 (N=6826) were included in this analysis. Sixty-four serum autoantibody markers were measured by multiplex bead-based serological assays. A two-step approach with selection of biomarkers in a training set, and validation of findings in a validation set, the latter exclusively including participants from the screening setting, was applied. Anti-MAGEA4 exhibited the highest sensitivity for detecting early stage CRC and advanced adenoma. Multi-marker combinations substantially increased sensitivity at the price of a moderate loss of specificity. Anti-TP53, anti-IMPDH2, anti-MDM2 and anti-MAGEA4 were consistently included in the best-performing 4-, 5-, and 6-marker combinations. This four-marker panel yielded a sensitivity of 26% (95% CI, 13–45%) for early stage CRC at a specificity of 90% (95% CI, 83–94%) in the validation set. Notably, it also detected 20% (95% CI, 13–29%) of advanced adenomas. Taken together, the identified biomarkers could contribute to the development of a useful multi-marker blood-based test for CRC early detection.
Collapse
Affiliation(s)
- Hongda Chen
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Simone Werner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Julia Butt
- Division of Molecular Diagnostics of Oncogenic Infections, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Inka Zörnig
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), Internal Medicine VI, University of Heidelberg, Heidelberg, Germany
| | - Phillip Knebel
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Angelika Michel
- Division of Molecular Diagnostics of Oncogenic Infections, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan B Eichmüller
- GMP & T cell Therapy Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dirk Jäger
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), Internal Medicine VI, University of Heidelberg, Heidelberg, Germany
| | - Tim Waterboer
- Division of Molecular Diagnostics of Oncogenic Infections, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Pawlita
- Division of Molecular Diagnostics of Oncogenic Infections, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
38
|
Hontani K, Tsuchikawa T, Hiwasa T, Nakamura T, Ueno T, Kushibiki T, Takahashi M, Inoko K, Takano H, Takeuchi S, Dosaka-Akita H, Kuwatani M, Sakamoto N, Hatanaka Y, Mitsuhashi T, Shimada H, Shichinohe T, Hirano S. Identification of novel serum autoantibodies against EID3 in non-functional pancreatic neuroendocrine tumors. Oncotarget 2017; 8:106206-106221. [PMID: 29290942 PMCID: PMC5739727 DOI: 10.18632/oncotarget.22175] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 10/14/2017] [Indexed: 12/16/2022] Open
Abstract
Pancreatic neuroendocrine tumors (pNETs) are relatively rare heterogenous tumors, comprising only 1–2% of all pancreatic neoplasms. The majority of pNETs are non-functional tumors (NF-pNETs) that do not produce hormones, and as such, do not cause any hormone-related symptoms. As a result, these tumors are often diagnosed at an advanced stage because patients do not present with specific symptoms. Although tumor markers are used to help diagnosis and predict some types of cancers, chromogranin A, a widely used tumor marker of pNETs, has significant limitations. To identify novel NF-pNET-associated antigens, we performed serological identification of antigens by recombinant cDNA expression cloning (SEREX) and identified five tumor antigens (phosphatase and tensin homolog, EP300-interacting inhibitor of differentiation 3 [EID3], EH domain-containing protein 1, galactoside-binding soluble 9, and BRCA1-associated protein). Further analysis using the AlphaLISA® immunoassay to compare serum antibody levels revealed that antibody levels against the EID3 antigen was significantly higher in the patient group than in the healthy donor group (n = 25, both groups). In addition, higher serum anti-EID3 antibody levels in NF-pNET patients correlated with shorter disease-free survival. The AUC calculated by ROC analysis was 0.784 with moderate diagnostic accuracy. In conclusion, serum anti-EID3 antibody levels may be useful as a tumor marker for prediction of tumor recurrence in NF-pNETs.
