1
|
Kajornsrichon W, Chaisaingmongkol J, Pomyen Y, Tit-Oon P, Wang XW, Ruchirawat M, Fuangthong M. Identification of autoantibodies as potential non-invasive biomarkers for intrahepatic cholangiocarcinoma. Sci Rep 2024; 14:20012. [PMID: 39198554 PMCID: PMC11358490 DOI: 10.1038/s41598-024-70595-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 08/19/2024] [Indexed: 09/01/2024] Open
Abstract
Intrahepatic cholangiocarcinoma (iCCA) presents a challenging diagnosis due to its nonspecific early clinical manifestations, often resulting in late-stage detection and high mortality. Diagnosing iCCA is further complicated by its limited accuracy, often necessitating multiple invasive procedures for precise identification. Despite carbohydrate antigen 19-9 (CA19-9) having been investigated and employed for iCCA diagnosis, it demonstrates modest diagnostic performance. Consequently, the identification of novel biomarkers with improved sensitivity and specificity remains an imperative yet formidable task. Autoantibodies, as early indicators of the immune response against cancer, offer a promising avenue for enhancing diagnostic accuracy. Our study aimed to identify non-invasive blood-based autoantibody biomarkers capable of distinguishing iCCA patients from healthy individuals (CTRs). We profiled autoantibodies in 26 serum samples (16 iCCAs and 10 CTRs) using protein microarrays containing 1622 functional proteins. Leveraging machine learning techniques, we identified a signature composed of three autoantibody biomarkers (NDE1, PYCR1, and VIM) in conjunction with CA19-9 for iCCA detection. This combined signature demonstrated superior diagnostic performance with an AUC of 96.9%, outperforming CA19-9 alone (AUC: 83.8%). These results suggest the potential of autoantibody biomarkers to develop a complementary non-invasive diagnostic utility for routine iCCA screening.
Collapse
Grants
- CGS2562/01 Chulabhorn Graduate Scholarship
- 2536699/42113 Ministry of Higher Education, Science, Research and Innovation, and Thailand Science Research and Innovation (TSRI), Chulabhorn Research Institute
- 48292/4691968 Ministry of Higher Education, Science, Research and Innovation, and Thailand Science Research and Innovation (TSRI), Chulabhorn Research Institute
- Intramural Program of the Center for Cancer Research, National Cancer Institute
- Center of Excellence on Environmental Health and Toxicology (EHT), OPS
Collapse
Affiliation(s)
- Wachira Kajornsrichon
- Program in Applied Biological Sciences, Chulabhorn Graduate Institute, Bangkok, 10210, Thailand
| | - Jittiporn Chaisaingmongkol
- Laboratory of Chemical Carcinogenesis, Chulabhorn Research Institute, Bangkok, 10210, Thailand
- Center of Excellence on Environmental Health and Toxicology (EHT), OPS, MHESI, Bangkok, Thailand
| | - Yotsawat Pomyen
- Translational Research Unit, Chulabhorn Research Institute, Bangkok, 10210, Thailand
| | - Phanthakarn Tit-Oon
- Translational Research Unit, Chulabhorn Research Institute, Bangkok, 10210, Thailand
| | - Xin Wei Wang
- Liver Cancer Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Mathuros Ruchirawat
- Laboratory of Chemical Carcinogenesis, Chulabhorn Research Institute, Bangkok, 10210, Thailand
- Center of Excellence on Environmental Health and Toxicology (EHT), OPS, MHESI, Bangkok, Thailand
- Translational Research Unit, Chulabhorn Research Institute, Bangkok, 10210, Thailand
| | - Mayuree Fuangthong
- Program in Applied Biological Sciences, Chulabhorn Graduate Institute, Bangkok, 10210, Thailand.
- Center of Excellence on Environmental Health and Toxicology (EHT), OPS, MHESI, Bangkok, Thailand.
- Translational Research Unit, Chulabhorn Research Institute, Bangkok, 10210, Thailand.
| |
Collapse
|
2
|
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
|
3
|
Jiang Y, Zhang G, Zhu J, Wang X, Tao Z, Yu P. Development and validation of a TAAbs and TAAs based non-invasive model for diagnosing lung cancer. Heliyon 2024; 10:e33888. [PMID: 39027487 PMCID: PMC11255565 DOI: 10.1016/j.heliyon.2024.e33888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/20/2024] Open
Abstract
Background Single Tumor-associated autoantibodies (TAAbs) and tumor-associated antigens (TAAs) have been found to have lower diagnostic efficacy in lung cancer. Our objective is to develop and validate a lung cancer prediction model that utilizes TAAbs and TAAs and to enhance the accuracy of lung cancer detection. Methods 1830 subjects were randomly divided into training and validation sets at a 7:3 ratio for this study. Lasso regression analysis was used to remove collinear variables, whereas univariate logistic regression analysis was employed to identify potential independent risk factors for lung cancer. A diagnostic model was constructed using multivariate logistic analysis. The results were presented as a nomogram and assessed for various performance measures, including area under the curve, calibration curve, and decision curve analysis. Results The diagnostic model was developed using gender, age, GAGE7, MAGE-A1, CA125, and CEA as variables. The training set had an AUC of 0.787, while the validation set had an AUC of 0.750. The calibration curves of the training and validation sets showed a strong agreement between anticipated and observed values. The nomogram performed better than any individual variable in both the training and validation sets in terms of net benefits for lung cancer detection, according to DCA analysis. Conclusions This study proposes a diagnostic model for lung cancer that uses TAAbs and TAAs and incorporates individual characteristics. This model can be easily applied to personalized diagnosis.
Collapse
Affiliation(s)
- Yan Jiang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Gong Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Jiayi Zhu
- Department of Pathology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Xuchu Wang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Zhihua Tao
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Pan Yu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| |
Collapse
|
4
|
Sreelatha SV, Shetty S, Karnaker VK, Jacob AM, Chowdhury CR. Assaying of p53 Autoantibodies in saliva for the detection of oral squamous cell carcinoma. A road not taken. Indian J Cancer 2024; 61:37-42. [PMID: 38159210 DOI: 10.4103/ijc.ijc_870_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 05/12/2021] [Indexed: 01/03/2024]
Abstract
BACKGROUND Autoantibody detection is a promising approach to cancer screening. Serum p53 antibodies have been time tested in various cancers, including oral squamous cell carcinoma (OSCC). This study is aimed to detect and determine the level of p53 autoantibodies (p53-AAbs) in saliva. The association of clinicopathological features among patients with and without OSCC was also explored as a novel method for the detection of autoantibodies. METHODS One hundred preoperative saliva samples from patients with histologically confirmed OSCC and a hundred from normal healthy individuals were collected. Anti p53 detection kit assessed levels of salivary p53-AAbs. The cut-off value was 1.3 U/mL by Enzyme-linked immunosorbent assay (ELISA). The p53-AAb levels were expressed in terms of the median and interquartile range (IQR). Fischer's exact test and Chi-square test were used to determine the association with clinicopathological features among patients with OSCC and healthy controls with tobacco consumption habits. RESULTS Median level of p53-AAb is 0.234 U/mL (IQR 0.18-0.37U/mL) in healthy controls and 0.285U/mL (IQR 0.16-0.58U/mL) in OSCC. p53-AAbs was positive in 15% of 100 patients with OSCC, which was statistically higher ( P < 0.001) among OSCC, and controls were negative for p53-AAb. No significant correlation of p53-AAbs with the patient's age, gender, site, clinical staging (TNM), and pathologic grade was observed. However, a significant association was seen between the node involvement and salivary p53-AAbs. CONCLUSION Salivary p53-AAb positivity was seen in a higher proportion in OSCC patients than in healthy controls with tobacco consumption, and the levels did differ significantly among OSCC and healthy controls.
