1
|
Allen J, Healey G, Macdonald I. Lung cancer associated autoantibody responses are detectable years before clinical presentation. PLoS One 2025; 20:e0315220. [PMID: 40067807 PMCID: PMC11896025 DOI: 10.1371/journal.pone.0315220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 11/21/2024] [Indexed: 03/15/2025] Open
Abstract
The EarlyCDT-Lung® test detects elevated levels of tumour-associated autoantibodies generated in response to immune recognition of cancerous cells, and these autoantibodies have previously been shown to precede clinical presentation of lung cancer. Using a longitudinal cohort from the United Kingdom Collaborative Trial of Ovarian Cancer Screening (UKCTOCS) study, we have established that elevated autoantibodies can be detected an average of four years in advance of clinical presentation, and in some cases up to 8 years prior to clinical presentation. This is the first study to establish pre-diagnostic elevation of autoantibodies using samples from a longitudinal prospective clinical trial using a clinically validated and commercially available biomarker panel.
Collapse
Affiliation(s)
- Jared Allen
- Division of Cancer and Stem Cells, University of Nottingham, Nottingham, United Kingdom
- Freenome Ltd., Nottingham, United Kingdom
| | | | | |
Collapse
|
2
|
Liu Q, Su J, Chen J, Yang S, Huang Y, Tang R, Jiang Z, Huang S. Bioinspired rational spatial-arrangement of antigens enables the accurate and rapid detection of anti-p53 autoantibody. Mikrochim Acta 2025; 192:123. [PMID: 39890668 DOI: 10.1007/s00604-025-06970-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 01/09/2025] [Indexed: 02/03/2025]
Abstract
A highly sensitive antibody detection strategy is presented that leverages the rational spatial arrangement of antigens at the sensing interface. Specifically, we employed rigid benzene ring-based coupling agents, carefully controlling their density and orientation on the biosensing interface to establish a well-defined spatial arrangement of receptor molecules, thereby enhancing antibody binding efficiency. Additionally, we utilized Au-decorated MoS2 nanosheets as an effective electrode modification, which also function as contact points for regulating the coupling agents. By optimizing both the electrode materials and the spatial arrangement of receptor molecules, our strategy enabled the precise and rapid detection of anti-p53 autoantibodies (anti-p53aAbs) in spiked plasma samples, achieving a broad linear range from 0.05 to 10 ng/mL and a low detection limit of 16.67 pg/mL, surpassing the performance of most existing methods. Notably, we introduce a biomimetic strategy for the spatial arrangement of antigens, inspired by the bionic recognition mechanism. This design effectively reduces steric hindrance between antibody molecules, enhances binding efficiency, and provides a novel approach for the rapid and sensitive detection of macromolecules, such as antibodies.
Collapse
Affiliation(s)
- Qiwen Liu
- Guangzhou Municipal and Guangdong Provincial KeyLaboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jianfen Su
- The Affiliated Panyu Central Hospital, Guangzhou Medical University, Guangzhou, 510000, China
| | - Jiamei Chen
- Guangzhou Municipal and Guangdong Provincial KeyLaboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Shuo Yang
- Guangzhou Municipal and Guangdong Provincial KeyLaboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yang Huang
- Guangzhou Municipal and Guangdong Provincial KeyLaboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Rentao Tang
- Guangzhou Municipal and Guangdong Provincial KeyLaboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Zhengjin Jiang
- Institute of Pharmaceutical Analysis, College of Pharmacy/State Key Laboratory of Bioactive Molecules and Druggability Assessment/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou, 510632, China.
| | - Shengfeng Huang
- Guangzhou Municipal and Guangdong Provincial KeyLaboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
3
|
Kim SY, Park YS, Kim IA, Kim HJ, Lee KY. Assessing the 9G Technology Blood Test for Predicting Lung Cancer in Patients with CT-Detected Lung Nodules: A Multicenter Clinical Trial. Cancers (Basel) 2024; 16:3737. [PMID: 39594693 PMCID: PMC11593157 DOI: 10.3390/cancers16223737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 10/31/2024] [Indexed: 11/28/2024] Open
Abstract
Background and Objectives: Lung nodules detected by chest computed tomography (CT) often require invasive biopsies for definitive diagnosis, leading to unnecessary procedures for benign lesions. A blood-based biomarker test that predicts lung cancer risk in CT-detected nodules could help stratify patients and direct invasive diagnostics toward high-risk individuals. Methods: In this multicenter, single-blinded clinical trial, we evaluated a test measuring plasma levels of p53, anti-p53 autoantibodies, CYFRA 21-1, and anti-CYFRA 21-1 autoantibodies in patients with CT-detected lung nodules. Sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV) were calculated, and subgroup analyses by gender, age, and smoking status were performed. A total of 1132 patients who had CT-detected lung nodules, including 885 lung cancer cases and 247 benign lesions, were enrolled from two academic hospitals in South Korea. Results: The test demonstrated a sensitivity of 78.4% (95% CI: 75.7-81.1) and specificity of 93.1% (95% CI: 90.0-96.3) in predicting lung cancer in CT-detected nodules. The PPV was 97.6%, and the NPV was 54.6%. Performance was consistent across gender (sensitivity 79.3% in men and 76.8% in women) and age groups, with a specificity of 93.4% in men and 92.7% in women. Stage I lung cancer was detected with a sensitivity of 80.6%. Conclusions: The Lung Cancer test based on 9G technology presented here offers a non-invasive method for stratifying lung cancer risk in patients with CT-detected nodules. Its integration into clinical practice could reduce unnecessary interventions and foster earlier detection.
Collapse
Affiliation(s)
- So Yeon Kim
- Division of Pulmonary and Critical Care Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea; (S.Y.K.); (Y.S.P.)
| | - Young Sik Park
- Division of Pulmonary and Critical Care Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea; (S.Y.K.); (Y.S.P.)
| | - In Ae Kim
- Precision Medicine Lung Cancer Center, Konkuk University Medical Center, Seoul 05029, Republic of Korea; (I.A.K.); (H.J.K.)
| | - Hee Joung Kim
- Precision Medicine Lung Cancer Center, Konkuk University Medical Center, Seoul 05029, Republic of Korea; (I.A.K.); (H.J.K.)
| | - Kye Young Lee
- Precision Medicine Lung Cancer Center, Konkuk University Medical Center, Seoul 05029, Republic of Korea; (I.A.K.); (H.J.K.)
| |
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
|
Abstract
Screening with low-dose computed tomography has been shown to decrease lung cancer mortality. However, the issues of low detection rates and false positive results remain, highlighting the need for adjunctive tools in lung cancer screening. To this end, researchers have investigated easily applicable, minimally invasive tests with high validity. We herein review some of the more promising novel markers utilizing plasma, sputum, and airway samples.
Collapse
Affiliation(s)
- Ju Ae Park
- Department of General Surgery, Inova Fairfax Medical Campus, 3300 Gallows Road, Falls Church, VA 22042, USA
| | - Kei Suzuki
- Inova Thoracic Surgery, Schar Cancer Institute, 8081 Innovation Park Drive, Fairfax, VA 22031, USA.
| |
Collapse
|
6
|
Yoon HS, Zheng W, Cai H, Wu J, Shidal C, Wang J, Shu XO, Waterboer T, Blot WJ, Cai Q. Pre-diagnostic circulating p53 autoantibodies and subsequent lung cancer risk in low-income African and European Americans. Cancer Epidemiol 2022; 81:102288. [PMID: 36332502 PMCID: PMC11296379 DOI: 10.1016/j.canep.2022.102288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 10/13/2022] [Accepted: 10/25/2022] [Indexed: 11/08/2022]
Abstract
INTRODUCTION Mutations of the TP53 gene lead to the production of autoantibodies against p53, a major tumor suppressor protein. Although studies have indicated the association of p53 autoantibodies with human cancers, epidemiologic evidence on lung cancer is still lacking. METHODS In this nested case-control study conducted within the Southern Community Cohort Study, we investigated the association of circulating p53 autoantibodies with the subsequent risk of developing lung cancer. Using blood samples collected prior to any cancer diagnosis from 295 cases and their individually matched controls, seroreactivity to p53 was assessed by fluorescent bead-based multiplex serology. Conditional logistic regression models were used to estimate odds ratios (OR) and 95 % confidence intervals (CI) for lung cancer risk associated with p53 autoantibodies. RESULTS After adjustment for potential confounders, p53 seropositivity was significantly associated with an increased risk of lung cancer (OR=2.98, 95 % CI: 1.10-8.06) among African Americans, but not among European Americans (OR=1.21, 95 % CI: 0.24-6.15). The positive associations were restricted to men (OR=4.59, 95 % CI: 1.30-16.16) and participants with a short interval (≤ 4 years) from blood collection to diagnosis (OR=4.30, 95 % CI: 1.33-13.89). CONCLUSION Our findings add to the evidence supporting p53 autoantibodies as a biomarker of lung cancer.
Collapse
Affiliation(s)
- Hyung-Suk Yoon
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, 37203, USA
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, 37203, USA
| | - Hui Cai
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, 37203, USA
| | - Jie Wu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, 37203, USA
| | - Chris Shidal
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, 37203, USA
| | - Jifeng Wang
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, 37203, USA
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, 37203, USA
| | - Tim Waterboer
- Division of Infections and Cancer Epidemiology, Infection, Inflammation and Cancer Program, German Cancer Research Center (DFKZ), Heidelberg, Germany
| | - William J Blot
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, 37203, USA
| | - Qiuyin Cai
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, 37203, USA.
| |
Collapse
|
7
|
He T, Wu Z, Xia P, Wang W, Sun H, Yu L, Lv W, Hu J. The combination of a seven-autoantibody panel with computed tomography scanning can enhance the diagnostic efficiency of non-small cell lung cancer. Front Oncol 2022; 12:1047019. [DOI: 10.3389/fonc.2022.1047019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 11/04/2022] [Indexed: 12/05/2022] Open
Abstract
BackgroundNon-small cell lung cancer (NSCLC) is still of concern in differentiating it from benign disease. This study aims to validate the diagnostic efficacy of a novel seven-autoantibody (7-AAB) panel for the diagnosis of NSCLC.MethodsWe retrospectively enrolled 2650 patients who underwent both the 7-AAB panel test and CT scanning. We compared the sensitivity, specificity, and PPV of 7-AAB, CT, and PET-CT in the diagnosis of NSCLC in different subgroups. Then, we established a nomogram based on CT image features and the 7-AAB panel to further improve diagnostic efficiency. Moreover, we compared the pathological and molecular results of NSCLC patients in the 7-AABs positive group and the negative group to verify the prognostic value of the 7-AAB panel.ResultsThe strategy of a “both-positive rule” combination of 7-AABs and CT had a specificity of 95.4% and a positive predictive value (PPV) of 95.8%, significantly higher than those of CT or PET-CT used alone (P<0.05). The nomogram we established has passed the calibration test (P=0.987>0.05) with an AUC of 0.791. Interestingly, it was found that the 7-AABs positive group was associated with higher proportion of EGFR mutations (P<0.001), lower pathological differentiation degrees (P=0.018), more advanced pathological stages (P=0.040) and higher Ki-67 indexes (P=0.011) in patients with adenocarcinoma.ConclusionThis study shows that combination of a 7-AAB panel with CT has can significantly enhance the diagnostic efficiency of lung cancer. Moreover, the 7-AAB panel also has potential prognostic value and has reference significance for the formulation of the treatment plan.
Collapse
|
8
|
Detection and Quantification of Tp53 and p53-Anti-p53 Autoantibody Immune Complex: Promising Biomarkers in Early Stage Lung Cancer Diagnosis. BIOSENSORS 2022; 12:bios12020127. [PMID: 35200387 PMCID: PMC8870326 DOI: 10.3390/bios12020127] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/03/2022] [Accepted: 02/14/2022] [Indexed: 11/29/2022]
Abstract
Lung cancer is a leading cause of death worldwide, claiming nearly 1.80 million lives in 2020. Screening with low-dose computed tomography (LDCT) reduces lung cancer mortality by about 20% compared to standard chest X-rays among current or heavy smokers. However, several reports indicate that LDCT has a high false-positive rate. In this regard, methods based on biomarker detection offer excellent potential for developing noninvasive cancer diagnostic tests to complement LDCT for detecting stage 0∼IV lung cancers. Herein, we have developed a method for detecting and quantifying a p53-anti-p53 autoantibody complex and the total p53 antigen (wild and mutant). The LOD for detecting Tp53 and PIC were 7.41 pg/mL and 5.74 pg/mL, respectively. The detection ranges for both biomarkers were 0–7500 pg/mL. The known interfering agents in immunoassays such as biotin, bilirubin, intra-lipid, and hemoglobin did not detect Tp53 and PIC, even at levels that were several folds higher levels than their normal levels. Furthermore, the present study provides a unique report on this preliminary investigation using the PIC/Tp53 ratio to detect stage I–IV lung cancers. The presented method detects lung cancers with 81.6% sensitivity and 93.3% specificity. These results indicate that the presented method has high applicability for the identification of lung cancer patients from the healthy population.
