1
|
Zhang P, Wu Z, Zhou T, Yang D, Mu Q, Zhang W, Yu L, Zhang S, Hu Y, Mu J, Jia W. Autoantibody repertoire profiling in tissue and blood identifies colorectal cancer-specific biomarkers. Cancer Sci 2024; 115:83-93. [PMID: 37985391 PMCID: PMC10823280 DOI: 10.1111/cas.16011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 10/19/2023] [Accepted: 10/25/2023] [Indexed: 11/22/2023] Open
Abstract
Autoantibodies (AAbs) in the blood of colorectal cancer (CRC) patients have been evaluated for tumor detection. However, it remains uncertain whether these AAbs are specific to tumor-associated antigens. In this study, we explored the IgG and IgM autoantibody repertoires in both the in situ tissue microenvironment and peripheral blood as potential tumor-specific biomarkers. We applied high-density protein arrays to profile AAbs in the tumor-infiltrating lymphocyte supernatants and corresponding serum from four patients with CRC, as well as in the serum of three noncancer controls. Our findings revealed that there were more reactive IgM AAbs than IgG in both the cell supernatant and corresponding serum, with a difference of approximately 3-5 times. Immunoglobulin G was predominant in the serum, while IgM was more abundant in the cell supernatant. We identified a range of AAbs present in both the supernatant and the corresponding serum, numbering between 432 and 780, with an average of 53.3% shared. Only 4.7% (n = 23) and 0.2% (n = 2) of reactive antigens for IgG and IgM AAbs, respectively, were specific to CRC. Ultimately, we compiled a list of 19 IgG AAb targets as potential tumor-specific AAb candidates. Autoantibodies against one of the top candidates, p15INK4b-related sequence/regulation of nuclear pre-mRNA domain-containing protein 1A (RPRD1A), were significantly elevated in 53 CRC patients compared to 119 controls (p < 0.0001). The project revealed that tissue-derived IgG AAbs, rather than IgM, are the primary source of tumor-specific AAbs in peripheral blood. It also identified potential tumor-specific AAbs that could be applied for noninvasive screening of CRC.
Collapse
Affiliation(s)
- Pei‐Fen Zhang
- Affiliated Tumor Hospital of Xinjiang Medical UniversityÜrümqiChina
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Ziyi Wu
- Department of Radiation OncologyFujian Medical University Cancer Hospital, Fujian Cancer HospitalFuzhouChina
| | - Ting Zhou
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Da‐Wei Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouChina
- School of Public HealthSun Yat‐sen UniversityGuangzhouChina
| | - Quan‐Kai Mu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Wen‐Bin Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Long Yu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Shao‐Dan Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Ye‐Zhu Hu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Jianbing Mu
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious DiseasesNational Institutes of HealthRockvilleMarylandUSA
| | - Wei‐Hua Jia
- Affiliated Tumor Hospital of Xinjiang Medical UniversityÜrümqiChina
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhouChina
| |
Collapse
|
2
|
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
|
3
|
Yu R, Yang S, Liu Y, Zhu Z. Identification and validation of serum autoantibodies in children with B-cell acute lymphoblastic leukemia by serological proteome analysis. Proteome Sci 2022; 20:3. [PMID: 35109855 PMCID: PMC8808998 DOI: 10.1186/s12953-021-00184-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 12/16/2021] [Indexed: 01/20/2023] Open
Abstract
Background B-cell acute lymphoblastic leukemia (B-ALL) is the most common malignancy of childhood. Even though significant progresses have been made in the treatment of B-ALL, some pediatric B-ALL have still poor prognosis. The identification of tumor autoantibodies may have utility in early cancer diagnosis and immunotherapy. In this study, we used serological proteome analysis (SERPA) to screen serum autoantibodies of pediatric B-ALL, aiming to contribute to the early detection of B-ALL in children. Methods The total proteins from three pooled B-ALL cell lines (NALM-6, REH and BALL-1 cells) were separated using two-dimensional gel electrophoresis (2-DE), which was followed by Western blot by mixed serum samples from children with B-ALL (n=20) or healthy controls (n=20). We analyzed the images of 2-D gel and Western blot by PDQuest software, and then identified the spots of immune responses in B-ALL samples compared with those in control samples. The proteins from spots were identified using mass spectrometry (MS). The autoantibodies against alpha-enolase (α-enolase) and voltage-dependent anion-selective channel protein 1 (VDAC1) were further validated in sera from another 30 children with B-ALL and 25 normal individuals by the use of enzyme-linked immunosorbent assay (ELISA). The protein expression levels of the candidate antigens α-enolase and VDAC1 in B-ALL were thoroughly studied by immunohistochemical analysis. Results Utilizing the SERPA approach, α-enolase and VDAC1 were identified as candidate autoantigens in children with B-ALL. The frequencies of autoantibodies against α-enolase and VDAC1 in children with B-ALL were 27% and 23% by using ELISA analysis, respectively, which were significantly higher than those in normal controls (4% and 0, p<0.05). Immunohistochemical analysis showed the expression of α-enolase and VDAC1 was positive in 95% and 85% of B-ALL patients, respectively, but negative expression levels were showed in the control group. Conclusions This study incidated that α-enolase and VDAC1 may be the autoantigens associated with B-ALL. Therefore, α-enolase and VDAC1 autoantibodies may be the potential serological markers for children with B-ALL. Supplementary Information The online version contains supplementary material available at 10.1186/s12953-021-00184-w.
Collapse
Affiliation(s)
- Runhong Yu
- Institute of Hematology, Henan Provincial People's Hospital, 7 Weiwu Road, Jinshui District, Zhengzhou, Henan, 450003, China.,Henan Key laboratory of Stem Cell Differentiation and Modification, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Shiwei Yang
- Institute of Hematology, Henan Provincial People's Hospital, 7 Weiwu Road, Jinshui District, Zhengzhou, Henan, 450003, China.,Henan Key laboratory of Stem Cell Differentiation and Modification, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Yufeng Liu
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe Road, Erqi District, Zhengzhou, Henan, 450052, China
| | - Zunmin Zhu
- Institute of Hematology, Henan Provincial People's Hospital, 7 Weiwu Road, Jinshui District, Zhengzhou, Henan, 450003, China. .,Henan Key laboratory of Stem Cell Differentiation and Modification, Henan Provincial People's Hospital, Zhengzhou, Henan, China. .,Department of Hematology, People's Hospital of Zhengzhou University, Henan, Zhengzhou, China.
| |
Collapse
|
4
|
Mummareddy S, Pradhan S, Narasimhan AK, Natarajan A. On Demand Biosensors for Early Diagnosis of Cancer and Immune Checkpoints Blockade Therapy Monitoring from Liquid Biopsy. BIOSENSORS 2021; 11:bios11120500. [PMID: 34940257 PMCID: PMC8699359 DOI: 10.3390/bios11120500] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/28/2021] [Accepted: 12/01/2021] [Indexed: 12/17/2022]
Abstract
Recently, considerable interest has emerged in the development of biosensors to detect biomarkers and immune checkpoints to identify and measure cancer through liquid biopsies. The detection of cancer biomarkers from a small volume of blood is relatively fast compared to the gold standard of tissue biopsies. Traditional immuno-histochemistry (IHC) requires tissue samples obtained using invasive procedures and specific expertise as well as sophisticated instruments. Furthermore, the turnaround for IHC assays is usually several days. To overcome these challenges, on-demand biosensor-based assays were developed to provide more immediate prognostic information for clinicians. Novel rapid, highly precise, and sensitive approaches have been under investigation using physical and biochemical methods to sense biomarkers. Additionally, interest in understanding immune checkpoints has facilitated the rapid detection of cancer prognosis from liquid biopsies. Typically, these devices combine various classes of detectors with digital outputs for the measurement of soluble cancer or immune checkpoint (IC) markers from liquid biopsy samples. These sensor devices have two key advantages: (a) a small volume of blood drawn from the patient is sufficient for analysis, and (b) it could aid physicians in quickly selecting and deciding the appropriate therapy regime for the patients (e.g., immune checkpoint blockade (ICB) therapy). In this review, we will provide updates on potential cancer markers, various biosensors in cancer diagnosis, and the corresponding limits of detection, while focusing on biosensor development for IC marker detection.