Collapse
Affiliation(s)
- Koji Hontani
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Takahiro Tsuchikawa
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Takaki Hiwasa
- Department of Biochemistry and Genetics, Chiba University, Chuo Ku, Chiba 260-8670, Japan
| | - Toru Nakamura
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Takashi Ueno
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Toshihiro Kushibiki
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Mizuna Takahashi
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Kazuho Inoko
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Hironobu Takano
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Satoshi Takeuchi
- Department of Medical Oncology, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Hirotoshi Dosaka-Akita
- Department of Medical Oncology, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Masaki Kuwatani
- Department of Gastroenterology and Hematology, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Naoya Sakamoto
- Department of Gastroenterology and Hematology, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Yutaka Hatanaka
- Department of Translational Pathology, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Tomoko Mitsuhashi
- Department of Translational Pathology, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Hideaki Shimada
- Department of Surgery, School of Medicine, Toho University, Ota-ku, Tokyo 143-8541, Japan
| | - Toshiaki Shichinohe
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Satoshi Hirano
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| |
Collapse
|
39
|
Yadav S, Masud MK, Islam MN, Gopalan V, Lam AKY, Tanaka S, Nguyen NT, Hossain MSA, Li C, Yamauchi Y, Shiddiky MJA. Gold-loaded nanoporous iron oxide nanocubes: a novel dispersible capture agent for tumor-associated autoantibody analysis in serum. NANOSCALE 2017. [PMID: 28627551 DOI: 10.1039/c7nr03006a] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Autoantibodies are produced against tumor associated antigens (TAAs) long before the appearance of any symptoms and thus can serve as promising, non-invasive biomarkers for early diagnosis of cancer. Current conventional methods for autoantibody detection are highly invasive and mostly provide diagnosis in the later stages of cancer. Herein we report a new electrochemical method for early detection of p53 autoantibodies against colon cancer using a strategy that combines the strength of gold-loaded nanoporous iron oxide nanocube (Au@NPFe2O3NC)-based capture and purification while incorporating the inherent simplicity, inexpensive, and portable nature of the electrochemical and naked-eye colorimetric readouts. After the functionalisation of Au@NPFe2O3NC with p53 antigens, our method utilises a two-step strategy that involves (i) magnetic capture and isolation of autoantibodies using p53/Au@NPFe2O3NC as 'dispersible nanocapture agents' in serum samples and (ii) subsequent detection of autoantibodies through a peroxidase-catalyzed reaction on a commercially available disposable screen-printed electrode or naked-eye detection in an Eppendorf tube. This method has demonstrated a good sensitivity (LOD = 0.02 U mL-1) and reproducibility (relative standard deviation, %RSD = <5%, for n = 3) for detecting p53 autoantibodies in serum and has also been successfully applied to analyse a small cohort of clinical samples obtained from colorectal cancer. We believe that the highly inexpensive, rapid, sensitive, and specific nature of our assay could potentially aid in the development of an early diagnostic tool for cancer and related diseases.
Collapse
Affiliation(s)
- Sharda Yadav
- School of Natural Sciences, Griffith University, Nathan Campus, QLD 4111, Australia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Protein Array-based Approaches for Biomarker Discovery in Cancer. GENOMICS PROTEOMICS & BIOINFORMATICS 2017; 15:73-81. [PMID: 28392481 PMCID: PMC5414965 DOI: 10.1016/j.gpb.2017.03.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 03/24/2017] [Accepted: 03/30/2017] [Indexed: 01/15/2023]
Abstract
Biomarkers are deemed to be potential tools in early diagnosis, therapeutic monitoring, and prognosis evaluation for cancer, with simplicity as well as economic advantages compared with computed tomography and biopsy. However, most of the current cancer biomarkers present insufficient sensitivity as well as specificity. Therefore, there is urgent requirement for the discovery of biomarkers for cancer. As one of the most exciting emerging technologies, protein array provides a versatile and robust platform in cancer proteomics research because it shows tremendous advantages of miniaturized features, high throughput, and sensitive detections in last decades. Here, we will present a relatively complete picture on the characteristics and advance of different types of protein arrays in application for biomarker discovery in cancer, and give the future perspectives in this area of research.
Collapse
|
41
|
Macdonald IK, Parsy-Kowalska CB, Chapman CJ. Autoantibodies: Opportunities for Early Cancer Detection. Trends Cancer 2017; 3:198-213. [PMID: 28718432 DOI: 10.1016/j.trecan.2017.02.003] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 02/06/2017] [Accepted: 02/07/2017] [Indexed: 12/18/2022]
Abstract
Cancer cells can induce an immunological response resulting in the production of tumor-associated (TA) autoantibodies. These serum immunobiomarkers have been detected for a range of cancers at an early stage before the development of clinical symptoms. Their measurement is minimally invasive and cost effective using established technologies. TA autoantibodies are present in a clinically significant number of individuals and could supplement current screening modalities to aid early diagnosis of high-risk populations and assist the clinical management of patients. Here we review their production, discovery, and validation as biomarkers for cancer and their current and future potential as clinical tools.