Collapse
Affiliation(s)
- S V Sreelatha
- Department of Oral and Maxillofacial Pathology and Oral Microbiology, AB Shetty Memorial Institute of Dental Sciences (ABSMIDS), Nitte (Deemed to be University), Deralakatte, Mangalore, Karnataka, India
| | - Sukanya Shetty
- Department of Biochemistry, KS Hegde Medical Academy (KSHEMA), Nitte (Deemed to be University), Deralakatte, Mangalore, Karnataka, India
| | - Vimal K Karnaker
- Department of Microbiology, KS Hegde Medical Academy (KSHEMA), Nitte (Deemed to be University), Deralakatte, Mangalore, Karnataka, India
| | - Ankeeta M Jacob
- Community Medicine, KS Hegde Medical Academy (KSHEMA), Nitte (Deemed to be University), Deralakatte, Mangalore, Karnataka, India
| | - Chitta R Chowdhury
- The University of Bolton City of London Dental School, Southgate Dental Science Campus, London, UK
| |
Collapse
|
5
|
Kim H, Lee JK, Oh AC, Kim HR, Hong YJ. The Usefulness of the Ratio of Antigen-Autoantibody Immune Complexes to Their Free Antigens in the Diagnosis of Non-Small Cell Lung Cancer. Diagnostics (Basel) 2023; 13:2999. [PMID: 37761366 PMCID: PMC10529727 DOI: 10.3390/diagnostics13182999] [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: 06/19/2023] [Revised: 08/21/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Autoantibodies against specific lung cancer-associated antigens have been suggested for the performance of lung cancer diagnosis. This study aimed to evaluate the diagnostic performance of the antigen-autoantibody immune complex (AIC) against its free antigens for CYFRA21-1, ProGRP, neutrophil gelatinase-associated lipocalin (NGAL), and neuron-specific enolase (NSE) in non-small cell lung cancer (NSCLC). In total, 85 patients with NSCLC and 120 healthy controls (HCs) were examined using a 9-guanine DNA chip method. The ratios of AICs to their antigens and the combinations of ratios consisting of two to four markers were calculated. The levels of AICs for CYFRA21-1, ProGRP, NGAL, and NSE were higher than those for their free antigens in all participants. The levels of each free antigens distinguished patients with NSCLC from the HCs. The ratios of the AIC to its antigen and seven combinations of two to four ratios were significantly higher in patients with NSCLC than in the HCs. Excellent diagnostic performance was observed for all combination ratios (C4-1), with 85.9% sensitivity and 86.7% specificity at a 3.51 cut-off. Higher sensitivity was observed in the early stages (0-I) and adenocarcinoma than in stages II-IV and other pathological types. Combining all ratios of AICs and their antigens for all four markers was useful when diagnosing NSCLC.
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.)
| | - Ae-Chin Oh
- 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
|
6
|
Gautam SK, Khan P, Natarajan G, Atri P, Aithal A, Ganti AK, Batra SK, Nasser MW, Jain M. Mucins as Potential Biomarkers for Early Detection of Cancer. Cancers (Basel) 2023; 15:1640. [PMID: 36980526 PMCID: PMC10046558 DOI: 10.3390/cancers15061640] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/25/2023] [Accepted: 02/27/2023] [Indexed: 03/10/2023] Open
Abstract
Early detection significantly correlates with improved survival in cancer patients. So far, a limited number of biomarkers have been validated to diagnose cancers at an early stage. Considering the leading cancer types that contribute to more than 50% of deaths in the USA, we discuss the ongoing endeavors toward early detection of lung, breast, ovarian, colon, prostate, liver, and pancreatic cancers to highlight the significance of mucin glycoproteins in cancer diagnosis. As mucin deregulation is one of the earliest events in most epithelial malignancies following oncogenic transformation, these high-molecular-weight glycoproteins are considered potential candidates for biomarker development. The diagnostic potential of mucins is mainly attributed to their deregulated expression, altered glycosylation, splicing, and ability to induce autoantibodies. Secretory and shed mucins are commonly detected in patients' sera, body fluids, and tumor biopsies. For instance, CA125, also called MUC16, is one of the biomarkers implemented for the diagnosis of ovarian cancer and is currently being investigated for other malignancies. Similarly, MUC5AC, a secretory mucin, is a potential biomarker for pancreatic cancer. Moreover, anti-mucin autoantibodies and mucin-packaged exosomes have opened new avenues of biomarker development for early cancer diagnosis. In this review, we discuss the diagnostic potential of mucins in epithelial cancers and provide evidence and a rationale for developing a mucin-based biomarker panel for early cancer detection.
Collapse
Affiliation(s)
- Shailendra K. Gautam
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Parvez Khan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Gopalakrishnan Natarajan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Pranita Atri
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Abhijit Aithal
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Apar K. Ganti
- Fred & Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Division of Oncology-Hematology, Department of Internal Medicine, VA Nebraska Western Iowa Health Care System, University of Nebraska Medical Center, Omaha, NE 68105, USA
| | - Surinder K. Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Mohd W. Nasser
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
7
|
Cancer Is Associated with the Emergence of Placenta-Reactive Autoantibodies. Biomedicines 2023; 11:biomedicines11020316. [PMID: 36830854 PMCID: PMC9953527 DOI: 10.3390/biomedicines11020316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 01/25/2023] Open
Abstract
Placenta-specific antigens are minimally expressed or unexpressed in normal adult tissues, while they are widely expressed in cancer. In the course of carcinogenesis, a vast array of autoantibodies (AAbs) is produced. Here, we used a quantitative approach to determine the reactivity of AAbs in the sera of patients with breast (BrC: N = 100, 100% female, median age: 51 years), gastric (GC: N = 30, 46.6% female, median age: 57 years), bladder (BC: N = 29, 34.4% female, median age: 57 years), and colorectal (CRC: N = 34, 41.1% female, median age: 51 years) cancers against first-trimester (FTP) and full-term placental proteome (TP) in comparison with age- and sex-matched non-cancer individuals. Human-on-human immunohistochemistry was used to determine reactive target cells in FTP. The effect of pregnancy on the emergence of placenta-reactive autoantibodies was tested using sera from pregnant women at different trimesters of pregnancy. Except for BC, patients with BrC (p < 0.0284), GC (p < 0.0002), and CRC (p < 0.0007) had significantly higher levels of placenta-reactive AAbs. BrC (p < 0.0001) and BC (p < 0.0409) in the early stages triggered higher autoantibody reactivity against FTP. The reactivities of BrC sera with FTP did not show an association with ER, PR, or HER2 expression. Pregnancy in the third trimester was associated with the induction of TP- and not FTP-reactive autoantibodies (=0.018). The reactivity of BrC sera with placental proteins was found to be independent of gravidity or abortion. BrC sera showed a very strong and specific pattern of reactivity with scattered cells beneath the syncytiotrophoblast layer. Our results reinforce the concept of the coevolution of placentation and cancer and shed light on the future clinical application of the placental proteome for the non-invasive early detection and treatment of cancer.