Collapse
Affiliation(s)
- Sai Mummareddy
- Department of Biology and Chemistry, Emory University, Atlanta, GA 30322, USA;
| | - Stuti Pradhan
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA 90095, USA;
| | - Ashwin Kumar Narasimhan
- Department of Biomedical Engineering, SRM Institute of Science and Technology, Chennai 603203, India;
| | - Arutselvan Natarajan
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, Stanford, CA 94305, USA
- Correspondence: ; Tel.: +1-650-736-9822
| |
Collapse
|
5
|
Kumar AR, Devan AR, Nair B, Vinod BS, Nath LR. Harnessing the immune system against cancer: current immunotherapy approaches and therapeutic targets. Mol Biol Rep 2021; 48:8075-8095. [PMID: 34671902 PMCID: PMC8605995 DOI: 10.1007/s11033-021-06752-9] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 09/15/2021] [Indexed: 02/08/2023]
Abstract
Cancer immunotherapy is a rapidly evolving concept that has been given the tag "fifth pillar" of cancer therapy while radiation therapy, chemotherapy, surgery and targeted therapy remain the other four pillars. This involves the stimulation of the immune system to control tumor growth and it specifically targets the neoplastic cells rather than the normal cells. Conventional chemotherapy has many limitations which include drug resistance, recurrence of cancer and severe adverse effects. Immunology has made major treatment breakthroughs for several cancers such as colorectal cancer, prostate cancer, breast cancer, lung cancer, liver cancer, kidney cancer, stomach cancer, acute lymphoblastic leukaemia etc. Currently, therapeutic strategies harnessing the immune system involve Checkpoint inhibitors, Chimeric antigen receptor T cells (CAR T cells), Monoclonal antibodies, Cancer vaccines, Cytokines, Radio-immunotherapy and Oncolytic virus therapy. The molecular characterization of several tumor antigens (TA) indicates that these TA can be utilized as promising candidates in cancer immunotherapy strategies. Here in this review, we highlight and summarize the different categories of emerging cancer immunotherapies along with the immunologically recognized tumor antigens involved in the tumor microenvironment.
Collapse
Affiliation(s)
- Ayana R Kumar
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala, 682041, India
| | - Aswathy R Devan
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala, 682041, India
| | - Bhagyalakshmi Nair
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala, 682041, India
| | - Balachandran S Vinod
- Department of Biochemistry, Sree Narayana College, Kollam, Kerala, 691001, India.
| | - Lekshmi R Nath
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala, 682041, India.
| |
Collapse
|
6
|
Abstract
Centromeric proteins are the foundation for assembling the kinetochore, a macromolecular complex that is essential for accurate chromosome segregation during mitosis. Anti-centromere antibodies (ACAs) are polyclonal autoantibodies targeting centromeric proteins (CENP-A, CENP-B, CENP-C), predominantly CENP-B, and are highly associated with rheumatologic disease (lcSSc/CREST syndrome). CENP-B autoantibodies have also been reported in cancer patients without symptoms of rheumatologic disease. The rise of oncoimmunotherapy stimulates inquiry into how and why anti-CENP-B autoantibodies are formed. In this review, we describe the clinical correlations between anti-CENP-B autoantibodies, rheumatologic disease, and cancer; the molecular features of CENP-B; possible explanations for autoantigenicity; and, finally, a possible mechanism for induction of autoantibody formation.
Collapse
|
7
|
Roney MSI, Lanagan C, Sheng YH, Lawler K, Schmidt C, Nguyen N, Begun J, Kijanka GS. IgM and IgA augmented autoantibody signatures improve early-stage detection of colorectal cancer prior to nodal and distant spread. Clin Transl Immunology 2021; 10:e1330. [PMID: 34603722 PMCID: PMC8473921 DOI: 10.1002/cti2.1330] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/30/2021] [Accepted: 08/02/2021] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVES Tumor-associated autoantibodies (AAbs) in individuals with cancer can precede clinical diagnosis by several months to years. The objective of this study was to determine whether the primary immune response in form of IgM and gut mucosa-associated IgA can aid IgG AAbs in the detection of early-stage colorectal cancer (CRC). METHODS We developed a novel protein array comprising 492 antigens seropositive in CRC. The array was used to profile IgG, IgM and IgA antibody signatures in 99 CRC patients and 99 sex- and age-matched non-cancer controls. A receiver operating curve (ROC), Kaplan-Meier survival analysis and univariate and multivariate Cox regression analyses were conducted. RESULTS We identified a panel of 16 multi-isotype AAbs with a cumulative sensitivity of 91% and specificity of 74% (AUC 0.90, 95% CI: 0.850-0.940) across all CRC stages. IgM and IgG isotypes were conversely associated with disease stage with IgM contributing significantly to improved stage I and II sensitivity of 96% at 78% specificity (AUC 0.928, 95% CI: 0.884-0.973). A single identified IgA AAb reached an overall sensitivity of 5% at 99% specificity (AUC 0.520, 95% CI: 0.440-0.601) balanced across all CRC stages. Kaplan-Meier analysis revealed that se33-1 (ZNF638) IgG AAbs were associated with reduced 5-year overall survival (log-rank test, P = 0.012), whereas cumulative IgM isotype signatures were associated with improved 5-year overall survival (log-rank test, P = 0.024). CONCLUSION IgM AAbs are associated with early-stage colorectal cancer. Combining IgG, IgM and IgA AAbs is a novel strategy to improve early diagnosis of cancers.