Collapse
|
9
|
Xiao K, Ma X, Wang Y, Zhu C, Guo L, Lu R. Diagnostic value of serum tumor-associated autoantibodies in esophageal cancer. Biomark Med 2021; 15:1333-1343. [PMID: 34541870 DOI: 10.2217/bmm-2021-0351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To explore the application value of serum autoantibodies in the early diagnosis of esophageal cancer. Materials & methods: A total of 130 patients with esophageal cancer and 110 controls were included and tested by ELISA. Results: According to the receiver operating characteristic curve, total sensitivity is 83.08%, total specificity is 72.73%. A nomogram was established based on the positive judgment standard, the area under the receiver operating characteristic curve was calculated to be 0.880 after verification with the calibration curve. A 2-week follow-up analysis found compared with the preoperative control, the postoperative model integral value will significantly decrease. Conclusion: The combination of serum autoantibody groups has certain clinical application value in the early diagnosis of esophageal cancer and can be used as an auxiliary index for early diagnosis.
Collapse
Affiliation(s)
- Kangjia Xiao
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032, China
| | - Xiaolu Ma
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032, China
| | - Yanchun Wang
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032, China
| | - Cheng Zhu
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032, China
| | - Lin Guo
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai, 200032, China
| | - Renquan Lu
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai, 200032, China
| |
Collapse
|
10
|
Bareke H, Juanes-Velasco P, Landeira-Viñuela A, Hernandez AP, Cruz JJ, Bellido L, Fonseca E, Niebla-Cárdenas A, Montalvillo E, Góngora R, Fuentes M. Autoimmune Responses in Oncology: Causes and Significance. Int J Mol Sci 2021; 22:ijms22158030. [PMID: 34360795 PMCID: PMC8347170 DOI: 10.3390/ijms22158030] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/19/2021] [Accepted: 07/23/2021] [Indexed: 12/20/2022] Open
Abstract
Specific anti-tumor immune responses have proven to be pivotal in shaping tumorigenesis and tumor progression in solid cancers. These responses can also be of an autoimmune nature, and autoantibodies can sometimes be present even before the onset of clinically overt disease. Autoantibodies can be generated due to mutated gene products, aberrant expression and post-transcriptional modification of proteins, a pro-immunogenic milieu, anti-cancer treatments, cross-reactivity of tumor-specific lymphocytes, epitope spreading, and microbiota-related and genetic factors. Understanding these responses has implications for both basic and clinical immunology. Autoantibodies in solid cancers can be used for early detection of cancer as well as for biomarkers of prognosis and treatment response. High-throughput techniques such as protein microarrays make parallel detection of multiple autoantibodies for increased specificity and sensitivity feasible, affordable, and quick. Cancer immunotherapy has revolutionized cancer treatments and has made a considerable impact on reducing cancer-associated morbidity and mortality. However, immunotherapeutic interventions such as immune checkpoint inhibition can induce immune-related toxicities, which can even be life-threatening. Uncovering the reasons for treatment-induced autoimmunity can lead to fine-tuning of cancer immunotherapy approaches to evade toxic events while inducing an effective anti-tumor immune response.
Collapse
Affiliation(s)
- Halin Bareke
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Institute of Health Sciences, Marmara University, Istanbul 34722, Turkey;
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (P.J.-V.); (A.L.-V.); (A.-P.H.); (E.M.); (R.G.)
| | - Pablo Juanes-Velasco
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (P.J.-V.); (A.L.-V.); (A.-P.H.); (E.M.); (R.G.)
| | - Alicia Landeira-Viñuela
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (P.J.-V.); (A.L.-V.); (A.-P.H.); (E.M.); (R.G.)
| | - Angela-Patricia Hernandez
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (P.J.-V.); (A.L.-V.); (A.-P.H.); (E.M.); (R.G.)
| | - Juan Jesús Cruz
- Medical Oncology Service, Hospital Universitario de Salamanca-IBSAL, 37007 Salamanca, Spain; (J.J.C.); (L.B.); (E.F.)
| | - Lorena Bellido
- Medical Oncology Service, Hospital Universitario de Salamanca-IBSAL, 37007 Salamanca, Spain; (J.J.C.); (L.B.); (E.F.)
| | - Emilio Fonseca
- Medical Oncology Service, Hospital Universitario de Salamanca-IBSAL, 37007 Salamanca, Spain; (J.J.C.); (L.B.); (E.F.)
| | - Alfonssina Niebla-Cárdenas
- Department of Nursing and Physiotherapy, Faculty of Nursing and Physiotherapy, University of Salamanca, 37007 Salamanca, Spain;
| | - Enrique Montalvillo
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (P.J.-V.); (A.L.-V.); (A.-P.H.); (E.M.); (R.G.)
| | - Rafael Góngora
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (P.J.-V.); (A.L.-V.); (A.-P.H.); (E.M.); (R.G.)
| | - Manuel Fuentes
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (P.J.-V.); (A.L.-V.); (A.-P.H.); (E.M.); (R.G.)
- Proteomics Unit, Cancer Research Center (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain
- Correspondence: ; Tel.: +34-923-294-811
| |
Collapse
|
11
|
Visaggi P, Barberio B, Ghisa M, Ribolsi M, Savarino V, Fassan M, Valmasoni M, Marchi S, de Bortoli N, Savarino E. Modern Diagnosis of Early Esophageal Cancer: From Blood Biomarkers to Advanced Endoscopy and Artificial Intelligence. Cancers (Basel) 2021; 13:3162. [PMID: 34202763 PMCID: PMC8268190 DOI: 10.3390/cancers13133162] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 12/16/2022] Open
Abstract
Esophageal cancer (EC) is the seventh most common cancer and the sixth cause of cancer death worldwide. Histologically, esophageal squamous cell carcinoma (ESCC) and esophageal adenocarcinoma (EAC) account for up to 90% and 20% of all ECs, respectively. Clinical symptoms such as dysphagia, odynophagia, and bolus impaction occur late in the natural history of the disease, and the diagnosis is often delayed. The prognosis of ESCC and EAC is poor in advanced stages, being survival rates less than 20% at five years. However, when the diagnosis is achieved early, curative treatment is possible, and survival exceeds 80%. For these reasons, mass screening strategies for EC are highly desirable, and several options are currently under investigation. Blood biomarkers offer an inexpensive, non-invasive screening strategy for cancers, and novel technologies have allowed the identification of candidate markers for EC. The esophagus is easily accessible via endoscopy, and endoscopic imaging represents the gold standard for cancer surveillance. However, lesion recognition during endoscopic procedures is hampered by interobserver variability. To fill this gap, artificial intelligence (AI) has recently been explored and provided encouraging results. In this review, we provide a summary of currently available options to achieve early diagnosis of EC, focusing on blood biomarkers, advanced endoscopy, and AI.
Collapse
Affiliation(s)
- Pierfrancesco Visaggi
- Gastroenterology Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124 Pisa, Italy; (P.V.); (S.M.); (N.d.B.)
| | - Brigida Barberio
- Division of Gastroenterology, Department of Surgery, Oncology and Gastroenterology, University of Padua, 35121 Padua, Italy; (B.B.); (M.G.)
| | - Matteo Ghisa
- Division of Gastroenterology, Department of Surgery, Oncology and Gastroenterology, University of Padua, 35121 Padua, Italy; (B.B.); (M.G.)
| | - Mentore Ribolsi
- Department of Digestive Diseases, Campus Bio Medico University of Rome, 00128 Roma, Italy;
| | - Vincenzo Savarino
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, 16143 Genoa, Italy;
| | - Matteo Fassan
- Surgical Pathology & Cytopathology Unit, Department of Medicine (DIMED), University of Padua, 35121 Padua, Italy;
| | - Michele Valmasoni
- Department of Surgical, Oncological and Gastroenterological Sciences, Center for Esophageal Disease, University of Padova, 35124 Padova, Italy;
| | - Santino Marchi
- Gastroenterology Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124 Pisa, Italy; (P.V.); (S.M.); (N.d.B.)
| | - Nicola de Bortoli
- Gastroenterology Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124 Pisa, Italy; (P.V.); (S.M.); (N.d.B.)
| | - Edoardo Savarino
- Division of Gastroenterology, Department of Surgery, Oncology and Gastroenterology, University of Padua, 35121 Padua, Italy; (B.B.); (M.G.)
| |
Collapse
|
12
|
Luo B, Mao G, Ma H, Chen S. The role of seven autoantibodies in lung cancer diagnosis. J Thorac Dis 2021; 13:3660-3668. [PMID: 34277058 PMCID: PMC8264704 DOI: 10.21037/jtd-21-835] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 06/08/2021] [Indexed: 11/06/2022]
Abstract
Background To investigate the expression and diagnostic value of seven autoantibodies (P53, PGP9.5, SOX2, GAGE7, GBU4-5, MAGE, and CACE) in lung cancer patients. Methods A total of 370 patients were admitted to the Thoracic Surgery of the First Affiliated Hospital of Suzhou University from 2017 to 2019, including 305 patients with lung cancer and 65 patients with benign lesions. The concentrations of the seven autoantibodies were determined by enzyme linked immunosorbent assay (ELISA).The expression levels of each antibody were compared between the two groups, and the levels of each antibody between lung cancer patients with different pathological types were also compared. We aimed to analyze the diagnostic efficiency of single antibody detection combined with seven antibodies, and also to explore whether there were differences among the positive rates of each antibody in sex, age, smoking history, pathological classification, and clinical stages in the lung cancer group. Results The expression levels of seven autoantibodies in the lung cancer group were higher than those in the benign lesion group. In the lung cancer group, the expression levels of the seven autoantibodies did not vary statistically among different pathological types. The area under the curve of combined detection of the seven antibodies reached 0.735, and the Y-index reached 0.35, which was higher than that of single antibody detection. P53 exhibited the highest sensitivity and lowest specificity; meanwhile, PGP9.5, SOX2, GAGE7, GBU4-5, and MAGEA1 exhibited low sensitivity and high specificity. The sensitivity and specificity of the CAGE were approximately 60%, respectively. There was no statistical difference in the positive rate of each antibody in age, smoking history, and clinical stage. The positive rate of MAGEA1 and CAGE was statistically different in sex, and the positive rate of MAGEA1 was statistically different in pathological classification. Conclusions The seven autoantibodies of lung cancer can potentially be used as an auxiliary examination method for the early diagnosis of lung cancer.