Collapse
|
8
|
Taylor J, Gandhi A, Gray E, Zaenker P. Checkpoint inhibitor immune-related adverse events: A focused review on autoantibodies and B cells as biomarkers, advancements and future possibilities. Front Immunol 2023; 13:991433. [PMID: 36713389 PMCID: PMC9874109 DOI: 10.3389/fimmu.2022.991433] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 12/07/2022] [Indexed: 01/13/2023] Open
Abstract
The use of immune checkpoint inhibitors (ICIs) has evolved rapidly with unprecedented treatment benefits being obtained for cancer patients, including improved patient survival. However, over half of the patients experience immune related adverse events (irAEs) or toxicities, which can be fatal, affect the quality of life of patients and potentially cause treatment interruption or cessation. Complications from these toxicities can also cause long term irreversible organ damage and other chronic health conditions. Toxicities can occur in various organ systems, with common observations in the skin, rheumatologic, gastrointestinal, hepatic, endocrine system and the lungs. These are not only challenging to manage but also difficult to detect during the early stages of treatment. Currently, no biomarker exists to predict which patients are likely to develop toxicities from ICI therapy and efforts to identify robust biomarkers are ongoing. B cells and antibodies against autologous antigens (autoantibodies) have shown promise and are emerging as markers to predict the development of irAEs in cancer patients. In this review, we discuss the interplay between ICIs and toxicities in cancer patients, insights into the underlying mechanisms of irAEs, and the involvement of the humoral immune response, particularly by B cells and autoantibodies in irAE development. We also provide an appraisal of the progress, key empirical results and advances in B cell and autoantibody research as biomarkers for predicting irAEs. We conclude the review by outlining the challenges and steps required for their potential clinical application in the future.
Collapse
Affiliation(s)
- John Taylor
- Centre for Precision Health, Edith Cowan University, Joondalup, WA, Australia,School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia,*Correspondence: John Taylor,
| | - Aesha Gandhi
- Sir Charles Gairdner Hospital, Department of Medical Oncology, Nedlands, WA, Australia
| | - Elin Gray
- Centre for Precision Health, Edith Cowan University, Joondalup, WA, Australia,School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Pauline Zaenker
- Centre for Precision Health, Edith Cowan University, Joondalup, WA, Australia,School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| |
Collapse
|
9
|
Kathrikolly T, Nair SN, Mathew A, Saxena PPU, Nair S. Can serum autoantibodies be a potential early detection biomarker for breast cancer in women? A diagnostic test accuracy review and meta-analysis. Syst Rev 2022; 11:215. [PMID: 36210467 PMCID: PMC9549667 DOI: 10.1186/s13643-022-02088-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 09/28/2022] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND The increasing incidence of breast cancer necessitates the need to explore alternate screening strategies that circumvent the setbacks of conventional techniques especially among population that report earlier age at diagnosis. Serum autoantibodies is one such potential area of interest. However, their ubiquitous presence across cancer types limits its applicability to any one specific type of cancer. This review was therefore carried out to explore and consolidate available evidence on autoantibodies for early detection of breast cancer and to identify those that demonstrated a higher sensitivity. METHODS A diagnostic test accuracy (DTA) review was carried out to ascertain serum autoantibodies that could be used for early detection of breast cancer among women. All relevant articles that investigated the role of autoantibodies in early detection of breast cancer were included for the review. MEDLINE, Scopus, ProQuest, Ovid SP, and Cochrane Library were searched extensively for eligible studies. Quality of the included studies was assessed using Quality Assessment of Diagnostic Accuracy Studies (QUADAS)-2 tool. RevMan 5.3 was used for exploratory and MetaDTA 2019 for hierarchical analyses. The review helped identify the most frequently investigated autoantibodies and a meta-analysis further consolidated the findings. RESULTS A total of 53 articles were included for the final analysis that reported over a 100 autoantibodies that were studied for early detection of breast cancer in women. P53, MUC1, HER2, HSP60, P16, Cyclin B1, and c-Myc were the most frequently investigated autoantibodies. Of these P53, MUC1, HER2, and HSP60 exhibited higher summary sensitivity measures. While the individual pooled sensitivity estimates ranged between 10 and 56%, the panel sensitivity values reported across studies were higher with an estimated range of 60-87%. CONCLUSION Findings from the review indicate a higher sensitivity for an autoantibody panel in comparison to individual assays. A panel comprising of P53, MUC1, HER2, and HSP60 autoantibodies has the potential to be investigated as an early detection biomarker for breast cancer.
Collapse
Affiliation(s)
- Thejas Kathrikolly
- Department of Community Oncology, Sri Shankara Cancer Hospital and Research Centre, Bengaluru, India.,Department of Community Medicine, Kasturba Medical College, Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Sreekumaran N Nair
- Department of Biostatistics, Jawaharlal Institute of Postgraduate Medical Education & Research (JIPMER), Puducherry, India
| | - Aju Mathew
- Department of Oncology, MOSC Medical College Kolenchery, Kerala, India.,Department of Internal Medicine, University of Kentucky Markey Cancer Center, Lexington, USA
| | - Prakash P U Saxena
- Department of Radiation Oncology, Kasturba Medical College, Mangalore, India
| | - Suma Nair
- Department of Community Medicine, Kasturba Medical College, Manipal Academy of Higher Education (MAHE), Manipal, India. .,School of Public Health, DY Patil Deemed to be University, Navi Mumbai, India.
| |
Collapse
|
10
|
Tsai IJ, Shen WC, Wu JZ, Chang YS, Lin CY. Autoantibodies to Oxidatively Modified Peptide: Potential Clinical Application in Coronary Artery Disease. Diagnostics (Basel) 2022; 12:diagnostics12102269. [PMID: 36291959 PMCID: PMC9600024 DOI: 10.3390/diagnostics12102269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 09/11/2022] [Accepted: 09/19/2022] [Indexed: 11/16/2022] Open
Abstract
Coronary artery disease (CAD) is a global health issue. Lipid peroxidation produces various by-products that associate with CAD, such as 4-hydroxynonenal (HNE) and malondialdehyde (MDA). The autoantibodies against HNE and MDA-modified peptides may be useful in the diagnosis of CAD. This study included 41 healthy controls (HCs) and 159 CAD patients with stenosis rates of <30%, 30−70%, and >70%. The plasma level of autoantibodies against four different unmodified and HNE-modified peptides were measured in this study, including CFAH1211−1230, HPT78−108, IGKC2−19, and THRB328−345. Furthermore, feature ranking, feature selection, and machine learning models have been utilized to exploit the diagnostic performance. Also, we combined autoantibodies against MDA and HNE-modified peptides to improve the models’ performance. The eXtreme Gradient Boosting (XGBoost) model received a sensitivity of 78.6% and a specificity of 90.4%. Our study demonstrated the combination of autoantibodies against oxidative modification may improve the model performance.