Collapse
|
42
|
Shi L, Chevolot Y, Souteyrand E, Laurenceau E. Autoantibodies against heat shock proteins as biomarkers for the diagnosis and prognosis of cancer. Cancer Biomark 2017; 18:105-116. [DOI: 10.3233/cbm-160117] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
43
|
Rastogi A, Ali A, Tan SH, Banerjee S, Chen Y, Cullen J, Xavier CP, Mohamed AA, Ravindranath L, Srivastav J, Young D, Sesterhenn IA, Kagan J, Srivastava S, McLeod DG, Rosner IL, Petrovics G, Dobi A, Srivastava S, Srinivasan A. Autoantibodies against oncogenic ERG protein in prostate cancer: potential use in diagnosis and prognosis in a panel with C-MYC, AMACR and HERV-K Gag. Genes Cancer 2017; 7:394-413. [PMID: 28191285 PMCID: PMC5302040 DOI: 10.18632/genesandcancer.126] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Overdiagnosis and overtreatment of prostate cancer (CaP) is attributable to widespread reliance on PSA screening in the US. This has prompted us and others to search for improved biomarkers for CaP, to facilitate early detection and disease stratification. In this regard, autoantibodies (AAbs) against tumor antigens could serve as potential candidates for diagnosis and prognosis of CaP. Towards this, our goals were: i) To investigate whether AAbs against ERG oncoprotein (overexpressed in 25-50% of Caucasian American and African American CaP) are present in the sera of CaP patients; ii) To evaluate an AAb panel to enhance CaP detection. The results using an enzyme-linked immunosorbent assay (ELISA) showed that anti-ERG AAbs are present in a significantly higher proportion in the sera of CaP patients compared to healthy controls (p = 0.0001). Furthermore, a panel of AAbs against ERG, AMACR and human endogenous retrovirus-K Gag successfully differentiated CaP patient sera from healthy controls (AUC = 0.791). These results demonstrate for the first time that anti-ERG AAbs are present in the sera of CaP patients. In addition, the data also suggest that AAbs against ERG together with AMACR and HERV-K Gag may be a useful panel of biomarkers for diagnosis and prognosis of CaP.
Collapse
Affiliation(s)
- Anshu Rastogi
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Amina Ali
- Urology Service, Department of Surgery, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Shyh-Han Tan
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Sreedatta Banerjee
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Yongmei Chen
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Jennifer Cullen
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Charles P Xavier
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Ahmed A Mohamed
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Lakshmi Ravindranath
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Jigisha Srivastav
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Denise Young
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | | | - Jacob Kagan
- Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, USA
| | - Sudhir Srivastava
- Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, USA
| | - David G McLeod
- Urology Service, Department of Surgery, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Inger L Rosner
- Urology Service, Department of Surgery, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Gyorgy Petrovics
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Albert Dobi
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Shiv Srivastava
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Alagarsamy Srinivasan
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| |
Collapse
|
44
|
Nizri E, Greenman-Maaravi N, Bar-David S, Ben-Yehuda A, Weiner G, Lahat G, Klausner J. Analysis of histological and immunological parameters of metastatic lymph nodes from colon cancer patients reveals that T-helper 1 type immune response is associated with improved overall survival. Medicine (Baltimore) 2016; 95:e5340. [PMID: 27828856 PMCID: PMC5106062 DOI: 10.1097/md.0000000000005340] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Lymph node (LN) involvement in colonic carcinoma (CC) is a grave prognostic sign and mandates the addition of adjuvant treatment. However, in light of the histological variability and outcomes observed, we hypothesized that patients with LN metastases (LNM) comprise different subgroups.We retrospectively analyzed the histological sections of 82 patients with CC and LNM. We studied various histological parameters (such as tumor grade, desmoplasia, and preservation of LN architecture) as well as the prevalence of specific peritumoral immune cells (CD8, CD20, T-bet, and GATA-3). We correlated the histological and immunological data to patient outcome.Tumor grade was a significant prognostic factor even in patients with LNM. So was the number of LN involved (N1/N2 stage). From the morphological parameters tested (LN extracapsular invasion, desmoplasia in LN, LN architecture preservation, and mode of metastases distribution), none was found to be significantly associated with overall survival (OS). The mean OS of CD8 low patients was 66.6 ± 6.25 versus 71.4 ± 5.1 months for CD8 high patients (P = 0.79). However, T-helper (Th) 1 immune response skewing (measured by Th1/Th2 ratio >1) was significantly associated with improved OS. For patients with low ratio, the median OS was 35.5 ± 5 versus 83.5 months for patients with high Th1/Th2 ratio (P = 0.001).The histological presentation of LNM does not entail specific prognostic information. However, the finding of Th1 immune response in LN signifies a protective immune response. Future studies should be carried to verify this marker and develop a strategy that augments this immune response during subsequent adjuvant treatment.