Collapse
Affiliation(s)
- Bin Luo
- Department of Thoracic Surgery, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Guocai Mao
- Department of Thoracic Surgery, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Haitao Ma
- Department of Thoracic Surgery, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shaomu Chen
- Department of Thoracic Surgery, First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
13
|
Sullivan FM, Mair FS, Anderson W, Armory P, Briggs A, Chew C, Dorward A, Haughney J, Hogarth F, Kendrick D, Littleford R, McConnachie A, McCowan C, McMeekin N, Patel M, Rauchhaus P, Ritchie L, Robertson C, Robertson J, Robles-Zurita J, Sarvesvaran J, Sewell H, Sproule M, Taylor T, Tello A, Treweek S, Vedhara K, Schembri S. Earlier diagnosis of lung cancer in a randomised trial of an autoantibody blood test followed by imaging. Eur Respir J 2021; 57:2000670. [PMID: 32732334 PMCID: PMC7806972 DOI: 10.1183/13993003.00670-2020] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 07/09/2020] [Indexed: 12/18/2022]
Abstract
The EarlyCDT-Lung test is a high-specificity blood-based autoantibody biomarker that could contribute to predicting lung cancer risk. We report on the results of a phase IV biomarker evaluation of whether using the EarlyCDT-Lung test and any subsequent computed tomography (CT) scanning to identify those at high risk of lung cancer reduces the incidence of patients with stage III/IV/unspecified lung cancer at diagnosis compared with the standard clinical practice at the time the study began.The Early Diagnosis of Lung Cancer Scotland (ECLS) trial was a randomised controlled trial of 12 208 participants at risk of developing lung cancer in Scotland in the UK. The intervention arm received the EarlyCDT-Lung test and, if test-positive, low-dose CT scanning 6-monthly for up to 2 years. EarlyCDT-Lung test-negative and control arm participants received standard clinical care. Outcomes were assessed at 2 years post-randomisation using validated data on cancer occurrence, cancer staging, mortality and comorbidities.At 2 years, 127 lung cancers were detected in the study population (1.0%). In the intervention arm, 33 out of 56 (58.9%) lung cancers were diagnosed at stage III/IV compared with 52 out of 71 (73.2%) in the control arm. The hazard ratio for stage III/IV presentation was 0.64 (95% CI 0.41-0.99). There were nonsignificant differences in lung cancer and all-cause mortality after 2 years.ECLS compared EarlyCDT-Lung plus CT screening to standard clinical care (symptomatic presentation) and was not designed to assess the incremental contribution of the EarlyCDT-Lung test. The observation of a stage shift towards earlier-stage lung cancer diagnosis merits further investigations to evaluate whether the EarlyCDT-Lung test adds anything to the emerging standard of low-dose CT.
Collapse
Affiliation(s)
| | - Frances S Mair
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | | | - Pauline Armory
- Tayside Clinical Trials Unit, University of Dundee, Dundee, UK
| | - Andrew Briggs
- Dept of Health Services Research and Policy, London School of Hygiene and Tropical Medicine, London, UK
| | - Cindy Chew
- Radiology, NHS Lanarkshire, Bothwell, UK
| | - Alistair Dorward
- Respiratory Medicine, NHS Greater Glasgow and Clyde, Glasgow, UK
| | - John Haughney
- General Practice, NHS Greater Glasgow and Clyde, Glasgow, UK
| | - Fiona Hogarth
- Tayside Clinical Trials Unit, University of Dundee, Dundee, UK
| | - Denise Kendrick
- School of Medicine, University of Nottingham, Nottingham, UK
| | - Roberta Littleford
- Centre for Clinical Research, University of Queensland, Saint Lucia, Australia
| | - Alex McConnachie
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Colin McCowan
- School of Medicine, University of St Andrews, St Andrews, UK
| | - Nicola McMeekin
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Manish Patel
- Respiratory Medicine, NHS Lanarkshire, Bothwell, UK
| | - Petra Rauchhaus
- Tayside Clinical Trials Unit, University of Dundee, Dundee, UK
| | - Lewis Ritchie
- The Institute of Applied Health Sciences, University of Aberdeen, Aberdeen, UK
| | - Chris Robertson
- Dept of Mathematics and Statistics, University of Strathclyde, Glasgow, UK
| | - John Robertson
- School of Medicine, University of Nottingham, Nottingham, UK
| | | | | | - Herbert Sewell
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | | | | | - Agnes Tello
- School of Medicine, University of St Andrews, St Andrews, UK
| | - Shaun Treweek
- Health Services Research Unit, University of Aberdeen, Aberdeen, UK
| | - Kavita Vedhara
- School of Medicine, University of Nottingham, Nottingham, UK
| | | |
Collapse
|
14
|
Adeniyi OK, Ngqinambi A, Mashazi PN. Ultrasensitive detection of anti-p53 autoantibodies based on nanomagnetic capture and separation with fluorescent sensing nanobioprobe for signal amplification. Biosens Bioelectron 2020; 170:112640. [DOI: 10.1016/j.bios.2020.112640] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 09/11/2020] [Accepted: 09/17/2020] [Indexed: 12/28/2022]
|
15
|
Ushigome M, Shimada H, Nabeya Y, Shiratori F, Soda H, Takiguchi N, Hoshino I, Kuwajima A, Kaneko T, Funahashi K. Possible predictive significance of serum RalA autoantibodies on relapse-free survival in patients with colorectal cancer. Mol Clin Oncol 2020; 14:18. [PMID: 33363728 PMCID: PMC7725215 DOI: 10.3892/mco.2020.2180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 10/27/2020] [Indexed: 12/18/2022] Open
Abstract
RalA protein, a member of the Ras superfamily of small GTPases, is a tumor antigen that induces serum RalA antibodies (s-RalA-Abs). The present study explored the clinicopathological and prognostic significance of s-RalA-Abs in patients with colorectal cancer. Serum samples were obtained from 314 patients with colorectal cancer at stage 0/I (n=71), stage II (n=86), stage III (n=78), stage IV (n=64) and recurrence (n=15). Samples were analyzed for the presence of s-RalA-Abs using ELISA. The cutoff optical density value was fixed at 0.324 (mean of heathy controls + 3 standard deviations). The overall positive rate for serum anti-RalA antibodies was 14%. The presence of s-RalA-Abs was not significantly associated with clinicopathological characteristic factors. Additionally, the s-RalA-Abs(+) group demonstrated significantly poor relapse-free survival rates. The s-RalA-Abs (+)/carcinoembryonic antigen (CEA)(+) group exhibited the worst prognosis and s-RalA-Abs(+)/CEA(+) was an independent risk factor for poor relapse-free survival. Although the positive rate was not high, s-RalA-Abs may be a useful predictor of poor relapse-free survival in patients with colorectal cancer.
Collapse
Affiliation(s)
- Mitsunori Ushigome
- Department of Surgery, School of Medicine, Toho University, Ota-ku, Tokyo 143-8541, Japan
| | - Hideaki Shimada
- Department of Surgery, School of Medicine, Toho University, Ota-ku, Tokyo 143-8541, Japan
| | - Yoshihiro Nabeya
- Division of Gastroenterological Surgery, Chiba Cancer Center, Chuo-ku, Chiba 260-8717, Japan
| | - Fumiaki Shiratori
- Department of Surgery, School of Medicine, Toho University, Ota-ku, Tokyo 143-8541, Japan.,Division of Gastroenterological Surgery, Chiba Cancer Center, Chuo-ku, Chiba 260-8717, Japan
| | - Hiroaki Soda
- Division of Gastroenterological Surgery, Chiba Cancer Center, Chuo-ku, Chiba 260-8717, Japan
| | - Nobuhiro Takiguchi
- Division of Gastroenterological Surgery, Chiba Cancer Center, Chuo-ku, Chiba 260-8717, Japan
| | - Isamu Hoshino
- Division of Gastroenterological Surgery, Chiba Cancer Center, Chuo-ku, Chiba 260-8717, Japan
| | - Akiko Kuwajima
- Medical and Biological Laboratories Co., Ltd, Naka-ku, Nagoya 460-0008, Japan
| | - Tomoaki Kaneko
- Department of Surgery, School of Medicine, Toho University, Ota-ku, Tokyo 143-8541, Japan
| | - Kimihiko Funahashi
- Department of Surgery, School of Medicine, Toho University, Ota-ku, Tokyo 143-8541, Japan
| |
Collapse
|
16
|
Wang H, Zhang B, Li X, Zhou D, Li Y, Jia S, Qi S, Xu A, Zhao X, Wang J, Bai Z, Cao B, Li N, Dai M, Chen H, Huang J. Identification and Validation of Novel Serum Autoantibody Biomarkers for Early Detection of Colorectal Cancer and Advanced Adenoma. Front Oncol 2020; 10:1081. [PMID: 32793472 PMCID: PMC7387658 DOI: 10.3389/fonc.2020.01081] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 05/29/2020] [Indexed: 12/24/2022] Open
Abstract
Background: Colorectal cancer (CRC) comprises a large proportion of malignant tumors, and early detection of CRC is critical for effective treatment and optimal prognosis. We aimed to discover and validate serum autoantibodies for early detection of CRC. Methods: Combined with CRC-associated autoantibodies discovered by serological proteome and multiplex analyses, 26 predefined autoantibodies were evaluated in 315 samples (130 CRCs, 75 advanced adenomas, and 110 healthy controls) by protein microarray analysis. Autoantibodies with potential detection value were verified by enzyme-linked immunosorbent assays (ELISAs). Receiver operating characteristic (ROC) curve analysis was conducted to evaluate the accuracy of the biomarkers. Results: Four serum autoantibodies (ALDH1B1, UQCRC1, CTAG1, and CENPF) showed statistically different levels between patients with advanced neoplasm (CRC or advanced adenoma) and controls in microarray analysis, which were validated by ELISAs. Among the four biomarkers, the ALDH1B1 autoantibody showed the highest detection value with area under the curve (AUC) values of 0.70 and 0.74 to detect CRC and advanced adenoma with sensitivities of 75.68 and 62.31% and specificities of 63.06 and 73.87%, respectively. By combining the four biomarkers, the performance was improved with an AUC of 0.79 to detect CRC and advanced adenomas. Conclusion: The ALDH1B1 autoantibody has a good potential for early detection of CRC and advanced adenoma, and measuring serum autoantibodies against tumor-associated antigens may improve detection of early CRC.
Collapse
Affiliation(s)
- Hejing Wang
- Experimental Centre, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Bei Zhang
- Experimental Centre, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xiaojin Li
- Experimental Centre, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Donghu Zhou
- Experimental Centre, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yanmeng Li
- Experimental Centre, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Siyu Jia
- Experimental Centre, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Saiping Qi
- Experimental Centre, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Anjian Xu
- Experimental Centre, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xiaomu Zhao
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jin Wang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhigang Bai
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Bangwei Cao
- National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Ni Li
- Office of Cancer Screening, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Min Dai
- Office of Cancer Screening, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hongda Chen
- Office of Cancer Screening, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jian Huang
- Experimental Centre, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
17
|
Chu LY, Peng YH, Weng XF, Xie JJ, Xu YW. Blood-based biomarkers for early detection of esophageal squamous cell carcinoma. World J Gastroenterol 2020; 26:1708-1725. [PMID: 32351288 PMCID: PMC7183865 DOI: 10.3748/wjg.v26.i15.1708] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 03/13/2020] [Accepted: 03/19/2020] [Indexed: 02/06/2023] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is a common malignant tumor of the digestive system worldwide, especially in China. Due to the lack of effective early detection methods, ESCC patients often present at an advanced stage at the time of diagnosis, which seriously affects the prognosis of patients. At present, early detection of ESCC mainly depends on invasive and expensive endoscopy and histopathological biopsy. Therefore, there is an unmet need for a non-invasive method to detect ESCC in the early stages. With the emergence of a large class of non-invasive diagnostic tools, serum tumor markers have attracted much attention because of their potential for detection of early tumors. Therefore, the identification of serum tumor markers for early detection of ESCC is undoubtedly one of the most effective ways to achieve early diagnosis and treatment of ESCC. This article reviews the recent advances in the discovery of blood-based ESCC biomarkers, and discusses the origins, clinical applications, and technical challenges of clinical validation of various types of biomarkers.