Collapse
Affiliation(s)
- I-Jung Tsai
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Wen-Chi Shen
- Institute of Biotechnology, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Jia-Zhen Wu
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Yu-Sheng Chang
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Shuang Ho Hospital, New Taipei City 23561, Taiwan
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Correspondence: (Y.-S.C.); (C.-Y.L.); Tel.: +886-2-22490088 (Y.-S.C.); +886-2-27361661 (ext. 3326) (C.-Y.L.)
| | - Ching-Yu Lin
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Correspondence: (Y.-S.C.); (C.-Y.L.); Tel.: +886-2-22490088 (Y.-S.C.); +886-2-27361661 (ext. 3326) (C.-Y.L.)
| |
Collapse
|
11
|
Li J, Dai L, Huang M, Ma Y, Guo Z, Wang X, Li W, Zhang JY. Immunoseroproteomic profiling in autoantibody to ENO1 as potential biomarker in immunodiagnosis of osteosarcoma by serological proteome analysis (SERPA) approach. Oncoimmunology 2021; 10:1966969. [PMID: 38260036 PMCID: PMC10802918 DOI: 10.1080/2162402x.2021.1966969] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 08/06/2021] [Indexed: 12/19/2022] Open
Abstract
Osteosarcoma (OS) is the most common highly malignant primary solid bone tumor. Despite its relatively low incidence among cancers, it remains one of the most harmful primary malignant tumors in childhood and adolescence. It is now evident that serum autoantibodies against tumor-associated antigens (TAAs) could be used as serological cancer biomarkers in types of cancers. Serological proteome analysis (SERPA) approach was applied to profile anti-TAA autoantibody response in sera from patients with OS and normal human, as well as explore difference between this response. This approach can detect autoantibodies that could serve as clinical biomarkers and immunotherapeutic agents. Enzyme-linked immunosorbent assay (ELISA) and Western blotting were further used to validate the level of identified TAAs. ENO1 as a 47kD TAA in OS was identified and characterized by SERPA. Analysis of 172 serum samples with OS, osteochondroma (OC), and normal human sera (NHS) by ELISA showed higher frequency of anti-ENO1 autoantibodies in OS sera compared to others. Interestingly, decrease of ENO1 immunoreactivity was observed in most patients after treatments, which may imply a potential association between anti-ENO1 autoantibody titers and disease progression. Nine of twelve sera reacted strongly against purified ENO1, but three reacted weakly against purified ENO1, which indicated 75.0% sera with positive optimal density values from ELISA were consistently positive in Western blotting. The expression of ENO1 in OS tissues was evaluated by immunohistochemistry in tumor microarray. ENO1 was one of the autoantibodies that elicit autoimmune responses in OS and can be used as biomarkers in immunodiagnosis and progression of OS.
Collapse
Affiliation(s)
- Jitian Li
- Department of Biological Sciences & NIH-sponsored Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX, USA
- Laboratory of Molecular Biology, Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital)/Henan Institute of Orthopedic and Traumatology, Luoyang, Henan, China
| | - Liping Dai
- Department of Biological Sciences & NIH-sponsored Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX, USA
| | - Manyu Huang
- Laboratory of Molecular Biology, Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital)/Henan Institute of Orthopedic and Traumatology, Luoyang, Henan, China
| | - Yan Ma
- Laboratory of Molecular Biology, Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital)/Henan Institute of Orthopedic and Traumatology, Luoyang, Henan, China
| | - Zhiping Guo
- Laboratory of Molecular Biology, Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital)/Henan Institute of Orthopedic and Traumatology, Luoyang, Henan, China
| | - Xiao Wang
- Department of Biological Sciences & NIH-sponsored Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX, USA
| | - Wuyin Li
- Laboratory of Molecular Biology, Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital)/Henan Institute of Orthopedic and Traumatology, Luoyang, Henan, China
| | - Jian-Ying Zhang
- Department of Biological Sciences & NIH-sponsored Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX, USA
| |
Collapse
|
12
|
Li J, Qin B, Huang M, Ma Y, Li D, Li W, Guo Z. Tumor-Associated Antigens (TAAs) for the Serological Diagnosis of Osteosarcoma. Front Immunol 2021; 12:665106. [PMID: 33995397 PMCID: PMC8119874 DOI: 10.3389/fimmu.2021.665106] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 04/08/2021] [Indexed: 01/07/2023] Open
Abstract
Osteosarcoma (OS) is the most common form of malignant bone tumor found in childhood and adolescence. Although its incidence rate is low among cancers, the prognosis of OS is usually poor. Although some biomarkers, such as p53, have been identified in OS, the association between the biomarkers and clinical outcome is not well understood. Thus, it is necessary to establish a method to identify patients diagnosed with OS at an early stage. It is becoming obvious that anti-tumor-associated antigens (TAAs) autoantibodies (TAAbs) in sera could be used as serological biomarkers in the detection of many different types of cancers. This notion indicates that TAAbs are considered as immunological “sentinels” associated with tumorigenesis underlying molecular events. It provides new insights into the molecular and cellular biology of the differential diagnosis of cancers. What’s more, it is reported that a customized TAA array could significantly increase the sensitivity/specificity. TAA arrays also have great application prospects in detecting cancer at an early stage, monitoring cancer progression, discovering new therapeutic targets, and designing personalized treatment. In this review, we provide an overview of the TAAs identified in OS as well as the possibility that TAAs and TAAbs system be used as biomarkers in the immunodiagnosis and prognosis of OS.