Collapse
Affiliation(s)
- Md Saiful Islam Roney
- Immune Profiling and Cancer GroupFaculty of MedicineMater Research Institute – The University of QueenslandTranslational Research InstituteWoolloongabbaQLDAustralia
| | - Catharine Lanagan
- Immune Profiling and Cancer GroupFaculty of MedicineMater Research Institute – The University of QueenslandTranslational Research InstituteWoolloongabbaQLDAustralia
| | - Yong Hua Sheng
- Inflammatory Bowel Diseases GroupFaculty of MedicineMater Research Institute – The University of QueenslandTranslational Research InstituteWoolloongabbaQLDAustralia
| | - Karen Lawler
- Pathology QueenslandQueensland HealthBrisbaneQLDAustralia
| | - Christopher Schmidt
- Immune Profiling and Cancer GroupFaculty of MedicineMater Research Institute – The University of QueenslandTranslational Research InstituteWoolloongabbaQLDAustralia
| | - Nam‐Trung Nguyen
- Queensland Micro‐ and Nanotechnology CentreGriffith UniversityBrisbaneQLDAustralia
| | - Jakob Begun
- Inflammatory Bowel Diseases GroupFaculty of MedicineMater Research Institute – The University of QueenslandTranslational Research InstituteWoolloongabbaQLDAustralia
- School of Clinical MedicineFaculty of MedicineThe University of QueenslandBrisbaneQLDAustralia
| | - Gregor Stefan Kijanka
- Immune Profiling and Cancer GroupFaculty of MedicineMater Research Institute – The University of QueenslandTranslational Research InstituteWoolloongabbaQLDAustralia
- Queensland Micro‐ and Nanotechnology CentreGriffith UniversityBrisbaneQLDAustralia
| |
Collapse
|
8
|
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
|
9
|
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: 7] [Impact Index Per Article: 1.8] [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
|
10
|
Cui C, Duan Y, Qiu C, Wang P, Sun G, Ye H, Dai L, Han Z, Song C, Wang K, Shi J, Zhang J. Identification of Novel Autoantibodies Based on the Human Proteomic Chips and Evaluation of Their Performance in the Detection of Gastric Cancer. Front Oncol 2021; 11:637871. [PMID: 33718231 PMCID: PMC7953047 DOI: 10.3389/fonc.2021.637871] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/27/2021] [Indexed: 12/24/2022] Open
Abstract
Autoantibodies against tumor-associated antigens (TAAbs) can be used as potential biomarkers in the detection of cancer. Our study aims to identify novel TAAbs for gastric cancer (GC) based on human proteomic chips and construct a diagnostic model to distinguish GC from healthy controls (HCs) based on serum TAAbs. The human proteomic chips were used to screen the candidate TAAbs. Enzyme-linked immunosorbent assay (ELISA) was used to verify and validate the titer of the candidate TAAbs in the verification cohort (80 GC cases and 80 HCs) and validation cohort (192 GC cases, 128 benign gastric disease cases, and 192 HCs), respectively. Then, the diagnostic model was established by Logistic regression analysis based on OD values of candidate autoantibodies with diagnostic value. Eleven candidate TAAbs were identified, including autoantibodies against INPP5A, F8, NRAS, MFGE8, PTP4A1, RRAS2, RGS4, RHOG, SRARP, RAC1, and TMEM243 by proteomic chips. The titer of autoantibodies against INPP5A, F8, NRAS, MFGE8, PTP4A1, and RRAS2 were significantly higher in GC cases while the titer of autoantibodies against RGS4, RHOG, SRARP, RAC1, and TMEM243 showed no difference in the verification group. Next, six potential TAAbs were validated in the validation cohort. The titer of autoantibodies against F8, NRAS, MFGE8, RRAS2, and PTP4A1 was significantly higher in GC cases. Finally, an optimal prediction model with four TAAbs (anti-NRAS, anti-MFGE8, anti-PTP4A1, and anti-RRAS2) showed an optimal diagnostic performance of GC with AUC of 0.87 in the training group and 0.83 in the testing group. The proteomic chip approach is a feasible method to identify TAAbs for the detection of cancer. Moreover, the panel consisting of anti-NRAS, anti-MFGE8, anti-PTP4A1, and anti-RRAS2 may be useful to distinguish GC cases from HCs.
Collapse
Affiliation(s)
- Chi Cui
- BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Epidemiology, Zhengzhou University, Zhengzhou, China
| | - Yaru Duan
- Henan Key Laboratory of Tumor Epidemiology, Zhengzhou University, Zhengzhou, China
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Cuipeng Qiu
- Henan Key Laboratory of Tumor Epidemiology, Zhengzhou University, Zhengzhou, China
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Peng Wang
- Henan Key Laboratory of Tumor Epidemiology, Zhengzhou University, Zhengzhou, China
- College of Public Health, Zhengzhou University, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
| | - Guiying Sun
- Henan Key Laboratory of Tumor Epidemiology, Zhengzhou University, Zhengzhou, China
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Hua Ye
- Henan Key Laboratory of Tumor Epidemiology, Zhengzhou University, Zhengzhou, China
- College of Public Health, Zhengzhou University, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
| | - Liping Dai
- BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Epidemiology, Zhengzhou University, Zhengzhou, China
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Zhuo Han
- Henan Key Laboratory of Tumor Epidemiology, Zhengzhou University, Zhengzhou, China
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Chunhua Song
- Henan Key Laboratory of Tumor Epidemiology, Zhengzhou University, Zhengzhou, China
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Kaijuan Wang
- Henan Key Laboratory of Tumor Epidemiology, Zhengzhou University, Zhengzhou, China
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Jianxiang Shi
- BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Epidemiology, Zhengzhou University, Zhengzhou, China
- College of Public Health, Zhengzhou University, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
| | - Jianying Zhang
- BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Tumor Epidemiology, Zhengzhou University, Zhengzhou, China
- College of Public Health, Zhengzhou University, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
11
|
Akhtar J, Priya R, Jain V, Sakhuja P, Agarwal AK, Goyal S, Polisetty RV, Sirdeshmukh R, Kar S, Gautam P. Immunoproteomics approach revealed elevated autoantibody levels against ANXA1 in early stage gallbladder carcinoma. BMC Cancer 2020; 20:1175. [PMID: 33261560 PMCID: PMC7709428 DOI: 10.1186/s12885-020-07676-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 11/22/2020] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Early diagnosis is important for the timely treatment of gallbladder carcinoma (GBC) patients and may lead to increased survival outcomes. Here, we have applied serological proteome analysis (SERPA), an immunoproteomics approach, for the detection of 'tumor-associated antigens (TAAs) that elicit humoral response' in early stage GBC patients. METHODS Total protein from pooled tumor tissue of GBC patients (n = 7) was resolved by two-dimensional gel electrophoresis (2-DE) followed by immunoblotting using pooled blood plasma from healthy volunteers (n = 11) or gallstone disease (GSD) cases (n = 11) or early stage GBC (Stage I and II) (n = 5) or GBC stage IIIA (n = 9). 2-D gel and immunoblot images were acquired and analyzed using PDQuest software to identify immunoreactive spots in GBC cases in comparison to controls. Proteins from immunoreactive spots were identified by liquid chromatography- tandem mass spectrometric analysis (LC-MS/MS). Autoantibody levels for two of the functionally relevant proteins were investigated in individual plasma samples (52 cases and 89 controls) by dot blot assay using recombinant proteins. RESULTS Image analysis using PDQuest software identified 25 protein spots with significantly high or specific immunoreactivity in GBC cases. Mass spectrometric analysis of 8 corresponding protein spots showing intense immunoreactivity (based on densitometric analysis) in early stage GBC or GBC stage IIIA cases led to the identification of 27 proteins. Some of the identified proteins include ANXA1, HSPD1, CA1, CA2, ALDOA and CTSD. Among the two proteins, namely ANXA1 and HSPD1 verified using a cohort of samples, significantly elevated autoantibody levels against ANXA1 were observed in early stage GBC cases in comparison to healthy volunteers or GSD cases (unpaired t-test, p < 0.05). Receiver operating characteristic (ROC) curve analysis for ANXA1 showed an Area under the Curve (AUC) of 0.69, with 41.7% sensitivity against a specificity of 89.9% for early stage GBC. IHC analysis for ANXA1 protein showed 'high' expression levels in 72% of GBC cases whereas all the controls showed 'low' expression levels. CONCLUSIONS The study suggests that the ANXA1 autoantibody levels against ANXA1 may be potentially employed for early stage detection of GBC patients. Other proteins could also be explored and verified in a large cohort of clinical samples.