Collapse
Affiliation(s)
- Ling-Yu Chu
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Yu-Hui Peng
- Department of Clinical Laboratory Medicine, the Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
- Precision Medicine Research Center, Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Xue-Fen Weng
- Department of Clinical Laboratory Medicine, the Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
- Precision Medicine Research Center, Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Jian-Jun Xie
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong Province, China
- Precision Medicine Research Center, Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Yi-Wei Xu
- Department of Clinical Laboratory Medicine, the Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
- Precision Medicine Research Center, Shantou University Medical College, Shantou 515041, Guangdong Province, China
| |
Collapse
|
18
|
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]
|
19
|
Yan Y, Sun N, Wang H, Kobayashi M, Ladd JJ, Long JP, Lo KC, Patel J, Sullivan E, Albert T, Goodman GE, Do KA, Hanash SM. Whole Genome-Derived Tiled Peptide Arrays Detect Prediagnostic Autoantibody Signatures in Non-Small-Cell Lung Cancer. Cancer Res 2019; 79:1549-1557. [PMID: 30723114 DOI: 10.1158/0008-5472.can-18-1536] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 09/26/2018] [Accepted: 01/25/2019] [Indexed: 02/07/2023]
Abstract
The majority of non-small-cell lung cancer (NSCLC) cases are diagnosed at advanced stages, primarily because earlier stages of the disease are either asymptomatic or may be attributed to other causes such as infection or long-term effects from smoking. Therefore, early detection of NSCLC would likely increase response and survival rates due to timely intervention. Here, we utilize a novel approach based on whole genome-derived tiled peptide arrays to identify epitopes associated with autoantibody reactivity in NSCLC as a potential means for early detection. Arrays consisted of 2,781,902 tiled peptides representing 20,193 proteins encoded in the human genome. Analysis of 86 prediagnostic samples and 86 matched normal controls from a high-risk cohort revealed 48 proteins with three or more reactive epitopes in NSCLC samples relative to controls. Independent mass spectrometry analysis identified 40 of the 48 proteins in prediagnostic sera from NSCLC samples, of which, 21 occurred in the immunoglobulin-bound fraction. In addition, 63 and 34 proteins encompassed three or more epitopes that were distinct for squamous cell lung cancer and lung adenocarcinoma, respectively. Collectively, these data show that tiled peptide arrays provide a means to delineate epitopes encoded across the genome that trigger an autoantibody response associated with tumor development. SIGNIFICANCE: This study provides a modality for early diagnosis of NSCLC for precision oncology that can be applied to other cancer types.
Collapse
Affiliation(s)
- Yuanqing Yan
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Nan Sun
- Red and Charline McCombs Institute for the Early Detection and Treatment of Cancer, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hong Wang
- Red and Charline McCombs Institute for the Early Detection and Treatment of Cancer, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Makoto Kobayashi
- Red and Charline McCombs Institute for the Early Detection and Treatment of Cancer, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jon J Ladd
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - James P Long
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ken C Lo
- Roche Diagnostics, Madison, Wisconsin
| | | | | | | | - Gary E Goodman
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Kim-Anh Do
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Samir M Hanash
- Red and Charline McCombs Institute for the Early Detection and Treatment of Cancer, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
20
|
Arjomandi A, Delanoy ML, Walker RP, Binder SR. A novel algorithm to improve specificity in ovarian cancer detection. Cancer Treat Res Commun 2018; 15:32-35. [PMID: 30207285 DOI: 10.1016/j.ctarc.2017.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 05/02/2015] [Indexed: 11/18/2022]
Abstract
BACKGROUND Measurement of autoantibodies (AAbs) to tumor associated antigens has been proposed to aid in the early detection of ovarian cancer with high specificity. Here we describe a multiplex approach to evaluate selected peptide epitopes of p53 protein, and propose a novel approach to increase specificity and potentially sensitivity for discrimination between healthy women and women with cancerous masses. MATERIALS AND METHODS 20-mer overlapping peptide epitopes of p53, generated by mapping the complete p53 sequence, were evaluated in a multiplex immunoassay for their detection of serum AAbs in patients with ovarian cancer, using Luminex technology. AAbs to the selected peptides and to p53 full length protein were then detected in a multiplex immunoassay evaluating 359 sera from healthy women and 285 sera from patients with early and late stage ovarian cancer. CA-125 levels were measured in all p53 AAb-positive sera. RESULTS We considered the AAb results together to identify sera where both the full length protein and at least one selected peptide epitope were positive and chose cutoffs that reduced false positives from these AAbs to 1/359 samples, improving specificity. Using this combined approach, we could identify 7 AAb-positive patients that were negative for CA-125 (concentrations below 35 IU/mL); this represents 26% of the p53 positive patients in the total population. CONCLUSION By detecting p53 AAbs in CA-125-negative sera, we demonstrated that combining measurement of AAbs to the full length p53 protein and one or more selected epitopes can potentially improve sensitivity and specificity for ovarian cancer detection.
Collapse
Affiliation(s)
- Audrey Arjomandi
- Clinical Diagnostics Group, Bio-Rad Laboratories, 4000 Alfred Nobel Drive, Hercules, CA 94547, USA.
| | - Michelle L Delanoy
- Clinical Diagnostics Group, Bio-Rad Laboratories, 4000 Alfred Nobel Drive, Hercules, CA 94547, USA
| | - Roger P Walker
- Clinical Diagnostics Group, Bio-Rad Laboratories, 4000 Alfred Nobel Drive, Hercules, CA 94547, USA
| | - Steven R Binder
- Clinical Diagnostics Group, Bio-Rad Laboratories, 4000 Alfred Nobel Drive, Hercules, CA 94547, USA
| |
Collapse
|
21
|
Hayes B, Murphy C, Crawley A, O'Kennedy R. Developments in Point-of-Care Diagnostic Technology for Cancer Detection. Diagnostics (Basel) 2018; 8:diagnostics8020039. [PMID: 29865250 PMCID: PMC6023377 DOI: 10.3390/diagnostics8020039] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 05/22/2018] [Accepted: 05/25/2018] [Indexed: 12/12/2022] Open
Abstract
Cancer is the cause of death for one in seven individuals worldwide. It is widely acknowledged that screening and early diagnosis are of vital importance for improving the likelihood of recovery. However, given the costly, time-consuming, and invasive nature of the many methods currently in use, patients often do not take advantage of the services available to them. Consequently, many researchers are exploring the possibility of developing fast, reliable, and non-invasive diagnostic tools that can be used directly or by local physicians at the point-of-care. Herein, we look at the use of established biomarkers in cancer therapy and investigate emerging biomarkers exhibiting future potential. The incorporation of these biomarkers into point-of-care devices could potentially reduce the strain currently experienced by screening programs in hospitals and healthcare systems. Results derived from point-of-care tests should be accurate, sensitive, and generated rapidly to assist in the selection of the best course of treatment for optimal patient care. Essentially, point-of-care diagnostics should enhance the well-being of patients and lead to a reduction in cancer-related deaths.
Collapse
Affiliation(s)
- Bryony Hayes
- Translational Health Sciences, Bristol Medical School, Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY, UK.
| | - Caroline Murphy
- School of Biotechnology, Dublin City University, Collins Avenue, Glasnevin, Dublin D09 Y5N0, Ireland.
| | - Aoife Crawley
- School of Biotechnology, Dublin City University, Collins Avenue, Glasnevin, Dublin D09 Y5N0, Ireland.
| | - Richard O'Kennedy
- School of Biotechnology, Dublin City University, Collins Avenue, Glasnevin, Dublin D09 Y5N0, Ireland.
- Hamad Bin Khalifa University, Research Complex, P.O. Box 34110 Doha, Qatar.
| |
Collapse
|
22
|
Ushigome M, Nabeya Y, Soda H, Takiguchi N, Kuwajima A, Tagawa M, Matsushita K, Koike J, Funahashi K, Shimada H. Multi-panel assay of serum autoantibodies in colorectal cancer. Int J Clin Oncol 2018; 23:917-923. [DOI: 10.1007/s10147-018-1278-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 04/10/2018] [Indexed: 12/15/2022]
|
23
|
Du Q, Yu R, Wang H, Yan D, Yuan Q, Ma Y, Slamon D, Hou D, Wang H, Wang Q. Significance of tumor-associated autoantibodies in the early diagnosis of lung cancer. CLINICAL RESPIRATORY JOURNAL 2018; 12:2020-2028. [PMID: 29356386 DOI: 10.1111/crj.12769] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 10/16/2017] [Accepted: 01/09/2018] [Indexed: 12/17/2022]
Abstract
OBJECTIVES Autoantibodies tumor-associated antigens (TAAs) could be a valuable tool for the diagnosis or early detection of cancer due to their relatively high specificity and stability. The purpose of this study is to detect the level of tumor-associated autoantibodies in lung cancer and assess the diagnostic potential of autoantibodies in screening strategy for early stage lung cancer. MATERIALS AND METHODS Levels of tumor-associated autoantibodies (AAbs) were measured against a panel of seven TAAs (p53, PGP9.5, SOX2, GAGE7, GBU4-5, CAGE and MAGEA1) in 397 patients with pulmonary lesions (305 with newly diagnosis of NSCLC, 47 with SCLC and 45 with benign nodule) and 74 control persons without any nodules in the lung after chest MDCT scan. The sensitivity, specificity for patients and control persons, positive rate of the panel in different pathology, stage, size of lesion, age and gender were compared and analyzed. RESULTS The AAbs panel could distinguish malignant lesions from benign lesions and control people, with sensitivity of 56.53% and specificity of 91.60%. The specificity could be further increased to 95.80%, when combined with CT. The AAbs also showed high diagnostic value of malignant nodule, and it would be a new method for judgment of malignant nodules that are less than 8 mm in diameter. No significant differences were seen based on pathology, NSCLC stages, tumor size, age or gender. CONCLUSION This assay confirms the value of AAbs panel as a diagnostic tool combined with CT scan.
Collapse
Affiliation(s)
- Qiang Du
- Department of Respiratory Medicine, The Second Affiliated Hospital, Dalian Medical University, Dalian, China.,Department of Respiratory Medicine, The North Area of Suzhou Municipal Hospital, Suzhou, China
| | - Ruofei Yu
- Department of Oncology, The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Han Wang
- Department of Respiratory Medicine, The Second Affiliated Hospital, Dalian Medical University, Dalian, China.,Department of Pharmacy, The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Dong Yan
- Department of Respiratory Medicine, The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Qi Yuan
- Department of Intensive Care Unit, People's Hospital of Liaoning Provincial, Shenyang, China
| | - Yixin Ma
- Department of Respiratory Medicine, The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Dennis Slamon
- Department of Medicine, Division of Hematology Oncology, Medical School of University of California at Los Angeles, Los Angeles 90095, California
| | - Dongmei Hou
- Department of Medicine, Division of Hematology Oncology, Medical School of University of California at Los Angeles, Los Angeles 90095, California
| | - Huiling Wang
- Department of Respiratory Medicine, The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Qi Wang
- Department of Respiratory Medicine, The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|
24
|
The roles and applications of autoantibodies in progression, diagnosis, treatment and prognosis of human malignant tumours. Autoimmun Rev 2017; 16:1270-1281. [PMID: 29042252 DOI: 10.1016/j.autrev.2017.10.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 09/06/2017] [Indexed: 02/07/2023]
Abstract
The existence of autoantibodies towards an individual's own proteins or nucleic acids has been established for more than 100years, and for a long period, these autoantibodies have been believed to be closely associated with autoimmune diseases. However, in recent years, researchers have become more interested in the role and application of autoantibodies in progression, diagnosis, treatment and prognosis of human malignant tumours. Over the past few decades, numerous epidemiological studies have shown that the risk of certain cancers is significantly altered (increased or decreased) in patients with autoimmune diseases, which suggests that autoantibodies may play either promoting or suppressing roles in cancer progression. The idea that autoantibodies are directly involved in tumour progression gains special support by the findings that some antibodies secreted by a variety of cancer cells can promote their proliferation and metastasis. Because the cancer cells generate cell antigenic changes (neoantigens), which trigger the immune system to produce autoantibodies, serum autoantibodies against tumour-associated antigens have been established as a novel type of cancer biomarkers and have been extensively studied in different types of cancer. The autoantibodies as biomarkers in cancer diagnosis are not only more sensitive and specific than antigens, but also could appear before clinical evidences of the tumours, thus disclosing them. The observations that cancer risk is lower in patients with some autoimmune diseases suggest that certain autoantibodies may be protective from certain cancers. Moreover, the presence of autoantibodies in healthy individuals implies that it could be safe to employ autoantibodies to treat cancer. Of note, an autoantibodies derived from lupus murine model received much attention due to their selective cytotoxicity for malignant tumour cell without harming normal ones. These studies showed the therapeutic value of autoantibodies in cancer. In this review, we revisited the pathological or protective role of autoantibodies in cancer progression, summarize the application of autoantibodies in cancer diagnosis and prognosis, and discuss the value of autoantibodies in cancer therapy. The studies established to date suggest that autoantibodies not only regulate cancer progression but also promise to be valuable instruments in oncological diagnosis and therapy.