Collapse
Affiliation(s)
- Eran Nizri
- Division of General Surgery, Tel Aviv Sourasky Medical Center
- Sackler Faculty of Medicine, Tel Aviv University
- Laboratory of Surgical Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv
- Correspondence: Eran Nizri, Division of General Surgery, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel (e-mail: )
| | | | - Shoshi Bar-David
- Laboratory of Surgical Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv
| | - Amir Ben-Yehuda
- Division of General Surgery, Tel Aviv Sourasky Medical Center
- Sackler Faculty of Medicine, Tel Aviv University
| | - Gilad Weiner
- Sackler Faculty of Medicine, Tel Aviv University
- Institute of Pathology, Tel Aviv Sourasky Medical Center
| | - Guy Lahat
- Division of General Surgery, Tel Aviv Sourasky Medical Center
- Sackler Faculty of Medicine, Tel Aviv University
- Laboratory of Surgical Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv
| | - Joseph Klausner
- Division of General Surgery, Tel Aviv Sourasky Medical Center
- Sackler Faculty of Medicine, Tel Aviv University
- Laboratory of Surgical Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv
- The Nikolas and Elizabeth Shlezak Cathedra for Experimental Surgery, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
45
|
Werner S, Chen H, Butt J, Michel A, Knebel P, Holleczek B, Zörnig I, Eichmüller SB, Jäger D, Pawlita M, Waterboer T, Brenner H. Evaluation of the diagnostic value of 64 simultaneously measured autoantibodies for early detection of gastric cancer. Sci Rep 2016; 6:25467. [PMID: 27140836 PMCID: PMC4853774 DOI: 10.1038/srep25467] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 04/18/2016] [Indexed: 02/08/2023] Open
Abstract
Autoantibodies against tumor-associated antigens (TAAs) have been suggested as biomarkers for early detection of gastric cancer. However, studies that systematically assess the diagnostic performance of a large number of autoantibodies are rare. Here, we used bead-based multiplex serology to simultaneously measure autoantibody responses against 64 candidate TAAs in serum samples from 329 gastric cancer patients, 321 healthy controls and 124 participants with other diseases of the upper digestive tract. At 98% specificity, sensitivities for the 64 tested autoantibodies ranged from 0–12% in the training set and a combination of autoantibodies against five TAAs (MAGEA4 + CTAG1 + TP53 + ERBB2_C + SDCCAG8) was able to detect 32% of the gastric cancer patients at a specificity of 87% in the validation set. Sensitivities for early and late stage gastric cancers were similar, while chronic atrophic gastritis, a precursor lesion of gastric cancer, was not detectable. However, the 5-marker combination also detected 26% of the esophageal cancer patients. In conclusion, the tested autoantibodies and combinations alone did not reach sufficient sensitivity for gastric cancer screening. Nevertheless, some autoantibodies, such as anti-MAGEA4, anti-CTAG1 or anti-TP53 and their combinations could possibly contribute to the development of cancer early detection tests (not necessarily restricted to gastric cancer) when being combined with other markers.