Collapse
Affiliation(s)
- Javed Akhtar
- Laboratory of Molecular Oncology, ICMR- National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, 110029, India
- Jamia Hamdard- Institute of Molecular Medicine, Jamia Hamdard, New Delhi, 110062, India
| | - Ratna Priya
- Laboratory of Molecular Oncology, ICMR- National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, 110029, India
- Jamia Hamdard- Institute of Molecular Medicine, Jamia Hamdard, New Delhi, 110062, India
| | - Vaishali Jain
- Laboratory of Molecular Oncology, ICMR- National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, 110029, India
- Manipal Academy of Higher Education (MAHE) , 576104, Manipal, India
| | - Puja Sakhuja
- Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, 110002, India
| | - Anil Kumar Agarwal
- Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, 110002, India
| | - Surbhi Goyal
- Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, 110002, India
| | - Ravindra Varma Polisetty
- Department of Biochemistry, Sri Venkateswara College, University of Delhi, New Delhi, 110021, India
| | - Ravi Sirdeshmukh
- Manipal Academy of Higher Education (MAHE) , 576104, Manipal, India
- Institute of Bioinformatics, International Tech Park, Bangalore, 560066, India
| | - Sudeshna Kar
- Jamia Hamdard- Institute of Molecular Medicine, Jamia Hamdard, New Delhi, 110062, India
| | - Poonam Gautam
- Laboratory of Molecular Oncology, ICMR- National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, 110029, India.
| |
Collapse
|
12
|
Chang SC, Yuan SHC, Li CY, Chang HM, Wang HC, Pan YA, Hsueh PC, Wu CC, Yang Y, Liu HP. Significant association of serum autoantibodies to TYMS, HAPLN1 and IGFBP5 with early stage canine malignant mammary tumours. Vet Comp Oncol 2020; 19:172-182. [PMID: 33038064 DOI: 10.1111/vco.12657] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 10/04/2020] [Accepted: 10/05/2020] [Indexed: 01/14/2023]
Abstract
Canine mammary tumours (CMTs) are the most prevalent neoplasms in female dogs. Despite the high incidence of such tumours, a lack of easily accessible biomarkers still impedes early diagnosis of malignant CMTs. Herein we identify thymidylate synthetase (TYMS), hyaluronan and proteoglycan link protein 1 (HAPLN1) and insulin-like growth factor-binding protein 5 (IGFBP5) as CMT antigens eliciting corresponding autoantibodies in CMT cases. We establish enzyme-linked immunosorbent assays (ELISAs) to detect autoantibodies to TYMS (TYMS-AAb), HAPLN1 (HAPLN1-AAb) and IGFBP5 (IGFBP5-AAb) in sera from 81 dogs with malignant CMTs (41 in Stage I), 24 with benign CMTs and 35 healthy controls. Levels of all the three autoantibodies are elevated in the malignant group compared with the healthy or the benign group; notably, the elevated autoantibody levels significantly correlate with the stage-I CMTs. For discriminating malignant CMTs from healthy control, the area under curve (AUC) of TYMS-AAb is 0.694 with specificity of 82.9% and sensitivity of 50.6%. The AUC of utilising HAPLN1-AAb for distinguishing the stage-I CMTs from healthy controls is 0.711 with specificity of 77.1% and sensitivity of 58.5%. In differentiating malignant CMTs from the benign, the AUC of IGFBP5-AAb reaches 0.696 with specificity of 70.8% and sensitivity of 67.9%, and a combination of IGFBP5-AAb and TYMS-AAb increases the AUC to 0.72. Finally, the AUC of combined HAPLN1-AAb and IGFBP5-AAb in discriminating the stage-I CMTs from the benign achieves 0.731. Collectively, this study highlights a significant association of the three serum autoantibodies with early stage malignant CMTs.
Collapse
Affiliation(s)
- Shih-Chieh Chang
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Stephen Hsien-Chi Yuan
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Chia-Yin Li
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Huan-Ming Chang
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Heng-Cian Wang
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Yun-An Pan
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Pei-Chun Hsueh
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chih-Ching Wu
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Youngsen Yang
- Division of Hematology-Oncology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Hao-Ping Liu
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
13
|
Zhang X, Liu M, Zhang X, Wang Y, Dai L. Autoantibodies to tumor-associated antigens in lung cancer diagnosis. Adv Clin Chem 2020; 103:1-45. [PMID: 34229848 DOI: 10.1016/bs.acc.2020.08.005] [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: 12/24/2022]
Abstract
Lung cancer (LC) accounts for the majority of cancer-related deaths worldwide. Although screening the high-risk population by low-dose CT (LDCT) has reduced mortality, the cost and high false positivity rate has prevented its general diagnostic use. As such, better and more specific minimally invasive biomarkers are needed in general and for early LC detection, specifically. Autoantibodies produced by humoral immune response to tumor-associated antigens (TAA) are emerging as a promising noninvasive biomarker for LC. Given the low sensitivity of any one single autoantibody, a panel approach could provide a more robust and promising strategy to detect early stage LC. In this review, we summarize the background of TAA autoantibodies (TAAb) and the techniques currently used for identifying TAA, as well as recent findings of LC specific antigens and TAAb. This review provides guidance toward the development of accurate and reliable TAAb as immunodiagnostic biomarkers in the early detection of LC.
Collapse
Affiliation(s)
- Xiuzhi Zhang
- Department of Pathology, Henan Medical College, Zhengzhou, Henan, China
| | - Man Liu
- Henan Institute of Medical and Pharmaceutical Sciences in Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; School of Basic Medical Sciences & Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; Henan Key Laboratory of Tumor Epidemiology, Zhengzhou University, Zhengzhou, Henan, China
| | - Xue Zhang
- Henan Institute of Medical and Pharmaceutical Sciences in Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; School of Basic Medical Sciences & Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; Henan Key Laboratory of Tumor Epidemiology, Zhengzhou University, Zhengzhou, Henan, China
| | - Yulin Wang
- Henan Institute of Medical and Pharmaceutical Sciences in Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; School of Basic Medical Sciences & Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; Henan Key Laboratory of Tumor Epidemiology, Zhengzhou University, Zhengzhou, Henan, China
| | - Liping Dai
- Henan Institute of Medical and Pharmaceutical Sciences in Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; School of Basic Medical Sciences & Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; Henan Key Laboratory of Tumor Epidemiology, Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
14
|
Profiling of Naturally Occurring Antibodies to the Thomsen-Friedenreich Antigen in Health and Cancer: The Diversity and Clinical Potential. BIOMED RESEARCH INTERNATIONAL 2020; 2020:9747040. [PMID: 32280709 PMCID: PMC7128052 DOI: 10.1155/2020/9747040] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 03/17/2020] [Indexed: 02/07/2023]
Abstract
The Thomsen-Friedenreich (TF) antigen is expressed in a majority of human tumors due to aberrant glycosylation in cancer cells. There is strong evidence that humoral immune response to TF represents an effective mechanism for the elimination of cancer cells that express TF-positive glycoconjugates. The presence of naturally occurring antibodies to tumor-associated TF and cancer-specific changes in their levels, isotype distribution and interrelation, avidity, and glycosylation profile make these Abs a convenient and ubiquitous marker for cancer diagnostics and prognostics. In this review, we attempt to summarize the latest data on the potential of TF-specific Abs for cancer diagnostics and prognostics.