Collapse
|
25
|
Ren S, Zhang S, Jiang T, He Y, Ma Z, Cai H, Xu X, Li Y, Cai W, Zhou J, Liu X, Hu X, Zhang J, Yu H, Zhou C, Hirsch FR. Early detection of lung cancer by using an autoantibody panel in Chinese population. Oncoimmunology 2017; 7:e1384108. [PMID: 29308305 DOI: 10.1080/2162402x.2017.1384108] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 09/17/2017] [Accepted: 09/19/2017] [Indexed: 12/17/2022] Open
Abstract
We have previously identified a panel of autoantibodies (AABs), including p53, GAGE7, PGP9.5, CAGE, MAGEA1, SOX2 and GBU4-5, that was helpful in the early diagnosis of lung cancer. This large-scale, multicenter study was undertaken to validate the clinical value of this 7-AABs panel for early detection of lung cancer in a Chinese population. Two independent sets of plasma samples from 2308 participants were available for the assay of AABs (training set = 300; validation set = 2008). The concentrations of AABs were quantitated by enzyme-linked immunosorbent assay (ELISA), and the optimal cutoff value for each AAB was determined in the training set and then applied in the validation set. The value of the 7-AABs panel for the early detection of lung cancer was assessed in 540 patients who presented with ground-glass nodules (GGNs) and/or solid nodules. In the validation set, the sensitivity and specificity of the 7-AABs panel were 61% and 90%, respectively. For stage I and stage II non-small cell lung cancer (NSCLC), the sensitivity of the 7-AABs panel was 62% and 59%, respectively, and for limited stage small cell lung cancer (SCLC) it was 59%; these sensitivity values were considerably higher than for traditional biomarkers (including CEA, NSE and CYFRA21-1). Importantly, the combination of the 7-AABs panel and low-dose computed tomography (CT) scanning significantly improved the diagnostic yield in patients presenting with GGNs and/or solid nodules. In conclusion, our 7-AABs panel has clinical value for early detection of lung cancer, including early-stage lung cancer presenting as GGNs.
Collapse
Affiliation(s)
- Shengxiang Ren
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Shucai Zhang
- Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Tao Jiang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Yayi He
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Zhiyong Ma
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Hourong Cai
- Department of Respiratory Medicine, Nanjing Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing, China
| | - Xiaohong Xu
- Clinical Laboratory Department, Zhejiang Province Cancer Hospital, Hangzhou, Zhejiang, China
| | - Yan Li
- Laboratory Department, Hubei General Hospital, Wuhan, China
| | - Weijing Cai
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Jing Zhou
- Hangzhou Cancer Probe Biotechnology Company, Hangzhou, China
| | - Xiaopeng Liu
- Hangzhou Cancer Probe Biotechnology Company, Hangzhou, China
| | - Xuejun Hu
- Hangzhou Cancer Probe Biotechnology Company, Hangzhou, China
| | - Jun Zhang
- Division of Hematology, Oncology and Blood & Marrow Transplantation, Department of Internal Medicine, Holden Comprehensive Cancer Center, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Hui Yu
- Department of Medicine, Division of Medical Oncology, University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO, USA
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Fred R Hirsch
- Department of Medicine, Division of Medical Oncology, University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
26
|
Meistere I, Werner S, Zayakin P, Siliņa K, Rulle U, Pismennaja A, Šantare D, Kikuste I, Isajevs S, Leja M, Kupčinskas L, Kupčinskas J, Jonaitis L, Wu CY, Brenner H, Linē A, Kalniņa Z. The Prevalence of Cancer-Associated Autoantibodies in Patients with Gastric Cancer and Progressive Grades of Premalignant Lesions. Cancer Epidemiol Biomarkers Prev 2017; 26:1564-1574. [PMID: 28768706 DOI: 10.1158/1055-9965.epi-17-0238] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 06/17/2017] [Accepted: 07/21/2017] [Indexed: 01/14/2023] Open
Abstract
Background: Serum autoantibodies against tumor-associated antigens (TAAs) are detectable in early-stage gastric cancer patients; however, the time point during cancerogenesis when they appear in circulation is still obscure.Methods: In this study, we developed a recombinant antigen microarray and analyzed the prevalence of autoantibodies against 102 TAAs in 829 gastric cancer patients and 929 healthy controls from Caucasian and Asian populations, as well as 100 patients with chronic atrophic gastritis and 775 individuals staged according to different grades of intestinal metaplasia.Results: Six antigens, including CTAG1B/CTAG2, DDX53, IGF2BP2, TP53, and MAGEA3, were predominantly reacting with sera from gastric cancer patients when compared with healthy controls, and the seroreactivity was associated with intestinal-type gastric cancer, but not with patients' Helicobacter pylori status, grade, age, gender, or stage of gastric cancer. We detected gastric cancer-associated seroreactivity in 13% of patients with advanced/severe intestinal metaplasia, which was increased in comparison with mild/moderate intestinal metaplasia (5.3%) and was comparable with that seen in early-stage gastric cancer patients (12%). Moreover, by testing serum samples taken 1 to 9 years before the clinical diagnosis of 18 incident gastric cancer cases, we detected autoantibody responses against several TAAs-SOX2, MYC, BIRC5, IGF2BP1, and MUC1.Conclusions: Our results suggest that humoral immune response against TAAs is generated already during premalignant stages.Impact: Based on the obtained results, cancer-associated autoantibodies might make a valuable contribution to the stratification of high-risk patients with premalignant lesions in the stomach through enhancing the positive predictive power of existing risk models. Cancer Epidemiol Biomarkers Prev; 26(10); 1564-74. ©2017 AACR.
Collapse
Affiliation(s)
- Irēna Meistere
- Cancer Biomarker and Immunotherapy Group, Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Simone Werner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Pawel Zayakin
- Cancer Biomarker and Immunotherapy Group, Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Karīna Siliņa
- Cancer Biomarker and Immunotherapy Group, Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Undīne Rulle
- Cancer Biomarker and Immunotherapy Group, Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Angelina Pismennaja
- Cancer Biomarker and Immunotherapy Group, Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Daiga Šantare
- Institute of Clinical and Preventive Medicine and Faculty of Medicine, University of Latvia, Riga, Latvia
- Riga East University Hospital, Riga, Latvia
| | - Ilze Kikuste
- Institute of Clinical and Preventive Medicine and Faculty of Medicine, University of Latvia, Riga, Latvia
- Digestive Diseases Centre GASTRO, Riga, Latvia
| | - Sergejs Isajevs
- Institute of Clinical and Preventive Medicine and Faculty of Medicine, University of Latvia, Riga, Latvia
- Riga East University Hospital, Riga, Latvia
| | - Mārcis Leja
- Institute of Clinical and Preventive Medicine and Faculty of Medicine, University of Latvia, Riga, Latvia
- Riga East University Hospital, Riga, Latvia
- Digestive Diseases Centre GASTRO, Riga, Latvia
| | - Limas Kupčinskas
- Institute for Digestive Research and Department of Gastroenterology Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Juozas Kupčinskas
- Institute for Digestive Research and Department of Gastroenterology Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Laimas Jonaitis
- Institute for Digestive Research and Department of Gastroenterology Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Chun-Ying Wu
- Division of Gastroenterology and Hepatology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - 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
| | - Aija Linē
- Cancer Biomarker and Immunotherapy Group, Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Zane Kalniņa
- Cancer Biomarker and Immunotherapy Group, Latvian Biomedical Research and Study Centre, Riga, Latvia.
| |
Collapse
|
27
|
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
|
28
|
Hoshino I, Nagata M, Takiguchi N, Nabeya Y, Ikeda A, Yokoi S, Kuwajima A, Tagawa M, Matsushita K, Satoshi Y, Hideaki S. Panel of autoantibodies against multiple tumor-associated antigens for detecting gastric cancer. Cancer Sci 2017; 108:308-315. [PMID: 28064445 PMCID: PMC5378227 DOI: 10.1111/cas.13158] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 01/05/2017] [Accepted: 01/06/2017] [Indexed: 12/20/2022] Open
Abstract
Gastric cancer is the second leading cause of cancer death in the world, and effective diagnosis is extremely important for good outcome. We assessed the diagnostic potential of an autoantibody panel that may provide a novel tool for the early detection of gastric cancer. We analyzed data from patients with gastric cancer and normal controls in test and validation cohorts. Autoantibody levels were measured against a panel of six tumor‐associated antigens (TAAs) by ELISA: p53, heat shock protein 70, HCC‐22‐5, peroxiredoxin VI, KM‐HN‐1, and p90 TAA. We assessed serum autoantibodies in 100 participants in the test cohort. The validation cohort comprised 248 participants. Autoantibodies to at least one of the six antigens showed a sensitivity/specificity of 49.0% (95% confidence interval [CI], 39.2–58.8%)/92.4% (95% CI, 87.2–97.6%), and 52.0% (95% CI, 42.2–61.8%)/90.5% (95% CI, 84.8–96.3%) in the test and validation cohorts, respectively. In the validation cohort, no significant differences were seen when patients were subdivided based on age, sex, depth of tumor invasion, lymph node metastasis, distant metastasis, peritoneal dissemination, or TNM stage. Patients who were positive for more than two antibodies in the panel tended to have a worse prognosis than those who were positive for one or no antibody. Measurement of autoantibody response to multiple TAAs in an optimized panel assay to discriminate patients with early stage gastric cancer from normal controls may aid in the early detection of gastric cancer.
Collapse
Affiliation(s)
- Isamu Hoshino
- Division of Gastroenterological Surgery, Chiba Cancer Center, Chiba, Japan
| | - Matsuo Nagata
- Division of Gastroenterological Surgery, Chiba Cancer Center, Chiba, Japan
| | - Nobuhiro Takiguchi
- Division of Gastroenterological Surgery, Chiba Cancer Center, Chiba, Japan
| | - Yoshihiro Nabeya
- Division of Gastroenterological Surgery, Chiba Cancer Center, Chiba, Japan
| | - Atsushi Ikeda
- Division of Gastroenterological Surgery, Chiba Cancer Center, Chiba, Japan
| | - Sana Yokoi
- Division of Chemotherapy and Cancer Diagnosis, Chiba Cancer Center, Chiba, Japan
| | - Akiko Kuwajima
- Medical & Biological Laboratories Co., Ltd, Nagoya, Japan
| | - Masatoshi Tagawa
- Division of Pathology and Cell Therapy, Chiba Cancer Center, Chiba, Japan
| | - Kazuyuki Matsushita
- Division of Clinical Genetics and Proteomics, Department of Laboratory Medicine, Chiba University Hospital, Chiba, Japan
| | - Yajima Satoshi
- Department of Surgery, School of Medicine, Toho University, Tokyo, Japan
| | - Shimada Hideaki
- Department of Surgery, School of Medicine, Toho University, Tokyo, Japan
| |
Collapse
|
29
|
Yu-Rice Y, Edassery SL, Urban N, Hellstrom I, Hellstrom KE, Deng Y, Li Y, Luborsky JL. Selenium-Binding Protein 1 (SBP1) autoantibodies in ovarian disorders and ovarian cancer. Reproduction 2016; 153:277-284. [PMID: 27965399 DOI: 10.1530/rep-16-0265] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 12/02/2016] [Accepted: 12/12/2016] [Indexed: 12/30/2022]
Abstract
Infertility is a risk factor for ovarian cancer (OvCa). The goal was to determine if antibodies to selenium-binding protein 1 (SBP1), an autoantibody we identified in patients with premature ovarian failure (POF), occurs in both infertility and OvCa patients, and thus could be associated with preneoplasia. Anti-SBP1 was measured by immunoassay against recombinant SBP1, in sera from OvCa (n = 41), infertility (n = 92) and control (n = 87) patients. Infertility causes were POF, unexplained, irregular ovulation or endometriosis. The percent of anti-SBP1-positive sera was higher in POF (P = 0.02), irregular ovulation (P = 0.001), unexplained causes (P = 0.02), late (III-IV)-stage OvCa (P = 0.02) but was not significant in endometriosis, benign ovarian tumors/cysts, early stage (I-II) OvCa or uterine cancer compared to healthy controls. Anti-SBP1 was significantly higher in women with serous (P = 0.04) but not non-serous (P = 0.33) OvCa compared to controls. Also, we determined if anti-SBP1 was associated with CA125 or anti-TP53, markers often studied in OvCa. Anti-TP53 and CA125 were measured by established immunoassays. The ability of anti-SBP1 alone to discriminate infertility or OvCa from controls or when combined with anti-TP53 and CA125, to identify OvCa was evaluated by comparing the area under the curve (AUC) in ROC analysis. Anti-SBP1 alone discriminated infertility (AUC = 0.7; P = 0.001) or OvCa (AUC = 0.67; P = 0.03) from controls. The sensitivity and specificity of OvCa identification was increased by combining CA125, anti-TP53 and anti-SBP1 (AUC = 0.96). Therefore, anti-SBP1 occurs in infertile women with POF, ovulatory disturbances or unexplained infertility and in serous OvCa. This suggests an autoimmune process is associated with the development of serous OvCa.