Collapse
Affiliation(s)
- Simone Werner
- Division of Clinical Epidemiology and Aging Research, DKFZ, Heidelberg, Germany
| | - Hongda Chen
- Division of Clinical Epidemiology and Aging Research, DKFZ, Heidelberg, Germany
| | - Julia Butt
- Division of Molecular Diagnostics of Oncogenic Infections, DKFZ, Heidelberg, Germany
| | - Angelika Michel
- Division of Molecular Diagnostics of Oncogenic Infections, DKFZ, Heidelberg, Germany
| | - Phillip Knebel
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | | | - Inka Zörnig
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) and Heidelberg University Hospital, Heidelberg, Germany
| | - Stefan B Eichmüller
- GMP &T cell Therapy Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dirk Jäger
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) and Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Pawlita
- Division of Molecular Diagnostics of Oncogenic Infections, DKFZ, Heidelberg, Germany
| | - Tim Waterboer
- Division of Molecular Diagnostics of Oncogenic Infections, DKFZ, Heidelberg, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, DKFZ, Heidelberg, Germany.,Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
46
|
Mock A, Warta R, Geisenberger C, Bischoff R, Schulte A, Lamszus K, Stadler V, Felgenhauer T, Schichor C, Schwartz C, Matschke J, Jungk C, Ahmadi R, Sahm F, Capper D, Glass R, Tonn JC, Westphal M, von Deimling A, Unterberg A, Bermejo JL, Herold-Mende C. Printed peptide arrays identify prognostic TNC serumantibodies in glioblastoma patients. Oncotarget 2016; 6:13579-90. [PMID: 25944688 PMCID: PMC4537035 DOI: 10.18632/oncotarget.3791] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 03/18/2015] [Indexed: 01/11/2023] Open
Abstract
Liquid biopsies come of age offering unexploited potential to monitor and react to tumor evolution. We developed a cost-effective assay to non-invasively determine the immune status of glioblastoma (GBM) patients. Employing newly developed printed peptide microarrays we assessed the B-cell response against tumor-associated antigens (TAAs) in 214 patients. Firstly, sera of long-term (36+ months, LTS, n=10) and short-term (6-10 months, STS, n=14) surviving patients were screened for prognostic antibodies against 1745 13-mer peptides covering known TAAs (TNC, EGFR, GLEA2, PHF3, FABP5, MAGEA3). Next, survival associations were investigated in two retrospective independent multicenter validation sets (n=61, n=129, all IDH1-wildtype). Reliability of measurements was tested using a second array technology (spotted arrays). LTS/STS screening analyses identified 106 differential antibody responses. Evaluating the Top30 peptides in validation set 1 revealed three prognostic peptides. Prediction of TNC peptide VCEDGFTGPDCAE was confirmed in a second set (p=0.043, HR=0.66 [0.44-0.99]) and was unrelated to TNC protein expression. Median signals of printed arrays correlated with pre-synthesized spotted microarrays (p<0.0002, R=0.33). Multiple survival analysis revealed independence of age, gender, KPI and MGMT status. We present a novel peptide microarray immune assay that identified increased anti-TNC VCEDGFTGPDCAE serum antibody titer as a promising non-invasive biomarker for prolonged survival.
Collapse
Affiliation(s)
- Andreas Mock
- Department of Neurosurgery, Experimental Neurosurgery, University of Heidelberg, Heidelberg, Germany
| | - Rolf Warta
- Department of Neurosurgery, Experimental Neurosurgery, University of Heidelberg, Heidelberg, Germany
| | - Christoph Geisenberger
- Department of Neurosurgery, Experimental Neurosurgery, University of Heidelberg, Heidelberg, Germany
| | - Ralf Bischoff
- PEPperPRINT GmbH, Heidelberg, Germany.,Division of Functional Genome Analysis, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Alexander Schulte
- Department of Neurosurgery, Laboratory for Brain Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katrin Lamszus
- Department of Neurosurgery, Laboratory for Brain Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | - Christian Schichor
- Department of Neurosurgery, Klinikum Grosshadern, Ludwigs-Maximilians-University, Munich, Germany
| | - Christoph Schwartz
- Department of Neurosurgery, Klinikum Grosshadern, Ludwigs-Maximilians-University, Munich, Germany
| | - Jakob Matschke
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christine Jungk
- Department of Neurosurgery, Experimental Neurosurgery, University of Heidelberg, Heidelberg, Germany
| | - Rezvan Ahmadi
- Department of Neurosurgery, Experimental Neurosurgery, University of Heidelberg, Heidelberg, Germany
| | - Felix Sahm
- Department of Neuropathology, Institute of Pathology, Heidelberg, Germany.,Clinical Cooperation Unit Neuropathology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - David Capper
- Department of Neuropathology, Institute of Pathology, Heidelberg, Germany.,Clinical Cooperation Unit Neuropathology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rainer Glass
- Department of Neurosurgery, Klinikum Grosshadern, Ludwigs-Maximilians-University, Munich, Germany
| | - Jörg-Christian Tonn
- Department of Neurosurgery, Klinikum Grosshadern, Ludwigs-Maximilians-University, Munich, Germany
| | - Manfred Westphal
- Department of Neurosurgery, Laboratory for Brain Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas von Deimling