Collapse
|
15
|
Multiplexed Remote SPR Detection of Biological Interactions through Optical Fiber Bundles. SENSORS 2020; 20:s20020511. [PMID: 31963277 PMCID: PMC7014493 DOI: 10.3390/s20020511] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/08/2020] [Accepted: 01/14/2020] [Indexed: 02/04/2023]
Abstract
The development of sensitive methods for in situ detection of biomarkers is a real challenge to bring medical diagnosis a step forward. The proof-of-concept of a remote multiplexed biomolecular interaction detection through a plasmonic optical fiber bundle is demonstrated here. The strategy relies on a fiber optic biosensor designed from a 300 µm diameter bundle composed of 6000 individual optical fibers. When appropriately etched and metallized, each optical fiber exhibits specific plasmonic properties. The surface plasmon resonance phenomenon occurring at the surface of each fiber enables to measure biomolecular interactions, through the changes of the retro-reflected light intensity due to light/plasmon coupling variations. The functionalization of the microstructured bundle by multiple protein probes was performed using new polymeric 3D-printed microcantilevers. Such soft cantilevers allow for immobilizing the probes in micro spots, without damaging the optical microstructures nor the gold layer. We show here the potential of this device to perform the multiplexed detection of two different antibodies with limits of detection down to a few tenths of nanomoles per liter. This tool, adapted for multiparametric, real-time, and label free monitoring is minimally invasive and could then provide a useful platform for in vivo targeted molecular analysis.
Collapse
|
16
|
Xing M, Li P, Wang X, Li J, Shi J, Qin J, Zhang X, Ma Y, Francia G, Zhang JY. Overexpression of p62/IMP2 can Promote Cell Migration in Hepatocellular Carcinoma via Activation of the Wnt/β-Catenin Pathway. Cancers (Basel) 2019; 12:cancers12010007. [PMID: 31861402 PMCID: PMC7017416 DOI: 10.3390/cancers12010007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/10/2019] [Accepted: 12/11/2019] [Indexed: 12/24/2022] Open
Abstract
p62/IMP2 is an oncofetal protein that was first reported as a tumor-associated antigen in hepatocellular carcinoma (HCC). In our previous studies, we demonstrated a high frequency of p62/IMP2 autoantibodies appearing in various types of cancer. Therefore, we hypothesize that p62/IMP2 plays an important role in the progression of HCC, although the mechanism remains to be explored. In this study, we evaluated the expression of p62/IMP2 protein both in human tissues and liver cancer cell lines by immunohistochemistry and western blotting analysis and found that p62/IMP2 protein is overexpressed in human HCC tissue in comparison to normal human liver tissue. To explore the role that p62/IMP2 plays in HCC, p62/IMP2 was knocked out in two p62/IMP2-positive liver cancer cell lines (SNU449 and HepG2). Due to the low expression level of p62/IMP2 in SNU449, we overexpressed p62/IMP2 in this cell line. We subsequently demonstrated that high expression of p62/IMP2 in both cell lines can promote cell migration and invasion abilities in vitro by activating the Wnt/β-catenin pathway. We also used the Wnt/β-catenin pathway inhibitor, XAV 939, and a phosphoproteome assay to confirm our findings. Conclusion: Our results suggest that p62/IMP2 is an essential regulator of Wnt signaling pathways and plays an important role in HCC progression and metastasis.
Collapse
Affiliation(s)
- Mengtao Xing
- Department of Biological Sciences & NIH-Sponsored Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA; (M.X.); (P.L.); (X.W.); (J.L.); (J.S.); (J.Q.); (X.Z.); (Y.M.)
| | - Pei Li
- Department of Biological Sciences & NIH-Sponsored Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA; (M.X.); (P.L.); (X.W.); (J.L.); (J.S.); (J.Q.); (X.Z.); (Y.M.)
| | - Xiao Wang
- Department of Biological Sciences & NIH-Sponsored Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA; (M.X.); (P.L.); (X.W.); (J.L.); (J.S.); (J.Q.); (X.Z.); (Y.M.)
- Henan Medical and Pharmaceutical Institute, Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Jitian Li
- Department of Biological Sciences & NIH-Sponsored Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA; (M.X.); (P.L.); (X.W.); (J.L.); (J.S.); (J.Q.); (X.Z.); (Y.M.)
| | - Jianxiang Shi
- Department of Biological Sciences & NIH-Sponsored Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA; (M.X.); (P.L.); (X.W.); (J.L.); (J.S.); (J.Q.); (X.Z.); (Y.M.)
- Henan Medical and Pharmaceutical Institute, Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Jiejie Qin
- Department of Biological Sciences & NIH-Sponsored Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA; (M.X.); (P.L.); (X.W.); (J.L.); (J.S.); (J.Q.); (X.Z.); (Y.M.)
| | - Xiaojun Zhang
- Department of Biological Sciences & NIH-Sponsored Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA; (M.X.); (P.L.); (X.W.); (J.L.); (J.S.); (J.Q.); (X.Z.); (Y.M.)
| | - Yangcheng Ma
- Department of Biological Sciences & NIH-Sponsored Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA; (M.X.); (P.L.); (X.W.); (J.L.); (J.S.); (J.Q.); (X.Z.); (Y.M.)
| | - Giulio Francia
- Department of Biological Sciences & NIH-Sponsored Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA; (M.X.); (P.L.); (X.W.); (J.L.); (J.S.); (J.Q.); (X.Z.); (Y.M.)
- Correspondence: (G.F.); (J.-Y.Z.)
| | - Jian-Ying Zhang
- Department of Biological Sciences & NIH-Sponsored Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA; (M.X.); (P.L.); (X.W.); (J.L.); (J.S.); (J.Q.); (X.Z.); (Y.M.)
- Correspondence: (G.F.); (J.-Y.Z.)
| |
Collapse
|
17
|
Pan J, Zheng QZ, Li Y, Yu LL, Wu QW, Zheng JY, Pan XJ, Xie BS, Wu YA, Qian J, Zhu H, Huang Y. Discovery and Validation of a Serologic Autoantibody Panel for Early Diagnosis of Esophageal Squamous Cell Carcinoma. Cancer Epidemiol Biomarkers Prev 2019; 28:1454-1460. [PMID: 31239266 DOI: 10.1158/1055-9965.epi-18-1269] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 02/14/2019] [Accepted: 06/21/2019] [Indexed: 11/16/2022] Open
Affiliation(s)
- Jianbo Pan
- Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Qing-Zhu Zheng
- Provincial Clinical College, Fujian Medical University, Fuzhou, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, China
| | - Yadong Li
- Provincial Clinical College, Fujian Medical University, Fuzhou, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, China
| | - Li-Li Yu
- Provincial Clinical College, Fujian Medical University, Fuzhou, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, China
| | - Qing-Wei Wu
- Provincial Clinical College, Fujian Medical University, Fuzhou, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, China
| | - Jia-Ying Zheng
- Provincial Clinical College, Fujian Medical University, Fuzhou, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, China
| | - Xiao-Jie Pan
- Provincial Clinical College, Fujian Medical University, Fuzhou, China
- Department of Thoracic Surgery, Fujian Provincial Hospital, Fuzhou, China
| | - Bao-Song Xie
- Provincial Clinical College, Fujian Medical University, Fuzhou, China
- Department of Respiratory Medicine, Fujian Provincial Hospital, Fuzhou, China
| | - Yan-An Wu
- Provincial Clinical College, Fujian Medical University, Fuzhou, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, China
| | - Jiang Qian
- Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, Maryland.
| | - Heng Zhu
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland.
| | - Yi Huang
- Provincial Clinical College, Fujian Medical University, Fuzhou, China.