Collapse
Affiliation(s)
- Jitian Li
- Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital)/Henan Institute of Orthopedic and Traumatology, Luoyang, China
| | - Bo Qin
- Transitional Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Manyu Huang
- Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital)/Henan Institute of Orthopedic and Traumatology, Luoyang, China
| | - Yan Ma
- Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital)/Henan Institute of Orthopedic and Traumatology, Luoyang, China
| | - Dongsheng Li
- Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital)/Henan Institute of Orthopedic and Traumatology, Luoyang, China
| | - Wuyin Li
- Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital)/Henan Institute of Orthopedic and Traumatology, Luoyang, China
| | - Zhiping Guo
- Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital)/Henan Institute of Orthopedic and Traumatology, Luoyang, China
| |
Collapse
|
13
|
Rashid F, Saeed A, Iqbal J. In Vitro Anticancer Effects of Stilbene Derivatives: Mechanistic Studies on HeLa and MCF-7 Cells. Anticancer Agents Med Chem 2021; 21:793-802. [PMID: 32781966 DOI: 10.2174/1871520620666200811123230] [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: 02/13/2020] [Revised: 04/14/2020] [Accepted: 05/10/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND AND OBJECTIVE The growing prevalence of cancer and the resulting chemoresistance exert a huge burden on healthcare systems and impose a great challenge to public health around the world. In efforts to develop new chemotherapeutic agents for cancer treatment, a class of heterocyclic compounds i.e. triazine-based molecules were investigated as anticancer agents. MATERIALS AND METHODS New triazine hybrids of stilbene were synthesized and evaluated as anticancer agents for cervical (HeLa) and breast (MCF-7) carcinoma cells. The compound (7e), sodium (E)-6,6'-(ethene-1,2- diyl)bis(3-((4-chloro-6-((3-luorophenyl)amino)-1,3,5-triazin-2-yl)amino)benzenesulfonate) was found to be most potent among synthesized derivatives and was explored further for detailed mechanistic studies. RESULTS In a set comprised of twelve derivatives, compound 7e, sodium (E)-6,6'-(ethene-1,2-diyl)bis(3-((4- chloro-6-((3-luorophenyl)amino)-1,3,5-triazin-2-yl)amino)benzenesulfonate) was found most potent inhibitor for HeLa and MCF-7 cells. DISCUSSION The present study has revealed that compound 7e may activate mitochondrial pathway of apoptosis in HeLa and MCF-7 cells which was assessed by DNA binding studies, estimation of the release of Lactate Dehydrogenase (LDH), fluorescence imaging, production of Reactive Oxygen Species (ROS) in cancer cells, analysis of cell cycle by flow cytometry, change in Mitochondrial Membrane Potential (MMP) and activation of caspase-9 and caspase-3. CONCLUSION Compound 7e may serve as a lead in designing new anticancer compounds based on stilbene scaffold.
Collapse
Affiliation(s)
- Faisal Rashid
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad-22060, Pakistan
| | - Aamer Saeed
- Department of Chemistry, Quaid-i-Azam University, Islamabad, Pakistan
| | - Jamshed Iqbal
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad-22060, Pakistan
| |
Collapse
|
14
|
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]
|
15
|
Xu ML, Kim HJ, Kim SC, Ju W, Kim YH, Chang KH, Kim HJ. Serum anti-GAPDH autoantibody levels reflect the severity of cervical lesions: A potential serum biomarker for cervical cancer screening. Oncol Lett 2019; 18:255-264. [PMID: 31289495 PMCID: PMC6539791 DOI: 10.3892/ol.2019.10326] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 04/17/2019] [Indexed: 12/22/2022] Open
Abstract
Recent studies have indicated that a certain level of autoantibodies may be essential for maintaining good health as well as preventing cancer development, and that the levels of serum autoantibodies can decline during malignant progression. The aim of the present study was to identify such an autoantibody-based biomarker for screening cervical lesions. An autoantigen reactive with healthy female sera was detected in the cytosolic fraction of HeLa cells, a cervical cancer cell line, and identified. Serum immunoglobulin (Ig)-G and IgM levels against the purified autoantigen in normal, cervical intraepithelial neoplasias (CINs) I, II and III, and cervical cancer were compared using ELISAs. The autoantigen in HeLa cells was identified to be GAPDH. The serum levels of anti-HeLa-GAPDH IgG decreased with increasing severity of cervical lesions, and similar decreases in IgM levels were revealed. Notably, the anti-HeLa-GAPDH IgG level was discovered to discriminate cervical cancer from normal samples with 80.0% sensitivity and 96.6% specificity. The serum anti-HeLa-GAPDH autoantibody level, as a single parameter, is a promising serum biomarker for screening cervical lesions.
Collapse
Affiliation(s)
- Mei Ling Xu
- Laboratory of Virology, College of Pharmacy, Chung-Ang University, Dongjak-Gu, Seoul 06974, Republic of Korea
| | - Hyoung Jin Kim
- Laboratory of Virology, College of Pharmacy, Chung-Ang University, Dongjak-Gu, Seoul 06974, Republic of Korea
| | - Seung Cheol Kim
- Department of Obstetrics and Gynecology, Ewha Woman's University College of Medicine, Yangcheon-Gu, Seoul 03760, Republic of Korea
| | - Woong Ju
- Department of Obstetrics and Gynecology, Ewha Woman's University College of Medicine, Yangcheon-Gu, Seoul 03760, Republic of Korea
| | - Yun Hwan Kim
- Department of Obstetrics and Gynecology, Ewha Woman's University College of Medicine, Yangcheon-Gu, Seoul 03760, Republic of Korea
| | - Kyu-Ho Chang
- Department of Biological Engineering, Inha University, Incheon 22212, Republic of Korea
| | - Hong-Jin Kim
- Laboratory of Virology, College of Pharmacy, Chung-Ang University, Dongjak-Gu, Seoul 06974, Republic of Korea
| |
Collapse
|
16
|
Music M, Soosaipillai A, Batruch I, Prassas I, Bogdanos DP, Diamandis EP. A proteome-wide immuno-mass spectrometric identification of serum autoantibodies. Clin Proteomics 2019; 16:25. [PMID: 31249498 PMCID: PMC6585069 DOI: 10.1186/s12014-019-9246-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 06/11/2019] [Indexed: 11/17/2022] Open
Abstract
Background Autoantibodies are produced when tolerance to self-antigens is broken and they can be mediators of tissue injury and systemic inflammation. They are excellent biomarkers because they are minimally invasive to screen and are highly abundant in serum due to limited proteolysis and slow clearance. Conventionally used methods of identifying autoantibodies in patient sera include indirect immunofluorescence, enzyme-linked immunoabsorbent assays (ELISAs) and protein microarrays. Here we present a novel proteome-wide immuno-mass spectrometric method to identify serum autoantibody targets. Methods Serum samples from patients with inflammatory bowel disease (IBD) were analyzed by ELISA for the presence of autoantibodies to CUB and zona pellucida-like domain-containing protein 1 (CUZD1). Protein was extracted from the human pancreas as well as 16 other human tissues to make a complex tissue lysate protein mixture. Antibodies in patient sera were immobilized and purified on protein G magnetic beads and subsequently incubated with pancreatic lysate containing CUZD1 or the aforementioned complex tissue lysate. After extensive washing, antibody-bound protein antigens were trypsin-digested and identified using shotgun mass spectrometry. Results The protocol was optimized for the immunoaffinity purification of autoantibody targets from tissue lysate, using CUZD1 from pancreatic lysate and anti-CUZD1 autoantibodies present in IBD patient serum as a proof-of-concept. Pancreatic secretory granule membrane major glycoprotein 2, whose autoantibodies are a known biomarker of Crohn’s disease, was also immunoprecipitated from IBD patient serum, as an additional internal positive control. Conclusions This study demonstrates the effectiveness of a proteomic approach to identify serum autoantibody targets, using immunoaffinity purification followed by tandem mass spectrometry. Our methodology is applicable for proteome-wide analysis of autoantibody targets in a wide variety of clinical settings. Electronic supplementary material The online version of this article (10.1186/s12014-019-9246-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Milena Music