- Department of Neuropathology, Institute of Pathology, Heidelberg, Germany.,Clinical Cooperation Unit Neuropathology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andreas Unterberg
- Department of Neurosurgery, Experimental Neurosurgery, University of Heidelberg, Heidelberg, Germany
| | - Justo Lorenzo Bermejo
- Institute of Medical Biometry and Informatics, University of Heidelberg, Heidelberg, Germany.,Research Group Molecular Genetics of Breast Cancer, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Christel Herold-Mende
- Department of Neurosurgery, Experimental Neurosurgery, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
47
|
In Vitro Validation of Survivin as Target Tumor-associated Antigen for Immunotherapy in Uterine Cancer. J Immunother 2016; 38:239-49. [PMID: 26049547 DOI: 10.1097/cji.0000000000000085] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Survivin is an antiapoptotic protein, not expressed in terminally differentiated adult tissues, yet overexpressed in several tumors. Therefore, it is an interesting target for immunotherapeutic strategies. In addition to specific overexpression in tumors, tumor survival is mediated by survivin and hence, tumor survival can be tackled by targeting survivin. Survivin expression in uterine cancer was validated by quantitative real-time polymerase chain reaction and immunohistochemistry. In addition, we evaluated survivin immunogenicity by analyzing spontaneous B-cell and T-cell responses in patients. Survivin as a protein was expressed in only a minority of normal tissues, whereas it was being expressed in all of the currently analyzed uterine cancers, both endometrial carcinoma (n = 52) and uterine sarcoma (n = 52). Survivin RNA transcripts were overexpressed in more aggressive tumors and survivin protein was overexpressed in recurrent endometrial tumors compared with primary tumors. Spontaneous T-cell responses were seen in 10/39 endometrial cancer patients and 3/16 uterine sarcoma patients. In normal controls, T-cell responses were found only in 1 donor (n = 21). Although increased antibody titers were found in more aggressive and far-advanced tumors, no differences in B-cell responses were seen. Overall, when compared with normal controls, a B-cell response was only measured in 1/41 uterine sarcoma patients. In conclusion, we currently validated the presence of survivin in uterine cancer. In addition, spontaneous T-cell responses were found in 23.6% of the total patient population. These data indicate that a survivin-specific immune response may be induced spontaneously in patients, further fortifying the eligibility of survivin as an immunotherapeutic target.
Collapse
|
48
|
Shi L, Gehin T, Chevolot Y, Souteyrand E, Mangé A, Solassol J, Laurenceau E. Anti-heat shock protein autoantibody profiling in breast cancer using customized protein microarray. Anal Bioanal Chem 2015; 408:1497-506. [DOI: 10.1007/s00216-015-9257-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 11/30/2015] [Accepted: 12/07/2015] [Indexed: 10/22/2022]
|
49
|
Jiang T, Xie P, Liu H. Circulating Anti-Matrix Metalloproteinase-7 Antibodies May Be a Potential Biomarker for Oral Squamous Cell Carcinoma. J Oral Maxillofac Surg 2015; 74:650-7. [PMID: 26454036 DOI: 10.1016/j.joms.2015.09.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 09/13/2015] [Accepted: 09/14/2015] [Indexed: 01/18/2023]
Abstract
PURPOSE The present study was conducted to evaluate the diagnostic and prognostic values of serum autoantibody against matrix metalloproteinase-7 (MMP-7) in patients with oral squamous cell carcinoma (OSCC). MATERIALS AND METHODS Anti-MMP-7 antibodies were measured in sera from 204 patients with OSCC and 212 normal controls using enzyme-linked immunosorbent assay, and clinicopathologic characteristics were correlated. Prognostic consequence was assessed with Kaplan-Meier curve and log-rank tests using Cox proportional hazard models. To check whether anti-MMP-7 antibody was related to tumor associated antigen, real-time polymerase chain reaction and western blot were used to measure MMP-7 mRNA and protein expression in tumor tissues from all 204 patients with OSCC. RESULTS Serum anti-MMP-7 antibody was higher in patients with OSCC (P < .05), and those with poorly differentiated tumors had more anti-MMP-7 antibody than those with well to moderate tumor differentiation (P < .01, P < .01, respectively). Patients with OSCC at late TNM stages (III, IV) and lymph node metastases had relatively higher serum anti-MMP-7 antibody levels than those with earlier stages (I, II) and those who lacked lymph node metastases (P < .05 for the 2 comparisons). OSCC prediction sensitivity as measured by receiver operating characteristics analysis was 0.485 and specificity was 0.896 (area under the curve, 0.761; 95% confidence interval, 0.716 to 0.806). Cox analysis showed that serum anti-MMP-7 antibody positivity independently predicted poor overall survival in patients with OSCC (hazard ratio, 1.82; 95% confidence interval, 1.07 to 4.61). MMP-7 mRNA and protein expression was increased in tumor tissues from patients with OSCC and high serum anti-MMP-7 antibody. CONCLUSION Serum anti-MMP-7 antibody might be a novel diagnostic and prognostic biomarker for OSCC.