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, China
- Center for Experimental Research in Clinical Medicine, Fujian Provincial Hospital, Fuzhou, China
| |
Collapse
|
18
|
Hussain S, Saxena S, Shrivastava S, Arora R, Singh RJ, Jena SC, Kumar N, Sharma AK, Sahoo M, Tiwari AK, Mishra BP, Singh RK. Multiplexed Autoantibody Signature for Serological Detection of Canine Mammary Tumours. Sci Rep 2018; 8:15785. [PMID: 30361548 PMCID: PMC6202347 DOI: 10.1038/s41598-018-34097-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 10/09/2018] [Indexed: 01/19/2023] Open
Abstract
Spontaneously occurring canine mammary tumours (CMTs) are the most common neoplasms of female unspayed dogs and are of potential importance as models for human breast cancer as well. Mortality rates are thrice higher in dogs as compared to humans with breast cancer, which can partly be attributed to lack of diagnostic techniques for their early detection. Human breast cancer studies reveal role of autoantibodies in early cancer diagnosis and also the usefulness of autoantibody panels in increasing the sensitivity, as well as, specificity of diagnostic assays. Therefore, in this study, we took advantage of high-throughput Luminex technique for developing a multiplex assay to detect autoantibody signatures against 5 canine mammary tumour-associated autoantigens (TAAs). These TAAs were expressed separately as fusion proteins with halo tag at the N-terminus, which allows easy and specific covalent coupling with magnetic microspheres. The multiplex assay, comprising a panel of candidate autoantigens (TPI, PGAM1, MNSOD, CMYC & MUC1) was used for screening circulating autoantibodies in 125 dog sera samples, including 75 mammary tumour sera and 50 healthy dog sera. The area under curve (AUC) of the combined panel of biomarkers is 0.931 (p < 0.0001), which validates the discriminative potential of the panel in differentiating tumour patients from healthy controls. The assay could be conducted in 3hrs using only 1ul of serum sample and could detect clinical cases of canine mammary tumour with sensitivity and specificity of 78.6% and 90%, respectively. In this study, we report for the first time a multiplexed assay for detection of autoantibodies in canine tumours, utilizing luminex technology and halo-tag coupling strategy. Further to the best of our knowledge, autoantibodies to CMYC and MUC1 have been reported for the first time in canines in this study.
Collapse
Affiliation(s)
- Shahid Hussain
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute [Deemed University] Izatnagar, Bareilly, UP, India
| | - Sonal Saxena
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute [Deemed University] Izatnagar, Bareilly, UP, India.
| | - Sameer Shrivastava
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute [Deemed University] Izatnagar, Bareilly, UP, India.
| | - Richa Arora
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute [Deemed University] Izatnagar, Bareilly, UP, India
| | - Rajkumar James Singh
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute [Deemed University] Izatnagar, Bareilly, UP, India
| | - Subas Chandra Jena
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute [Deemed University] Izatnagar, Bareilly, UP, India
| | - Naveen Kumar
- Division of Veterinary Surgery, ICAR-Indian Veterinary Research Institute [Deemed University], Izatnagar, Bareilly, UP, India
| | - Anil Kumar Sharma
- Division of Veterinary Pathology, ICAR-Indian Veterinary Research Institute [Deemed University], Izatnagar, Bareilly, UP, India
| | - Monalisa Sahoo
- Division of Veterinary Pathology, ICAR-Indian Veterinary Research Institute [Deemed University], Izatnagar, Bareilly, UP, India
| | - Ashok Kumar Tiwari
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute [Deemed University] Izatnagar, Bareilly, UP, India
| | - Bishnu Prasad Mishra
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute [Deemed University] Izatnagar, Bareilly, UP, India
| | - Raj Kumar Singh
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute [Deemed University] Izatnagar, Bareilly, UP, India.
| |
Collapse
|
19
|
Li J, Dai L, Lei N, Xing M, Li P, Luo C, Casiano CA, Zhang JY. Evaluation and characterization of anti-RalA autoantibody as a potential serum biomarker in human prostate cancer. Oncotarget 2017; 7:43546-43556. [PMID: 27286458 PMCID: PMC5190043 DOI: 10.18632/oncotarget.9869] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 05/05/2016] [Indexed: 01/24/2023] Open
Abstract
Autoantibodies against intracellular tumor-associated antigens (TAAs) are commonly found in human cancers. In this study, we characterized the serum autoantibody response to the RalA, Ras-like GTPase, in patients with prostate cancer (PCa). The autoantibodies were detected by immunofluorescence assay in PCa cell lines, ELISA, and immunoblotting in 339 serum samples from patients with PCa and benign prostatic hyperplasia (BPH), and in normal human sera (NHS). The expression of RalA in prostate tumor tissues was evaluated by immunohistochemistry (IHC) in tumor microarrays. The autoantibody level to RalA (median) in NHS was significantly lower than in PCa (0.053 vs 0.138; P < 0.001) and BPH (0.053 vs 0.132; P < 0.005) groups. The circulating anti-RalA autoantibody could distinguish PCa patients from normal individuals with the area under the receiver operating characteristic (ROC) curve (AUC) performing at 0.861, with sensitivity of 52.9% and specificity of 91.0%. Elevation in serum immunoreactivity was observed in PCa patients after radical prostatectomy. The combined use of both anti-RalA autoantibody and PSA showed a significantly higher discriminatory ability compared with either of those markers alone. RalA protein expression was detected by IHC in 85.3% of tumor tissues from PCa patients, but without significant difference compared to BPH or normal control tissues. Together, our study shows the additional benefits of anti-RalA autoantibody as a potential serological biomarker for PCa, particularly in patients with normal PSA, and further demonstrate the utility of biomarker combinations in the immunodiagnosis of PCa.
Collapse
Affiliation(s)
- Jitian Li
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX 79968, USA
| | - Liping Dai
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX 79968, USA.,Henan Key Laboratory of Tumor Epidemiology and Henan Academy of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Ningjing Lei
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX 79968, USA
| | - Mengtao Xing
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX 79968, USA
| | - Pei Li
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX 79968, USA
| | - Chenglin Luo
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX 79968, USA
| | - Carlos A Casiano
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| | - Jian-Ying Zhang
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX 79968, USA.,Henan Key Laboratory of Tumor Epidemiology and Henan Academy of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, China
| |
Collapse
|
20
|
Jin Y, Kim SC, Kim HJ, Ju W, Kim YH, Kim HJ. Use of autoantibodies against tumor-associated antigens as serum biomarkers for primary screening of cervical cancer. Oncotarget 2017; 8:105425-105439. [PMID: 29285261 PMCID: PMC5739648 DOI: 10.18632/oncotarget.22231] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 10/02/2017] [Indexed: 12/20/2022] Open
Abstract
Serum autoantibodies against tumor-associated antigens (TAAs) have received much attention as potential biomarkers for early detection of cancers, since they can be detected in the early stages of cancers. Autoantibodies against Cancer Antigen 15-3 (CA15-3), carcinoembryonic antigen (CEA), Cancer Antigen 19-9 (CA19-9), c-Myc, p53, heat shock protein (Hsp)27 and Hsp70 have been suggested as potential markers for detecting several types of cancer. In the present study, the seven types of antibody listed above were evaluated for detecting cervical lesions. Enzyme-linked immunosorbent assays (ELISAs) were used to measure IgG levels of the autoantibodies in women with normal cytology, cervical intraepithelial neoplasia (CIN) I, CIN II, CIN III and cervical cancer. The increases of anti-CA15-3 and anti-CEA IgG in cervical cancer were more pronounced than the increases of the other markers, and the level of anti-CA19-9 IgG in CIN III stage was higher than in normal CIN I, CIN II or cervical cancer. A combination of ELISAs detecting anti-CA15-3, anti-CEA and anti-CA19-9 IgGs was found to reliably discriminate CINs from normal and to strongly differentiate cancer from normal (90.3% of sensitivity and 82.1% of specificity). We suggest that the combination of three ELISA may be useful for detecting cervical lesions.