- 1Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Antoninus Soosaipillai
- 2Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, 60 Murray St [Box 32]; Flr 6 - Rm L6-201-1, Toronto, ON M5T 3L9 Canada
| | - Ihor Batruch
- 2Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, 60 Murray St [Box 32]; Flr 6 - Rm L6-201-1, Toronto, ON M5T 3L9 Canada
| | - Ioannis Prassas
- 2Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, 60 Murray St [Box 32]; Flr 6 - Rm L6-201-1, Toronto, ON M5T 3L9 Canada
| | - Dimitrios P Bogdanos
- 3Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, 41110 Larissa, Greece
| | - Eleftherios P Diamandis
- 1Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.,2Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, 60 Murray St [Box 32]; Flr 6 - Rm L6-201-1, Toronto, ON M5T 3L9 Canada.,4Department of Clinical Biochemistry, University Health Network, Toronto, Canada.,5Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 60 Murray St [Box 32]; Flr 6 - Rm L6-201-1, Toronto, ON M5T 3L9 Canada
| |
Collapse
|
17
|
Baldin AV, Grishina AN, Korolev DO, Kuznetsova EB, Golovastova MO, Kalpinskiy AS, Alekseev BY, Kaprin AD, Zinchenko DV, Savvateeva LV, Varshavsky VA, Zernii EY, Vinarov AZ, Bazhin AV, Philippov PP, Zamyatnin AA. Autoantibody against arrestin-1 as a potential biomarker of renal cell carcinoma. Biochimie 2018; 157:26-37. [PMID: 30389514 DOI: 10.1016/j.biochi.2018.10.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 10/25/2018] [Indexed: 12/28/2022]
Abstract
Renal cell carcinoma (RCC) is the second-most common uronephrological cancer. In the absence of specific symptoms, early diagnosis of RCC is challenging. Monitoring of the aberrant expression of tumour-associated antigens (TAAs) and related autoantibody response is considered as a novel approach of RCC diagnostics. The aim of this study was to examine the aberrant expression of arrestin-1 in renal tumours, to investigate the possible epigenetic mechanism underlying arrestin-1 expression, and to assess the frequency of anti-arrestin-1 autoantibody response. Immunohistochemistry was used to assess the presence of arrestin-1 in primary tumours and metastases of 39 patients with RCC and renal oncocytoma. Bisulfite sequencing was employed to analyse the methylation status of the promoter of the SAG gene encoding arrestin-1. Western blot analysis was performed to detect autoantibodies against arrestin-1 in serum samples of 36 RCC and oncocytoma patients. Arrestin-1 was found to be expressed in RCC (58.7% of cases) and renal oncocytoma (90% of cases) cells, while being absent in healthy kidney. The expression of arrestin-1 in RCC metastases was more prominent than in primary tumours. Hypomethylation of the SAG gene promoter is unlikely to be the mechanism for the aberrant expression of arrestin-1. Autoantibodies against arrestin-1 were detected in sera of 75% of RCC patients. Taken together, our findings suggest employment of autoantibody against arrestin-1 as biomarker of RCC.
Collapse
Affiliation(s)
- Alexey V Baldin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991, Moscow, Russia.
| | - Alena N Grishina
- Anatomic Pathology Department, Sechenov First Moscow State Medical University, 119991, Moscow, Russia.
| | - Dmitry O Korolev
- Institute of Uronephrology and Human Reproductive Health, Sechenov First Moscow State Medical University, 119991, Moscow, Russia
| | - Ekaterina B Kuznetsova
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991, Moscow, Russia; Research Centre for Medical Genetics, 115522, Moscow, Russia.
| | - Marina O Golovastova
- Department of Cell Signalling, Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991, Moscow, Russia
| | - Alexey S Kalpinskiy
- P.A. Hertzen Moscow Oncology Research Center, National Medical Research Center of Radiology, 125284, Moscow, Russia
| | - Boris Y Alekseev
- P.A. Hertzen Moscow Oncology Research Center, National Medical Research Center of Radiology, 125284, Moscow, Russia
| | - Andrey D Kaprin
- P.A. Hertzen Moscow Oncology Research Center, National Medical Research Center of Radiology, 125284, Moscow, Russia.
| | - Dmitry V Zinchenko
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Pushchino, Moscow Region, 142290 Russia.
| | - Lyudmila V Savvateeva
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991, Moscow, Russia
| | - Vladimir A Varshavsky
- Anatomic Pathology Department, Sechenov First Moscow State Medical University, 119991, Moscow, Russia
| | - Evgeni Yu Zernii
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991, Moscow, Russia; Department of Cell Signalling, Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991, Moscow, Russia.
| | - Andrey Z Vinarov
- Institute of Uronephrology and Human Reproductive Health, Sechenov First Moscow State Medical University, 119991, Moscow, Russia.
| | - Alexandr V Bazhin
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany.
| | - Pavel P Philippov
- Department of Cell Signalling, Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991, Moscow, Russia
| | - Andrey A Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991, Moscow, Russia; Department of Cell Signalling, Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991, Moscow, Russia.
| |
Collapse
|
18
|
Mohsenzadegan M, Saebi F, Yazdani M, Abolhasani M, Saemi N, Jahanbani F, Farajollahi MM. Autoantibody against new gene expressed in prostate protein is traceable in prostate cancer patients. Biomark Med 2018; 12:1125-1138. [DOI: 10.2217/bmm-2018-0069] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Aim: We assessed an autoantibody against new gene expressed in prostate (NGEP) protein for prostate cancer (PCa) that may better diagnosis and prognosis approaches in the patients with PCa. Methods: Autoantibodies against NGEP were measured in sera of PCa patients by ELISA. Results: The autoantibody against NGEP is present in a significantly higher proportion in the sera of PCa patients as compared with healthy controls (p < 0.001). An inverse significant correlation was found between seropositive patients and Gleason score (p < 0.05) and serum prostate-specific antigen (recombinant NGEP; p < 0.05). Conclusion: The data showed that measurement of autoantibody against NGEP as a novel prostate-specific antigen in sera can be used as a potential biomarker to discriminate well-differentiated PCa patients from normal subjects.