Collapse
Affiliation(s)
- Yi Yu-Rice
- Department of PharmacologyRush University Medical Center, Chicago, Illinois, USA
| | - Seby L Edassery
- Department of PharmacologyRush University Medical Center, Chicago, Illinois, USA
| | - Nicole Urban
- Fred Hutchinson Cancer Research CenterSeattle, Washington, USA
| | - Ingegerd Hellstrom
- Department of PathologyHarborview Medical Center, University of Washington, Seattle, Washington, USA
| | - Karl Erik Hellstrom
- Department of PathologyHarborview Medical Center, University of Washington, Seattle, Washington, USA
| | - Youping Deng
- Department of Bioinformatics and BiostatisticsRush University Medical Center, Chicago, Illinois, USA
| | - Yan Li
- Department of Bioinformatics and BiostatisticsRush University Medical Center, Chicago, Illinois, USA
| | - Judith L Luborsky
- Department of PharmacologyRush University Medical Center, Chicago, Illinois, USA
| |
Collapse
|
30
|
Kalniņa Z, Meistere I, Kikuste I, Tolmanis I, Zayakin P, Linē A. Emerging blood-based biomarkers for detection of gastric cancer. World J Gastroenterol 2015; 21:11636-11653. [PMID: 26556992 PMCID: PMC4631966 DOI: 10.3748/wjg.v21.i41.11636] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Revised: 07/08/2015] [Accepted: 09/30/2015] [Indexed: 02/06/2023] Open
Abstract
Early detection and efficient monitoring of tumor dynamics are prerequisites for reducing disease burden and mortality, and for improving the management of patients with gastric cancer (GC). Blood-based biomarker assays for the detection of early-stage GC could be of great relevance both for population-wide or risk group-based screening programs, while circulating biomarkers that reflect the genetic make-up and dynamics of the tumor would allow monitoring of treatment efficacy, predict recurrences and assess the genetic heterogeneity of the tumor. Recent research to identify blood-based biomarkers of GC has resulted in the identification of a wide variety of cancer-associated molecules, including various proteins, autoantibodies against tumor associated antigens, cell-free DNA fragments, mRNAs and various non-coding RNAs, circulating tumor cells and cancer-derived extracellular vesicles. Each type of these biomarkers provides different information on the disease status, has different advantages and disadvantages, and distinct clinical usefulness. In the current review, we summarize the recent developments in blood-based GC biomarker discovery, discuss the origin of various types of biomarkers and their clinical usefulness and the technological challenges in the development of biomarker assays for clinical use.
Collapse
|
31
|
Pan PT, Zou FY, Mao XF, Cao DH, Wei SL, Gao X, Wang L. WITHDRAWN: Dual display bacteriophage as a platform for high sensitive detection of serum p53 antibodies in breast cancer patients. Clin Chim Acta 2015:S0009-8981(15)30003-6. [PMID: 26434550 DOI: 10.1016/j.cca.2015.09.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 09/30/2015] [Accepted: 09/30/2015] [Indexed: 11/22/2022]
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.
Collapse
Affiliation(s)
- Peng-Tao Pan
- School of Life Sciences and Technology, Xinxiang University, 191 Jinsui Road, Xinxiang, Henan Province, PR China
| | - Fan-Yu Zou
- School of Life Sciences and Technology, Xinxiang University, 191 Jinsui Road, Xinxiang, Henan Province, PR China
| | - Xue-Fei Mao
- School of Life Sciences and Technology, Xinxiang University, 191 Jinsui Road, Xinxiang, Henan Province, PR China
| | - Dong-Hui Cao
- First Hospital of Jilin University, 71 Xinmin Street, Changchun, Jilin Province, PR China
| | - Shi-Lin Wei
- JiLin Brother Medical Immune Products Co., Ltd, Jilin, Jilin Province, PR China
| | - Xiang Gao
- Institute of Genetics and Cytology, Northeast Normal University, 5268 Renmin Street, Changchun, Jilin Province, PR China.
| | - Li Wang
- Institute of Genetics and Cytology, Northeast Normal University, 5268 Renmin Street, Changchun, Jilin Province, PR China.
| |
Collapse
|
32
|
O’Reilly JA, Fitzgerald J, Fitzgerald S, Kenny D, Kay EW, O’Kennedy R, Kijanka GS. Diagnostic potential of zinc finger protein-specific autoantibodies and associated linear B-cell epitopes in colorectal cancer. PLoS One 2015; 10:e0123469. [PMID: 25875936 PMCID: PMC4395473 DOI: 10.1371/journal.pone.0123469] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 03/04/2015] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer is one of the most common cancers worldwide with almost 700,000 deaths every year. Detection of colorectal cancer at an early stage significantly improves patient survival. Cancer-specific autoantibodies found in sera of cancer patients can be used for pre-symptomatic detection of the disease. In this study we assess the zinc finger proteins ZNF346, ZNF638, ZNF700 and ZNF768 as capture antigens for the detection of autoantibodies in colorectal cancer. Sera from 96 patients with colorectal cancer and 35 control patients with no evidence of cancer on colonoscopy were analysed for the presence of ZNF-specific autoantibodies using an indirect ELISA. Autoantibodies to individual ZNF proteins were detected in 10-20% of colorectal cancer patients and in 0-5.7% of controls. A panel of all four ZNF proteins resulted in an assay specificity of 91.4% and sensitivity of 41.7% for the detection of cancer patients in a cohort of non-cancer controls and colorectal cancer patients. Clinicopathological and survival analysis revealed that ZNF autoantibodies were independent of disease stage and did not correlate with disease outcome. Since ZNF autoantibodies were shared between patients and corresponding ZNF proteins showed similarities in their zinc finger motifs, we performed an in silico epitope sequence analysis. Zinc finger proteins ZNF700 and ZNF768 showed the highest sequence similarity with a bl2seq score of 262 (E-value 1E-81) and their classical C2H2 ZNF motifs were identified as potential epitopes contributing to their elevated immunogenic potential. Our findings show an enhanced and specific immunogenicity to zinc finger proteins, thereby providing a multiplexed autoantibody assay for minimally invasive detection of colorectal cancer.
Collapse
Affiliation(s)
- Julie-Ann O’Reilly
- Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland
| | - Jenny Fitzgerald
- Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland
| | - Seán Fitzgerald
- Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland
- School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Dermot Kenny
- Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Elaine W. Kay
- Department of Pathology, Royal College of Surgeons in Ireland and Beaumont Hospital, Dublin, Ireland
| | - Richard O’Kennedy
- Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland
- School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Gregor S. Kijanka
- Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland
| |
Collapse
|
33
|
Chai Y, Peng B, Dai L, Qian W, Zhang Y, Zhang JY. Autoantibodies response to MDM2 and p53 in the immunodiagnosis of esophageal squamous cell carcinoma. Scand J Immunol 2014; 80:362-368. [PMID: 24965442 DOI: 10.1111/sji.12202] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 06/05/2014] [Indexed: 01/01/2023]
Abstract
The human homologue of the mouse double minute 2 (MDM2) is known to be overexpressed in a variety of human malignancies. As one of E3 ubiquitin-protein ligases, MDM2 interacts with the tumour suppressor p53 by mediating ubiquitination and degradation of p53. Since abnormally expressed proteins can induce autoimmune response, to further examine whether sera from patients with esophageal squamous cell carcinoma (ESCC) exhibited immunoreactivity against MDM2 and p53, autoantibody responses to MDM2 and p53 were evaluated by enzyme-linked immunosorbent assay (ELISA) in sera from patients with ESCC and normal individuals. Positive results were also confirmed by Western blotting and indirect immunofluorescence assay. The results demonstrated that the positive rate of autoantibody against p53 and MDM2 in ESCC sera was 22.9% (36/157) and 14.0% (22/157), whereas this rate was 0% (0/85) and 1.2% (1/85), respectively, in normal individuals. Some of the sera with antibodies specific for MDM2 also contained antibodies against p53. And there was an increase of positive antibody reactions reaching a frequency of 35% (55/157) combination with MDM2 and p53. This was significantly higher than the frequency of antibodies in normal individuals (P < 0.01). Our preliminary results suggest that autoantibodies against MDM2 and p53 may be useful serum biomarkers in the immunodiagnosis of ESCC.
Collapse
Affiliation(s)
- Y Chai
- Center for Tumor Biotherapy, The First Affiliated Hospital, College of Public Health, Zhengzhou University, Zhengzhou, Henan, 450052, China ; Department of Biological Sciences, The University of Texas at El Paso, 500 W. University Ave., El Paso, TX, 79968, USA
| | | | | | | | | | | |
Collapse
|
34
|
Annels NE, Simpson GR, Denyer M, McGrath SE, Falgari G, Killick E, Eeles R, Stebbing J, Pchejetski D, Cutress R, Murray N, Michael A, Pandha H. Spontaneous antibodies against Engrailed-2 (EN2) protein in patients with prostate cancer. Clin Exp Immunol 2014; 177:428-38. [PMID: 24654775 PMCID: PMC4226594 DOI: 10.1111/cei.12332] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2014] [Indexed: 11/27/2022] Open
Abstract
We reported the expression of the homeodomain-containing transcription factor Engrailed-2 (EN2) in prostate cancer and showed that the presence of EN2 protein in the urine was highly predictive of prostate cancer. This study aimed to determine whether patients with prostate cancer have EN2 autoantibodies, what the prevalence of these antibodies is and whether they are associated with disease stage. The spontaneous immunoglobulin (Ig)G immune response against EN2 and for comparison the tumour antigen New York Esophageal Squamous Cell Carcinoma 1 (NY-ESO-1), were tested by enzyme-linked immunosorbent assay (ELISA) in three different cohorts of prostate cancer patients as well as a group of men genetically predisposed to prostate cancer. Thirty-two of 353 (9·1%) of the SUN cohort representing all stages of prostate cancer demonstrated EN2 IgG responses, 12 of 107 patients (11·2%) in the advanced prostate cancer patients showed responses, while only four of 121 patients (3·3%) with castrate-resistant prostate cancer showed EN2 autoantibodies. No significant responses were found in the predisposed group. Anti-EN2 IgG responses were significantly higher in patients with prostate cancer compared to healthy control males and similarly prevalent to anti-NY-ESO-1 responses. While EN2 autoantibodies are not a useful diagnostic or monitoring tool, EN2 immunogenicity provides the rationale to pursue studies using EN2 as an immunotherapeutic target.
Collapse
Affiliation(s)
- N E Annels
- Oncology, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, UK
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Xu YW, Peng YH, Chen B, Wu ZY, Wu JY, Shen JH, Zheng CP, Wang SH, Guo HP, Li EM, Xu LY. Autoantibodies as potential biomarkers for the early detection of esophageal squamous cell carcinoma. Am J Gastroenterol 2014; 109:36-45. [PMID: 24296751 PMCID: PMC3887578 DOI: 10.1038/ajg.2013.384] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 09/17/2013] [Indexed: 02/05/2023]
Abstract
OBJECTIVES Esophageal squamous cell carcinoma (ESCC) is one of the most frequent causes of cancer death worldwide and effective diagnosis is needed. We assessed the diagnostic potential of an autoantibody panel that may benefit early diagnosis. METHODS We analyzed data for patients with ESCC and normal controls in a test cohort and a validation cohort. Autoantibody levels were measured against a panel of six tumor-associated antigens (p53, NY-ESO-1, matrix metalloproteinase-7 (MMP-7), heat shock protein 70 (Hsp70), peroxiredoxin VI (Prx VI), and BMI1 polycomb ring finger oncogene (Bmi-1)) by enzyme-linked immunosorbent assay. RESULTS We assessed serum autoantibodies in 513 participants: 388 with ESCC and 125 normal controls. The validation cohort comprised 371 participants: 237 with ESCC, and 134 normal controls. Autoantibodies to at least 1 of 6 antigens demonstrated a sensitivity/specificity of 57% (95% confidence interval (CI): 52-62%)/95% (95% CI: 89-98%) and 51% (95% CI: 45-57%)/96% (95% CI: 91-99%) in the test and validation cohorts, respectively. Measurement of the autoantibody panel could differentiate early-stage ESCC patients from normal controls (sensitivity 45% (95% CI: 32-59%) and specificity 95% (95% CI: 89-98%) in the test cohort; 46% (95% CI: 35-58%) and 96% (95% CI: 91-99%) in the validation cohort). In either cohort, no significant differences were seen when patients were subdivided by age, gender, smoking status, size of tumor, site of tumor, depth of tumor invasion, histological grade, lymph node status, TNM stage, or early-stage and late-stage groups. CONCLUSIONS Measurement of an autoantibody response to multiple tumor-associated antigens in an optimized panel assay, to help discriminate early-stage ESCC patients from normal controls, may aid in early detection of ESCC.