Collapse
Affiliation(s)
- Yingji Jin
- Laboratory of Virology, College of Pharmacy, Chung-Ang University, Dongjak-Gu, Seoul 06974, South Korea
| | - Seung Cheol Kim
- Department of Obstetrics and Gynecology, Ewha Womans University College of Medicine, Yangcheon-Gu, Seoul 03760, South Korea
| | - Hyoung Jin Kim
- Laboratory of Virology, College of Pharmacy, Chung-Ang University, Dongjak-Gu, Seoul 06974, South Korea
| | - Woong Ju
- Department of Obstetrics and Gynecology, Ewha Womans University College of Medicine, Yangcheon-Gu, Seoul 03760, South Korea
| | - Yun Hwan Kim
- Department of Obstetrics and Gynecology, Ewha Womans University College of Medicine, Yangcheon-Gu, Seoul 03760, South Korea
| | - Hong-Jin Kim
- Laboratory of Virology, College of Pharmacy, Chung-Ang University, Dongjak-Gu, Seoul 06974, South Korea
| |
Collapse
|
21
|
Liu XX, Ye H, Wang P, Zhang Y, Zhang JY. Identification of 14‑3‑3ζ as a potential biomarker in gastric cancer by proteomics‑based analysis. Mol Med Rep 2017; 16:7759-7765. [PMID: 28944820 DOI: 10.3892/mmr.2017.7496] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 07/20/2017] [Indexed: 11/06/2022] Open
Abstract
The identification of tumor biomarkers to support early diagnosis and tumor progression monitoring may potentially reduce the mortality of gastric cancer (GC). The present study aimed to detect novel tumor‑associated antigens from the AGS GC cell line, and to identify their associated autoantibodies in sera from patients with GC by proteomics‑based approaches. Proteins from AGS cell lysates were isolated using two‑dimensional polyacrylamide gel electrophoresis, and western blotting was subsequently performed, to determine autoantibody responses in sera derived from patients with GC and healthy individuals. Positive protein spots were removed from gels stained with Coomassie blue, and were then evaluated by liquid chromatography‑tandem mass spectrometry. Sera from patients with GC produced numerous spots, one of which was identified as 14‑3‑3ζ. Autoantibody frequency to 14‑3‑3ζ was 17.6% (15/85) in patients with GC, which was significantly higher than that in healthy control individuals (2.4%; 2/85; P<0.01). These results suggested that the autoantibody against 14‑3‑3ζ may be a potential serological biomarker for the detection and diagnosis of GC.
Collapse
Affiliation(s)
- Xin-Xin Liu
- Center for Tumor Biotherapy, The First Affiliated Hospital of Zhengzhou University and College of Public Health, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Hua Ye
- Center for Tumor Biotherapy, The First Affiliated Hospital of Zhengzhou University and College of Public Health, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Peng Wang
- Center for Tumor Biotherapy, The First Affiliated Hospital of Zhengzhou University and College of Public Health, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yi Zhang
- Center for Tumor Biotherapy, The First Affiliated Hospital of Zhengzhou University and College of Public Health, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Jian-Ying Zhang
- Center for Tumor Biotherapy, The First Affiliated Hospital of Zhengzhou University and College of Public Health, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
22
|
Hori SS, Lutz AM, Paulmurugan R, Gambhir SS. A Model-Based Personalized Cancer Screening Strategy for Detecting Early-Stage Tumors Using Blood-Borne Biomarkers. Cancer Res 2017; 77:2570-2584. [PMID: 28283654 DOI: 10.1158/0008-5472.can-16-2904] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 12/07/2016] [Accepted: 03/02/2017] [Indexed: 12/18/2022]
Abstract
An effective cancer blood biomarker screening strategy must distinguish aggressive from nonaggressive tumors at an early, intervenable time. However, for blood-based strategies to be useful, the quantity of biomarker shed into the blood and its relationship to tumor growth or progression must be validated. To study how blood biomarker levels correlate with early-stage viable tumor growth in a mouse model of human cancer, we monitored early tumor growth of engineered human ovarian cancer cells (A2780) implanted orthotopically into nude mice. Biomarker shedding was monitored by serial blood sampling, whereas tumor viability and volume were monitored by bioluminescence imaging and ultrasound imaging. From these metrics, we developed a mathematical model of cancer biomarker kinetics that accounts for biomarker shedding from tumor and healthy cells, biomarker entry into vasculature, biomarker elimination from plasma, and subject-specific tumor growth. We validated the model in a separate set of mice in which subject-specific tumor growth rates were accurately predicted. To illustrate clinical translation of this strategy, we allometrically scaled model parameters from mouse to human and used parameters for PSA shedding and prostate cancer. In this manner, we found that blood biomarker sampling data alone were capable of enabling the detection and discrimination of simulated aggressive (2-month tumor doubling time) and nonaggressive (18-month tumor doubling time) tumors as early as 7.2 months and 8.9 years before clinical imaging, respectively. Our model and screening strategy offers broad impact in their applicability to any solid cancer and associated biomarkers shed, thereby allowing a distinction between aggressive and nonaggressive tumors using blood biomarker sampling data alone. Cancer Res; 77(10); 2570-84. ©2017 AACR.
Collapse
Affiliation(s)
- Sharon Seiko Hori
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, California
| | - Amelie M Lutz
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, California
| | - Ramasamy Paulmurugan
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, California
| | - Sanjiv Sam Gambhir
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, California. .,Departments of Bioengineering and Materials Science & Engineering, Stanford University, Stanford, California
| |
Collapse
|
23
|
Shi L, Chevolot Y, Souteyrand E, Laurenceau E. Autoantibodies against heat shock proteins as biomarkers for the diagnosis and prognosis of cancer. Cancer Biomark 2017; 18:105-116. [DOI: 10.3233/cbm-160117] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
24
|
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
|
25
|
A panel of autoantibodies as potential early diagnostic serum biomarkers in patients with breast cancer. Int J Clin Oncol 2016; 22:291-296. [DOI: 10.1007/s10147-016-1047-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 09/27/2016] [Indexed: 12/16/2022]
|
26
|
Sanchez TW, Zhang G, Li J, Dai L, Mirshahidi S, Wall NR, Yates C, Wilson C, Montgomery S, Zhang JY, Casiano CA. Immunoseroproteomic Profiling in African American Men with Prostate Cancer: Evidence for an Autoantibody Response to Glycolysis and Plasminogen-Associated Proteins. Mol Cell Proteomics 2016; 15:3564-3580. [PMID: 27742740 DOI: 10.1074/mcp.m116.060244] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 10/12/2016] [Indexed: 01/21/2023] Open
Abstract
African American (AA) men suffer from a disproportionately high incidence and mortality of prostate cancer (PCa) compared with other racial/ethnic groups. Despite these disparities, African American men are underrepresented in clinical trials and in studies on PCa biology and biomarker discovery. We used immunoseroproteomics to profile antitumor autoantibody responses in AA and European American (EA) men with PCa, and explored differences in these responses. This minimally invasive approach detects autoantibodies to tumor-associated antigens that could serve as clinical biomarkers and immunotherapeutic agents. Sera from AA and EA men with PCa were probed by immunoblotting against PC3 cell proteins, with AA sera showing stronger immunoreactivity. Mass spectrometry analysis of immunoreactive protein spots revealed that several AA sera contained autoantibodies to a number of proteins associated with both the glycolysis and plasminogen pathways, particularly to alpha-enolase (ENO1). The proteomic data is deposited in ProteomeXchange with identifier PXD003968. Analysis of sera from 340 racially diverse men by enzyme-linked immunosorbent assays (ELISA) showed higher frequency of anti-ENO1 autoantibodies in PCa sera compared with control sera. We observed differences between AA-PCa and EA-PCa patients in their immunoreactivity against ENO1. Although EA-PCa sera reacted with higher frequency against purified ENO1 in ELISA and recognized by immunoblotting the endogenous cellular ENO1 across a panel of prostate cell lines, AA-PCa sera reacted weakly against this protein by ELISA but recognized it by immunoblotting preferentially in metastatic cell lines. These race-related differences in immunoreactivity to ENO1 could not be accounted by differential autoantibody recognition of phosphoepitopes within this antigen. Proteomic analysis revealed differences in the posttranslational modification profiles of ENO1 variants differentially recognized by AA-PCa and EA-PCa sera. These intriguing results suggest the possibility of race-related differences in the antitumor autoantibody response in PCa, and have implications for defining novel biological determinants of PCa health disparities.