Collapse
Affiliation(s)
- Tao Jiang
- Professor, Department of Stomatology, Chinese PLA General Hospital, Beijing, China; Department of General Dentistry, Jinan Stomatological Hospital, Jinan, Shandong, China
| | - Pengfeng Xie
- Resident, Department of Oral and Maxillofacial Surgery, Jinan Stomatological Hospital, Jinan, Shandong, China
| | - Hongchen Liu
- Department Head, Department of Stomatology, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
50
|
Al-Shammari AM, Alshami MA, Umran MA, Almukhtar AA, Yaseen NY, Raad K, Hussien AA. Establishment and characterization of a receptor-negative, hormone-nonresponsive breast cancer cell line from an Iraqi patient. BREAST CANCER (DOVE MEDICAL PRESS) 2015; 7:223-30. [PMID: 26300657 PMCID: PMC4536763 DOI: 10.2147/bctt.s74509] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
A new breast cancer cell line (AMJ13) has been established from an Iraqi breast cancer patient. It is considered unique because it is the first for an Iraqi population, and is expected to be a useful tool in breast cancer research. The AMJ13 cell line was established from the primary tumor of a 70-year-old Iraqi woman with a histological diagnosis of infiltrating ductal carcinoma. The cells were morphologically characterized by light and scanning electron microscopy, and found to be elongated multipolar epithelial-like cells with a population doubling time of 22 hours. The anchorage-independent growth ability test showed that the cells were able to grow in semisolid agarose, confirming their transformed nature. Cytogenetic study of these cells showed chromosomal aberrations with many structural and numerical abnormalities, producing chromosomes of unknown origin called marker chromosomes. Immunocytochemistry showed that the estrogen receptor and the progesterone receptor were not expressed, and a weak positive result was found for HER2/neu gene expression. AMJ13 cells were positive for BRCA1 and BRCA2, as well as for vimentin. This cell line should be useful when testing new therapies for breast cancer in the Middle East.
Collapse
Affiliation(s)
- Ahmed Majeed Al-Shammari
- Experimental Therapy Department, Iraqi Center for Cancer and Medical Genetic Research, Mustansiriya University, Baghdad, Iraq
| | - Mortadha A Alshami
- Biotechnology Department, Collage of Science, Baghdad University, Baghdad, Iraq
| | | | - Asmaa Amer Almukhtar
- Medical Genetics Department, Iraqi Center for Cancer and Medical Genetic Research, Mustansiriya University, Baghdad, Iraq
| | - Nahi Y Yaseen
- Experimental Therapy Department, Iraqi Center for Cancer and Medical Genetic Research, Mustansiriya University, Baghdad, Iraq
| | - Khansaa Raad
- Experimental Therapy Department, Iraqi Center for Cancer and Medical Genetic Research, Mustansiriya University, Baghdad, Iraq
| | - Ayman A Hussien
- Experimental Therapy Department, Iraqi Center for Cancer and Medical Genetic Research, Mustansiriya University, Baghdad, Iraq
| |
Collapse
|