Collapse
Affiliation(s)
- Yi-Wei Xu
- Institute of Oncologic Pathology, The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, Guangdong, China
- Department of Clinical Laboratory, the Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Yu-Hui Peng
- Department of Clinical Laboratory, the Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Bo Chen
- Institute of Oncologic Pathology, The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, Guangdong, China
| | - Zhi-Yong Wu
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, Guangdong, China
- Department of Oncology Surgery, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou, Guangdong, China
| | - Jian-Yi Wu
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, Guangdong, China
| | - Jin-Hui Shen
- Department of Pathology, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou, Guangdong, China
| | - Chun-Peng Zheng
- Department of Oncology Surgery, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou, Guangdong, China
| | - Shao-Hong Wang
- Department of Pathology, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou, Guangdong, China
| | - Hai-Peng Guo
- Department of Head and Neck Surgery, the Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - En-Min Li
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, Guangdong, China
| | - Li-Yan Xu
- Institute of Oncologic Pathology, The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, Guangdong, China
- Institute of Oncologic Pathology, Shantou University Medical College, No. 22, Xinling Road, Shantou 515041, Guangdong, China or Department of Biochemistry and Molecular Biology, Shantou University Medical College, No. 22, Xinling Road, Shantou 515041, Guangdong, China. E-mail: or
| |
Collapse
|
36
|
Serum anti-CCNY autoantibody is an independent prognosis indicator for postoperative patients with early-stage nonsmall-cell lung carcinoma. DISEASE MARKERS 2013; 35:317-25. [PMID: 24167380 PMCID: PMC3789285 DOI: 10.1155/2013/935943] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Revised: 08/20/2013] [Accepted: 08/21/2013] [Indexed: 02/06/2023]
Abstract
Cyclin Y (CCNY) is a novel cyclin and almost nothing is known about its role in human cancers. To investigate the clinical significance of serum anti-CCNY autoantibodies in nonsmall-cell lung carcinoma (NSCLC), the serum levels of CCNY protein in 264 patients with NSCLC, 103 patients with tuberculosis, and 89 healthy controls were analyzed by immunohistochemistry. The result shows that, compared with normal lung tissues, the NSCLC tissues contained higher levels of CCNY protein. The levels of anti-CCNY autoantibodies were higher in the sera of the patients with NSCLC than in the sera of the healthy controls (P < 0.001) or the patients with tuberculosis (P = 0.027). Moreover, in a Cox regression analysis, anti-CCNY autoantibody was an independent factor that predicted poor prognosis for postoperative patients with early-stage NSCLC (P = 0.026) as well as for those with distant metastasis (P = 0.012). Our data indicated that Anti-CCNY autoantibody may be useful as a latent tumor marker to facilitate diagnosis and may represent a novel prognostic indicator for patients with early stage NSCLC.
Collapse
|
37
|
Autoantibodies to MUC1 glycopeptides cannot be used as a screening assay for early detection of breast, ovarian, lung or pancreatic cancer. Br J Cancer 2013; 108:2045-55. [PMID: 23652307 PMCID: PMC3670483 DOI: 10.1038/bjc.2013.214] [Citation(s) in RCA: 231] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background: Autoantibodies have been detected in sera before diagnosis of cancer leading to interest in their potential as screening/early detection biomarkers. As we have found autoantibodies to MUC1 glycopeptides to be elevated in early-stage breast cancer patients, in this study we analysed these autoantibodies in large population cohorts of sera taken before cancer diagnosis. Methods: Serum samples from women who subsequently developed breast cancer, and aged-matched controls, were identified from UK Collaborative Trial of Ovarian Cancer Screening (UKCTOCS) and Guernsey serum banks to formed discovery and validation sets. These were screened on a microarray platform of 60mer MUC1 glycopeptides and recombinant MUC1 containing 16 tandem repeats. Additional case–control sets comprised of women who subsequently developed ovarian, pancreatic and lung cancer were also screened on the arrays. Results: In the discovery (273 cases, 273 controls) and the two validation sets (UKCTOCS 426 cases, 426 controls; Guernsey 303 cases and 606 controls), no differences were found in autoantibody reactivity to MUC1 tandem repeat peptide or glycoforms between cases and controls. Furthermore, no differences were observed between ovarian, pancreatic and lung cancer cases and controls. Conclusion: This robust, validated study shows autoantibodies to MUC1 peptide or glycopeptides cannot be used for breast, ovarian, lung or pancreatic cancer screening. This has significant implications for research on the use of MUC1 in cancer detection.
Collapse
|
38
|
Chatterjee M, Dyson G, Levin NK, Shah JP, Morris R, Munkarah A, Tainsky MA. Tumor autoantibodies as biomarkers for predicting ovarian cancer recurrence. Cancer Biomark 2013; 11:59-73. [PMID: 23011153 DOI: 10.3233/cbm-2012-0265] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Ovarian cancer (OVCA) has a high incidence of recurrence and a high rate of mortality. We performed a pilot study to evaluate the usefulness of tumor autoantibodies to tumor associated antigens (TAA) to predict OVCA recurrence. A validation study with 56 antigens, previously identified in the initial phase of the study, along with 13 known tumor antigens on protein arrays was performed on an independent cohort of recurrent and non-recurrent OVCA patients. Statistical analyses revealed that a panel of 3 antigens predicted recurrence at a median time of 9.07 months prior to clinical recurrence in a study population, where majority of patients had CA125 values less than 35 U/ml, with an average sensitivity, specificity and accuracy of 94.7%, 86.7% and 93.3% respectively. One of the top 3 antigens has been associated with the development of polymyositis (PM) which has been shown in some cases to precede the occurrence of ovarian carcinoma. Our results indicate that these 3 antigens have potential for predicting recurrence at an early time and may have better prognostic utility than CA125 alone for early therapeutic intervention. These biomarkers could guide us to identify those patients that could benefit most from maintenance or consolidation therapy.
Collapse
Affiliation(s)
- Madhumita Chatterjee
- Program in Molecular Biology and Genetics, Karmanos Cancer Institute, Detroit, MI 48201-3917, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Mattioni M, Chinzari P, Soddu S, Strigari L, Cilenti V, Mastropasqua E. Serum p53 antibody detection in patients with impaired lung function. BMC Cancer 2013; 13:62. [PMID: 23384026 PMCID: PMC3570357 DOI: 10.1186/1471-2407-13-62] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Accepted: 01/30/2013] [Indexed: 11/10/2022] Open
Abstract
Background TP53 gene mutations can lead to the expression of a dysfunctional protein that in turn may enable genetically unstable cells to survive and change into malignant cells. Mutant p53 accumulates early in cells and can precociously induce circulating anti-p53 antibodies (p53Abs); in fact, p53 overexpression has been observed in pre-neoplastic lesions, such as bronchial dysplasia, and p53Abs have been found in patients with Chronic Obstructive Pulmonary Disease, before the diagnosis of lung and other tobacco-related tumors. Methods A large prospective study was carried out, enrolling non-smokers, ex-smokers and smokers with or without the impairment of lung function, to analyze the incidence of serum p53Abs and the correlation with clinicopathologic features, in particular smoking habits and impairment of lung function, in order to investigate their possible role as early markers of the onset of lung cancer or other cancers. The p53Ab levels were evaluated by a specific ELISA in 675 subjects. Results Data showed that significant levels of serum p53Abs were present in 35 subjects (5.2%); no difference was observed in the presence of p53Abs with regard to age and gender, while p53Abs correlated with the number of cigarettes smoked per day and packs-year. Furthermore, serum p53Abs were associated with the worst lung function impairment. The median p53Ab level in positive subjects was 3.5 units/ml (range 1.2 to 65.3 units/ml). Only fifteen positive subjects participated in the follow-up, again resulting positive for serum p53Abs, and no evidence of cancer was found in these patients. Conclusion The presence of serum p53Abs was found to be associated with smoking level and lung function impairment, both risk factors of cancer development. However, in our study we have not observed the occurrence of lung cancer or other cancers in the follow-up of positive subjects, therefore we cannot directly correlate the presence of serum p53Abs with cancer risk.
Collapse
Affiliation(s)
- Manlio Mattioni
- Experimental Research Centre, Regina Elena National Cancer Institute, via delle Messi d'Oro 156, 00158, Rome, Italy.
| | | | | | | | | | | |
Collapse
|
40
|
Pedersen JW, Gentry-Maharaj A, Fourkala EO, Dawnay A, Burnell M, Zaikin A, Pedersen AE, Jacobs I, Menon U, Wandall HH. Early detection of cancer in the general population: a blinded case-control study of p53 autoantibodies in colorectal cancer. Br J Cancer 2012; 108:107-14. [PMID: 23169294 PMCID: PMC3553520 DOI: 10.1038/bjc.2012.517] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background: Recent reports from cancer screening trials in high-risk populations suggest that autoantibodies can be detected before clinical diagnosis. However, there is minimal data on the role of autoantibody signatures in cancer screening in the general population. Methods: Informative p53 peptides were identified in sera from patients with colorectal cancer using an autoantibody microarray with 15-mer overlapping peptides covering the complete p53 sequence. The selected peptides were evaluated in a blinded case–control study using stored serum from the multimodal arm of the United Kingdom Collaborative Trial of Ovarian Cancer Screening where women gave annual blood samples. Cases were postmenopausal women who developed colorectal cancer following recruitment, with 2 or more serum samples preceding diagnosis. Controls were age-matched women with no history of cancer. Results: The 50 640 women randomised to the multimodal group were followed up for a median of 6.8 (inter-quartile range 5.9–8.4) years. Colorectal cancer notification was received in 101 women with serial samples of whom 97 (297 samples) had given consent for secondary studies. They were matched 1 : 1 with 97 controls (296 serial samples). The four most informative peptides identified 25.8% of colorectal cancer patients with a specificity of 95%. The median lead time was 1.4 (range 0.12–3.8) years before clinical diagnosis. Conclusion: Our findings suggest that in the general population, autoantibody signatures are detectable during preclinical disease and may be of value in cancer screening. In colorectal cancer screening in particular, where the current need is to improve compliance, it suggests that p53 autoantibodies may contribute towards risk stratification.
Collapse
Affiliation(s)
- J W Pedersen
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen N, DK-2200, Denmark
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Development and validation of a high throughput system for discovery of antigens for autoantibody detection. PLoS One 2012; 7:e40759. [PMID: 22815807 PMCID: PMC3399887 DOI: 10.1371/journal.pone.0040759] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Accepted: 06/12/2012] [Indexed: 12/03/2022] Open
Abstract
An assay employing a panel of tumor-associated antigens has been validated and is available commercially (EarlyCDT®-Lung) to aid the early detection of lung cancer by measurement of serum autoantibodies. The high throughput (HTP) strategy described herein was pursued to identify new antigens to add to the EarlyCDT-Lung panel and to assist in the development of new panels for other cancers. Two ligation-independent cloning vectors were designed and synthesized, producing fusion proteins suitable for the autoantibody ELISA. We developed an abridged HTP version of the validated autoantibody ELISA, determining that results reflected the performance of the EarlyCDT assay, by comparing results on both formats. Once validated this HTP ELISA was utilized to screen multiple fusion proteins prepared on small-scale, by a HTP expression screen. We determined whether the assay performance for these HTP protein batches was an accurate reflection of the performance of R&D or commercial batches. A HTP discovery platform for the identification and optimal production of tumor- associated antigens which detects autoantibodies has been developed and validated. The most favorable conditions for the exposure of immunogenic epitopes were assessed to produce discriminatory proteins for use in a commercial ELISA. This process is rapid and cost-effective compared to standard cloning and screening technologies and enables rapid advancement in the field of autoantibody assay discovery. This approach will significantly reduce timescale and costs for developing similar panels of autoantibody assays for the detection of other cancer types with the ultimate aim of improved overall survival due to early diagnosis and treatment.