Collapse
Affiliation(s)
- Monireh Mohsenzadegan
- Cellular & Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Laboratory Science, Faculty of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Saebi
- Cellular & Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mehdi Yazdani
- Hasheminejad Kidney Center Laboratory, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Abolhasani
- Hasheminejad Kidney Center Laboratory, Iran University of Medical Sciences, Tehran, Iran
- Oncopathology Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Nazanin Saemi
- Department of Medical Biotechnology, Faculty of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Jahanbani
- Department of Genetic, Standford University School of Medicine, CA, USA
| | - Mohammad M Farajollahi
- Cellular & Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Biotechnology, Faculty of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
The Thomsen-Friedenreich Antigen: A Highly Sensitive and Specific Predictor of Microsatellite Instability in Gastric Cancer. J Clin Med 2018; 7:jcm7090256. [PMID: 30189652 PMCID: PMC6162870 DOI: 10.3390/jcm7090256] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 08/30/2018] [Indexed: 02/07/2023] Open
Abstract
Microsatellite instability (MSI) is a distinct molecular subtype of gastric cancer. In recent years, the clinical consequences of MSI and the therapeutic opportunities to target this peculiar cancer subtype became evident. However, despite the importance of MSI for the stratification of patients, the time and resources required for diagnosis still present an obstacle. In an attempt to identify a new marker for MSI in gastric cancer, we evaluated the expression of five cancer-associated glycan epitopes in a cohort of 13 MSI and 17 microsatellite stable (MSS) cases. Our analysis revealed a highly significant (p < 0.001) association between the expression of the Thomsen-Friedenreich (TF) antigen and MSI status. Hence, we present here the identification of the first single marker for MSI in gastric cancer, excelling with a specificity of 94% (16/17), sensitivity of 69.2% (9/13), negative predictive value of 80% (16/20), and positive predictive value of 90% (9/10). The TF antigen, detected by simple antibody-based assays, is highly specific for carcinoma being undetectable in gastric healthy and premalignant epithelia. This finding lays the basis for new studies and holds promise in improving the rapid identification of MSI in the clinical setting.
Collapse
|
20
|
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
|
21
|
Mustafa MZ, Nguyen VH, Le Naour F, De Martin E, Beleoken E, Guettier C, Johanet C, Samuel D, Duclos-Vallee JC, Ballot E. Autoantibody signatures defined by serological proteome analysis in sera from patients with cholangiocarcinoma. J Transl Med 2016; 14:17. [PMID: 26774260 PMCID: PMC4715332 DOI: 10.1186/s12967-015-0751-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 12/08/2015] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND The challenging diagnosis and poor prognosis of cholangiocarcinoma require the determination of biomarkers. Autoantibodies could be used in the clinic as diagnostic markers for the early detection of tumours. By proteomic approaches, several autoantibodies were proposed as potential markers. We tried in this study, to perform a serological proteome analysis, using various antigenic substrates, including tumours and human liver. METHODS Sera from patients (n = 13) and healthy donors (n = 10) were probed on immunoblots performed using 2-dimensionally separated proteins from cholangiocarcinoma cell lines (CCLP1 and CCSW1), from the liver of healthy subject and interestingly, from tumour and adjacent non-tumour liver tissues from five patients with cholangiocarcinoma and tested with their corresponding serum. Spots of interest were identified using mass spectrometry and classified according gene ontology analysis. RESULTS A comparison of the whole immunoblotting patterns given by cholangiocarcinoma sera against those obtained with normal control sera enabled the definition of 862 spots. Forty-five different proteins were further analysed, corresponding to (1) spots stained with more than four of 13 (30 %) sera tested with the CCLP1 or the CCSW1 cell line and with the normal liver, and (2) to spots immunoreactive with at least two of the five sera probed with their tumour and non-tumour counter-part of cholangiocarcinoma. Immunoreactive proteins with catalytic activity as molecular function were detected at rates of 93 and 64 % in liver from healthy subjects or cholangiocarcinoma non-tumour tissues respectively, compared to 43, 33, 33 % in tumour tissues, or CCSW1 and CCLP1 cell lines. A second pattern was represented by structural proteins with rates of 7 and 7 % in normal liver or non-tumour tissues compared to 14, 33 and 67 % in tumour tissue, CCSW1 or CCLP1 cell lines. Proteins with a binding function were detected at rates of 7 % in non-tumour tissue and 14 % in tumour tissue. Using the extracted tumour tissue, serotransferrin was targeted by all cholangiocarcinoma-related sera. CONCLUSIONS Immunological patterns depended on the type of antigen substrate used; i.e. tumour versus non tumour specimens. Nevertheless, a combination of multiple autoantibodies tested with the most appropriate substrate might be more sensitive and specific for the diagnosis of cholangiocarcinoma.
Collapse
Affiliation(s)
- Mohammad Zahid Mustafa
- Inserm, Unité 1193, Université Paris-Saclay, 94800, Villejuif, France.
- Univ Paris-Sud, UMR-S 1193, Université Paris-Saclay, 94800, Villejuif, France.
- CASVAB, University of Balochistan, Quetta, Pakistan.
- DHU Hepatinov, 94800, Villejuif, France.
| | - Viet Hung Nguyen
- Inserm, Unité 1193, Université Paris-Saclay, 94800, Villejuif, France.
- Univ Paris-Sud, UMR-S 1193, Université Paris-Saclay, 94800, Villejuif, France.
| | - François Le Naour
- Inserm, Unité 1193, Université Paris-Saclay, 94800, Villejuif, France.
- Univ Paris-Sud, UMR-S 1193, Université Paris-Saclay, 94800, Villejuif, France.
- DHU Hepatinov, 94800, Villejuif, France.
| | - Eleonora De Martin
- Inserm, Unité 1193, Université Paris-Saclay, 94800, Villejuif, France.
- Univ Paris-Sud, UMR-S 1193, Université Paris-Saclay, 94800, Villejuif, France.
- Centre Hépato-Biliaire, AP-HP Hôpital Paul-Brousse, 94800, Villejuif, France.
- DHU Hepatinov, 94800, Villejuif, France.
| | - Elvire Beleoken
- Inserm, Unité 1193, Université Paris-Saclay, 94800, Villejuif, France.
- Univ Paris-Sud, UMR-S 1193, Université Paris-Saclay, 94800, Villejuif, France.
- DHU Hepatinov, 94800, Villejuif, France.
| | - Catherine Guettier
- Inserm, Unité 1193, Université Paris-Saclay, 94800, Villejuif, France.
- Univ Paris-Sud, UMR-S 1193, Université Paris-Saclay, 94800, Villejuif, France.
- DHU Hepatinov, 94800, Villejuif, France.
- Laboratoire Anatomie Pathologique, AP-HP Hôpital Bicêtre, 94270, Le Kremlin-Bicêtre, France.
| | - Catherine Johanet
- Inserm, Unité 1193, Université Paris-Saclay, 94800, Villejuif, France.
- Univ Paris-Sud, UMR-S 1193, Université Paris-Saclay, 94800, Villejuif, France.
- Département d'Immunologie Biologique, Unité d'autoimmunité, AP-HP Hôpital Saint Antoine, 75012, Paris, France.
- UFR 967 Faculté de Médecine, Université Pierre et Marie Curie, 75006, Paris, France.
| | - Didier Samuel
- Inserm, Unité 1193, Université Paris-Saclay, 94800, Villejuif, France.
- Univ Paris-Sud, UMR-S 1193, Université Paris-Saclay, 94800, Villejuif, France.
- Centre Hépato-Biliaire, AP-HP Hôpital Paul-Brousse, 94800, Villejuif, France.
- DHU Hepatinov, 94800, Villejuif, France.
| | - Jean-Charles Duclos-Vallee
- Inserm, Unité 1193, Université Paris-Saclay, 94800, Villejuif, France.
- Univ Paris-Sud, UMR-S 1193, Université Paris-Saclay, 94800, Villejuif, France.
- Centre Hépato-Biliaire, AP-HP Hôpital Paul-Brousse, 94800, Villejuif, France.
- DHU Hepatinov, 94800, Villejuif, France.
| | - Eric Ballot
- Inserm, Unité 1193, Université Paris-Saclay, 94800, Villejuif, France.
- Univ Paris-Sud, UMR-S 1193, Université Paris-Saclay, 94800, Villejuif, France.
- Département d'Immunologie Biologique, Unité d'autoimmunité, AP-HP Hôpital Saint Antoine, 75012, Paris, France.
- DHU Hepatinov, 94800, Villejuif, France.
| |
Collapse
|
22
|
Sensitive array-based assay for determination of serological protein kinase A autoantibody levels based on its antigen protein activation. Clin Biochem 2016; 49:127-31. [DOI: 10.1016/j.clinbiochem.2015.08.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 08/21/2015] [Accepted: 08/25/2015] [Indexed: 11/18/2022]
|
23
|
Mir AR, Uddin M, Khan F, Alam K, Ali A. Dicarbonyl Induced Structural Perturbations Make Histone H1 Highly Immunogenic and Generate an Auto-Immune Response in Cancer. PLoS One 2015; 10:e0136197. [PMID: 26317779 PMCID: PMC4552624 DOI: 10.1371/journal.pone.0136197] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Accepted: 07/31/2015] [Indexed: 12/28/2022] Open
Abstract
Increased oxidative stress under hyperglycemic conditions, through the interaction of AGEs with RAGE receptors and via activation of interleukin mediated transcription signalling, has been reported in cancer. Proteins modifications are being explored for their roles in the development and progression of cancer and autoantibody response against them is gaining interest as a probe for early detection of the disease. This study has analysed the changes in histone H1 upon modification by methylglyoxal (MG) and its implications in auto-immunopathogenesis of cancer. Modified histone showed modifications in the aromatic residues, changed tyrosine microenvironment, intermolecular cross linking and generation of AGEs. It showed masking of hydrophobic patches and a hypsochromic shift in the in ANS specific fluorescence. MG aggressively oxidized histone H1 leading to the accumulation of reactive carbonyls. Far UV CD measurements showed di-carbonyl induced enhancement of the alpha structure and the induction of beta sheet conformation; and thermal denaturation (Tm) studies confirmed the thermal stability of the modified histone. FTIR analysis showed amide I band shift, generation of a carboxyethyl group and N-Cα vibrations in the modified histone. LCMS analysis confirmed the formation of Nε-(carboxyethyl)lysine and electron microscopic studies revealed the amorphous aggregate formation. The modified histone showed altered cooperative binding with DNA. Modified H1 induced high titre antibodies in rabbits and the IgG isolated form sera of rabbits immunized with modified H1 exhibited specific binding with its immunogen in Western Blot analysis. IgG isolated from the sera of patients with lung cancer, prostate cancer, breast cancer and cancer of head and neck region showed better recognition for neo-epitopes on the modified histone, reflecting the presence of circulating autoantibodies in cancer. Since reports suggest a link between AGE-RAGE axis and carcinogenesis, glycoxidation of histone H1 and its immunogenicity paves ways for understanding role of glycoxidatively damaged nuclear proteins in cancer.
Collapse
Affiliation(s)
- Abdul Rouf Mir
- Department of Biochemistry, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Moin Uddin
- Department of Biochemistry, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
- * E-mail:
| | - Farzana Khan
- Department of Biochemistry, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Khursheed Alam
- Department of Biochemistry, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Asif Ali
- Department of Biochemistry, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| |
Collapse
|
24
|
Increased sialylation of anti-Thomsen-Friedenreich antigen (CD176) antibodies in patients with gastric cancer: a diagnostic and prognostic potential. BIOMED RESEARCH INTERNATIONAL 2014; 2014:830847. [PMID: 25276822 PMCID: PMC4168149 DOI: 10.1155/2014/830847] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 08/18/2014] [Accepted: 08/18/2014] [Indexed: 11/17/2022]
Abstract
AIM To study whether alterations in the sialylation of antibodies (Ab) specific to the Thomsen-Friedenreich (TF) glycotope have a diagnostic and prognostic potential in gastric cancer. METHODS Serum samples were taken from patients with gastric carcinoma (n = 142) and controls (n = 61). The level of TF-specific antibodies and their sialylation was detected using ELISA with synthetic TF-polyacrylamide conjugate as antigen and sialic acid-specific Sambucus nigra agglutinin (SNA). RESULTS The level of TF-specific IgM was significantly decreased in cancer compared with controls (P ≤ 0.001). Cancer patients showed a higher level of SNA binding to anti-TF IgM and IgA (P ≤ 0.001) irrespective of disease stage, tumor morphology, and gender. Changes in the SNA/Ab index demonstrated moderate sensitivity (66-71%) and specificity (60-73%) for stomach cancer. The best diagnostic accuracy (100%) was achieved in 29% patients with high SNA binding and low anti-TF IgM level. This subset of patients demonstrated the poorest survival. CONCLUSION Our findings are the first evidence that the increased sialylation of TF-specific Abs combined with a low level of anti-TF IgM is strongly linked to gastric cancer and patients survival, which can be used as a novel biomarker for cancer detection and prognosis.
Collapse
|
25
|
Tumor protein D52 (TPD52) and cancer-oncogene understudy or understudied oncogene? Tumour Biol 2014; 35:7369-82. [PMID: 24798974 DOI: 10.1007/s13277-014-2006-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 04/22/2014] [Indexed: 12/16/2022] Open
Abstract
The Tumor protein D52 (TPD52) gene was identified nearly 20 years ago through its overexpression in human cancer, and a substantial body of data now strongly supports TPD52 representing a gene amplification target at chromosome 8q21.13. This review updates progress toward understanding the significance of TPD52 overexpression and targeting, both in tumors known to be characterized by TPD52 overexpression/amplification, and those where TPD52 overexpression/amplification has been recently or variably reported. We highlight recent findings supporting microRNA regulation of TPD52 expression in experimental systems and describe progress toward deciphering TPD52's cellular functions, particularly in cancer cells. Finally, we provide an overview of TPD52's potential as a cancer biomarker and immunotherapeutic target. These combined studies highlight the potential value of genes such as TPD52, which are overexpressed in many cancer types, but have been relatively understudied.
Collapse
|