Collapse
|
42
|
Lu H, Ladd J, Feng Z, Wu M, Goodell V, Pitteri SJ, Li CI, Prentice R, Hanash SM, Disis ML. Evaluation of known oncoantibodies, HER2, p53, and cyclin B1, in prediagnostic breast cancer sera. Cancer Prev Res (Phila) 2012; 5:1036-43. [PMID: 22715141 DOI: 10.1158/1940-6207.capr-11-0558] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Serum autoantibodies, directed against oncogenic proteins, have been frequently detected in the sera of patients with breast cancer. It is unknown whether serum antibodies that are identified in patients with established disease could also be detected in patients with newly diagnosed disease or even predate the diagnosis of breast cancer. Using sera collected at the time of treatment, at the time of diagnosis, or before the time of diagnosis, the current study aimed to address the temporal relationship between breast cancer development and serum antibody response. Starting from serum antibodies to eight known breast cancer antigens, we first identified four serum antibodies, HER2/neu, p53, carcinoembryonic antigen (CEA), and cyclin B1, which are significantly increased in the sera collected from patients with breast cancer at the time of treatment. These antibodies were also elevated in breast cancer sera collected at the time of diagnosis. Finally, comparison of antibody responses in prediagnostic samples from women before the development of breast cancer and in controls showed that antibodies to the HER2/neu and p53 can be detected in sera that were collected on average more than 150 days before a breast cancer diagnosis. These results showed that serum autoantibodies commonly reported in sera from patients with established disease can also be detected in prediagnostic sera and may be useful for the early detection of breast cancer.
Collapse
Affiliation(s)
- Hailing Lu
- Tumor Vaccine Group, University of Washington, Box 358050, Seattle, WA 98109, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Murphy MA, O'Connell DJ, O'Kane SL, O'Brien JK, O'Toole S, Martin C, Sheils O, O'Leary JJ, Cahill DJ. Epitope presentation is an important determinant of the utility of antigens identified from protein arrays in the development of autoantibody diagnostic assays. J Proteomics 2012; 75:4668-75. [PMID: 22415278 DOI: 10.1016/j.jprot.2012.02.031] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Revised: 02/24/2012] [Accepted: 02/25/2012] [Indexed: 01/19/2023]
Abstract
Autoantibodies represent an attractive biomarker for diagnostic assays principally due to the stability of immunoglobulin in patient serum facilitating measurement with conventional assays. Immune responses to tumorigenesis may facilitate detection of ovarian cancer in the early stages of the disease with identification of a panel of tumour specific autoantibodies. Despite the reporting of many tumour associated autoantibodies using arrays of tumour antigens, this has not led to the advance in diagnostic capability as rapidly as was initially expected. Here we examine the potential diagnostic utility of candidate autoantibody biomarkers identified via screening of serum samples on a high content human protein array from a unique cohort of early stage and late stage ovarian cancer patients. We analyse the performance of autoantibodies to the tumour suppressor protein p53 and the novel autoantigens alpha adducin and endosulfine alpha identified in our array screen. Each antigen has different performance characteristics using conventional ELISA format and Western blot immunoassay. The high attrition rate of promising autoantigens identified by array screening can in part be explained by the presentation of the epitope of the antigen in the subsequent method of validation and this study provides directions on maximising the potential of candidate biomarkers. This article is part of a Special Issue entitled: Translational Proteomics.
Collapse
Affiliation(s)
- Mairead A Murphy
- School of Medicine and Medical Sciences, Conway Institute of Biomedical and Biomolecular Research, University College Dublin, Dublin 4, Ireland
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Auto-anticorps dirigés contre les antigènes associés aux tumeurs : nouveaux outils pour la détection précoce du cancer du poumon. Bull Cancer 2011; 98:1419-30. [DOI: 10.1684/bdc.2011.1499] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
45
|
Lam S, Boyle P, Healey GF, Maddison P, Peek L, Murray A, Chapman CJ, Allen J, Wood WC, Sewell HF, Robertson JFR. EarlyCDT-Lung: an immunobiomarker test as an aid to early detection of lung cancer. Cancer Prev Res (Phila) 2011; 4:1126-34. [PMID: 21733826 DOI: 10.1158/1940-6207.capr-10-0328] [Citation(s) in RCA: 117] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Recent publications have reported the technical and clinical validation of EarlyCDT-Lung, an autoantibody test which detected elevated autoantibodies in 40% of lung cancers at diagnosis. This manuscript reports the results of EarlyCDT-Lung run on four new (postvalidation) data sets. The following four cohorts of patients (n = 574) with newly diagnosed lung cancer were identified: group 1 (n = 122), 100% small cell lung cancer (SCLC); group 2 (n = 249), 97% non-small cell lung cancer (NSCLC); group 3 (n = 122), 100% NSCLC; group 4 (n = 81), 62% NSCLC. Serum samples were obtained after diagnosis, prior to any anticancer treatment. Autoantibody levels were measured against a panel of six tumor-related antigens (p53, NY-ESO-1, CAGE, GBU4-5, Annexin 1, and SOX2) in the EarlyCDT-Lung panel and previously established cutoffs applied. In groups 2, 3, and 4, patients were individually matched by gender, age, and smoking history to a control individual with no history of malignant disease. Assay sensitivity was tested in relation to cancer type and stage, and in the matched normals to demographic variables. The autoantibody panel showed sensitivity/specificity of 57%/n.d (not done) for SCLC in group 1, 34%/87% for NSCLC in group 2, 31% and 84% for NSCLC in group 3, and 35%/89% for NSCLC and 43%/89% for SCLC in group 4. There was no significant difference in positivity of EarlyCDT-Lung and different lung cancer stages. These studies confirm the value of an autoantibody assay, EarlyCDT-Lung, as an aid to detecting lung cancer in patients at high risk of the disease.
Collapse
Affiliation(s)
- Stephen Lam
- Department of Pulmonary Medicine, British Columbia Cancer Agency, Vancouver, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Tsuji T, Matsuzaki J, Ritter E, Miliotto A, Ritter G, Odunsi K, Old LJ, Gnjatic S. Split T cell tolerance against a self/tumor antigen: spontaneous CD4+ but not CD8+ T cell responses against p53 in cancer patients and healthy donors. PLoS One 2011; 6:e23651. [PMID: 21858191 PMCID: PMC3155555 DOI: 10.1371/journal.pone.0023651] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2011] [Accepted: 07/22/2011] [Indexed: 12/20/2022] Open
Abstract
Analyses of NY-ESO-1-specific spontaneous immune responses in cancer patients revealed that antibody and both CD4+ and CD8+ T cell responses were induced together in cancer patients. To explore whether such integrated immune responses are also spontaneously induced for other tumor antigens, we have evaluated antibody and T cell responses against self/tumor antigen p53 in ovarian cancer patients and healthy individuals. We found that 21% (64/298) of ovarian cancer patients but no healthy donors showed specific IgG responses against wild-type p53 protein. While none of 12 patients with high titer p53 antibody showed spontaneous p53-specific CD8+ T cell responses following a single in vitro sensitization, significant p53-specific IFN-γ producing CD4+ T cells were detected in 6 patients. Surprisingly, similar levels of p53-specific CD4+ T cells but not CD8+ T cells were also detected in 5/10 seronegative cancer patients and 9/12 healthy donors. Importantly, p53-specific CD4+ T cells in healthy donors originated from a CD45RA− antigen-experienced T cell population and recognized naturally processed wild-type p53 protein. These results raise the possibility that p53-specific CD4+ T cells reflect abnormalities in p53 occurring in normal individuals and that they may play a role in processes of immunosurveillance or immunoregulation of p53-related neoplastic events.
Collapse
Affiliation(s)
- Takemasa Tsuji
- Ludwig Institute for Cancer Research Ltd., New York Branch at Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Junko Matsuzaki
- Department of Gynecologic Oncology, Roswell Park Cancer Institute, Buffalo, New York, United States of America
| | - Erika Ritter
- Ludwig Institute for Cancer Research Ltd., New York Branch at Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Anthony Miliotto
- Department of Gynecologic Oncology, Roswell Park Cancer Institute, Buffalo, New York, United States of America
| | - Gerd Ritter
- Ludwig Institute for Cancer Research Ltd., New York Branch at Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Kunle Odunsi
- Department of Gynecologic Oncology, Roswell Park Cancer Institute, Buffalo, New York, United States of America
| | - Lloyd J. Old
- Ludwig Institute for Cancer Research Ltd., New York Branch at Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Sacha Gnjatic
- Ludwig Institute for Cancer Research Ltd., New York Branch at Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
- * E-mail: .
| |
Collapse
|
47
|
Hanash S. Harnessing the immune response for cancer detection. Cancer Epidemiol Biomarkers Prev 2011; 20:569-70. [PMID: 21454417 DOI: 10.1158/1055-9965.epi-11-0183] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Samir Hanash
- Fred Hutchinson Cancer Research Center, Molecular Diagnostics, 1100 Fairview Avenue North, M5-C800, PO Box 19024, Seattle, WA 98109, USA.
| |
Collapse
|
48
|
Desmetz C, Lacombe J, Mange A, Maudelonde T, Solassol J. Autoanticorps et diagnostic précoce des cancers. Med Sci (Paris) 2011; 27:633-8. [DOI: 10.1051/medsci/2011276016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
49
|
Lu D, Kuhn E, Bristow RE, Giuntoli RL, Kjær SK, Shih IM, Roden RBS. Comparison of candidate serologic markers for type I and type II ovarian cancer. Gynecol Oncol 2011; 122:560-6. [PMID: 21704359 DOI: 10.1016/j.ygyno.2011.05.039] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Revised: 05/26/2011] [Accepted: 05/29/2011] [Indexed: 12/28/2022]
Abstract
OBJECTIVE To examine the value of individual and combinations of ovarian cancer associated blood biomarkers for the discrimination between plasma of patients with type I or II ovarian cancer and disease-free volunteers. METHODS Levels of 14 currently promising ovarian cancer-related biomarkers, including CA125, macrophage inhibitory factor-1 (MIF-1), leptin, prolactin, osteopontin (OPN), insulin-like growth factor-II (IGF-II), autoantibodies (AAbs) to eight proteins: p53, NY-ESO-1, p16, ALPP, CTSD, B23, GRP78, and SSX, were measured in the plasma of 151 ovarian cancer patients, 23 with borderline ovarian tumors, 55 with benign tumors and 75 healthy controls. RESULTS When examined individually, seven candidate biomarkers (MIF, Prolactin, CA-125, OPN, Leptin, IGF-II and p53 AAbs) had significantly different plasma levels between type II ovarian cancer patients and healthy controls. Based on the receiver operating characteristic (ROC) curves constructed and area under the curve (AUC) calculated, CA125 exhibited the greatest power to discriminate the plasma samples of type II cancer patients from normal volunteers (AUC 0.9310), followed by IGF-II (AUC 0.8514), OPN (AUC 0.7888), leptin (AUC 0.7571), prolactin (AUC 0.7247), p53 AAbs (AUC 0.7033), and MIF (AUC 0.6992). p53 AAbs levels exhibited the lowest correlation with CA125 levels among the six markers, suggesting the potential of p53 AAbs as a biomarker independent of CA125. Indeed, p53 AAbs increased the AUC of ROC curve to the greatest extent when combining CA125 with one of the other markers. At a fixed specificity of 100%, the addition of p53 AAbs to CA125 increased sensitivity from 73.8% to 85.7% to discriminate type II cancer patients from normal controls. Notably, seropositivity of p53 AAbs is comparable in type II ovarian cancer patients with negative and positive CA125, but has no value for type I ovarian cancer patients. CONCLUSIONS p53 AAbs might be a useful blood-based biomarker for the detection of type II ovarian cancer, especially when combined with CA125 levels.
Collapse
Affiliation(s)
- Dan Lu
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
|