Collapse
Affiliation(s)
- Tino W Sanchez
- From the ‡Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California 92350;
| | - Guangyu Zhang
- ¶Mass Spectrometry Core Facility, Division of Biochemistry, LLU School of Medicine, Loma Linda, California 92350
| | - Jitian Li
- §Department of Biological Sciences, University of Texas, El Paso, Texas 79968
| | - Liping Dai
- §Department of Biological Sciences, University of Texas, El Paso, Texas 79968
| | - Saied Mirshahidi
- From the ‡Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California 92350.,‖LLU Cancer Center Biospecimen Laboratory, Loma Linda, California 92350
| | - Nathan R Wall
- From the ‡Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California 92350
| | - Clayton Yates
- ‡‡Department of Biology and Center for Cancer Research, Tuskegee University, Tuskegee Alabama 36088
| | - Colwick Wilson
- From the ‡Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California 92350.,**LLU School of Behavioral Health, Loma Linda, California 92350
| | - Susanne Montgomery
- From the ‡Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California 92350.,**LLU School of Behavioral Health, Loma Linda, California 92350
| | - Jian-Ying Zhang
- §Department of Biological Sciences, University of Texas, El Paso, Texas 79968
| | - Carlos A Casiano
- From the ‡Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California 92350.,§§Department of Medicine, Division of Rheumatology, LLU School of Medicine, Loma Linda, California 92350
| |
Collapse
|
27
|
He X, Hong Y, Wang X, Zhang X, Long J, Li H, Zhang B, Chen S, Liu Q, Li H, Wang X, Ou X, Huang J. Identification and clinical significance of an elevated level of serum aminoacylase-1 autoantibody in patients with hepatitis B virus-related liver cirrhosis. Mol Med Rep 2016; 14:4255-4262. [PMID: 27633755 DOI: 10.3892/mmr.2016.5740] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 08/10/2016] [Indexed: 11/06/2022] Open
Abstract
Chronic hepatitis B (CHB) is prevalent worldwide and can develop into liver cirrhosis and liver carcinoma. Early discrimination of liver cirrhosis from chronic hepatitis is critical for effective treatment and optimal prognosis. The aim of the present study was to assess the diagnostic value of a panel of cellular proteins that can be recognized by autoantibodies in patient serum for hepatitis B virus (HBV)‑related liver cirrhosis. Twenty‑two candidate autoantigens screened using a serum proteomics assay in our previous study were assessed retrospectively in 443 participants, comprising 89 patients with HBV‑related liver cirrhosis, 89 patients with CHB, and 265 healthy controls. The levels of autoantibodies against the candidate autoantigens were measured by protein microarrays containing the candidate antigen proteins. Receiver operating characteristic (ROC) curves were used to calculate the diagnostic accuracy. The present study determined that seven of the 22 candidate autoantibodies differed significantly in serum level between HBV‑related liver cirrhosis and CHB (P<0.0001), with area under curve (AUC) values >0.7. The seven autoantibodies recognized aminoacylase‑1 (ACY1), histidine triad nucleotide‑binding protein 1, insulin‑like growth factor 2 mRNA‑binding protein 2, heat shock 70 kDa protein 6, peroxiredoxin 3, apoptosis‑inducing factor and regucalcin. Among these, the ACY1 autoantibody had the highest value for discriminating HBV‑related liver cirrhosis from CHB, with an AUC value of 0.872 (95% confidence interval: 0.810‑0.934, P<0.0001), sensitivity of 77.3% and specificity of 85.0%. In conclusion, with the elevated level in the disease progression of CHB, ACY1 autoantibody may be a valuable serum biomarker for discriminating HBV‑related liver cirrhosis from CHB.
Collapse
Affiliation(s)
- Xiaomin He
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Yu Hong
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Xiaomei Wang
- Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100026, P.R. China
| | - Xiaohong Zhang
- Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100026, P.R. China
| | - Jiang Long
- Department of Oncology Minimally Invasive Interventional Radiology, Beijing You‑an Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Hai Li
- Department of Hepatopancreatobiliary and Splenic Medicine, Affiliated Hospital of Medical College of Chinese People's Armed Police Force, Tianjin 300192, P.R. China
| | - Bei Zhang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Suhong Chen
- Biotechnology Center, Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
| | - Qiqi Liu
- Biotechnology Center, Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
| | - Hongyi Li
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Xiaoming Wang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Xiaojuan Ou
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Jian Huang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| |
Collapse
|
28
|
p62/IMP2 stimulates cell migration and reduces cell adhesion in breast cancer. Oncotarget 2016; 6:32656-68. [PMID: 26416451 PMCID: PMC4741720 DOI: 10.18632/oncotarget.5328] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 09/11/2015] [Indexed: 12/23/2022] Open
Abstract
p62/IMP2 is an oncofetal protein that is overexpressed in several types of cancer, and is a member of the family of insulin-like growth factor 2 mRNA binding proteins. We previously reported that high levels of p62/IMP2 autoantibody are present in sera from cancer patients, compared to healthy individuals. Here, we report the overexpression of p62/IMP2 in tumor tissues of 72 out of 104 cases of human breast cancer, and high levels of p62/IMP2 autoantibody in patients’ sera (in 63 out of 216 cases). To explore the role of p62/IMP2 in breast cancer progression, we generated p62/IMP2 transfected variants of two human breast cancer cell lines: MDA-MB-231 and LM2-4. Using in vitro assays we found that overexpression of p62/IMP2 can increase cell migration, and reduce cell adhesion to extracellular matrix (ECM) proteins. A Human Extracellular Matrix and Adhesion Molecules qPCR array was performed with our generated variants, and it identified a group of mRNAs whose expression was altered with p62/IMP2 overexpression, including connective tissue growth factor (CTGF) mRNA – which we show to be a p62/IMP2 binding partner. Overall, our results provide new insights into the molecular mechanism by which p62/IMP2 can contribute to breast cancer progression.
Collapse
|
29
|
Soler M, Estevez MC, Villar-Vazquez R, Casal JI, Lechuga LM. Label-free nanoplasmonic sensing of tumor-associate autoantibodies for early diagnosis of colorectal cancer. Anal Chim Acta 2016; 930:31-8. [DOI: 10.1016/j.aca.2016.04.059] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 04/25/2016] [Accepted: 04/30/2016] [Indexed: 12/15/2022]
|
30
|
Li Y, Nakka M, Kelly AJ, Lau CC, Krailo M, Barkauskas DA, Hicks JM, Man TK. p27 Is a Candidate Prognostic Biomarker and Metastatic Promoter in Osteosarcoma. Cancer Res 2016; 76:4002-11. [PMID: 27197201 DOI: 10.1158/0008-5472.can-15-3189] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 03/31/2016] [Indexed: 11/16/2022]
Abstract
Metastatic progression is the major cause of death in osteosarcoma, the most common bone malignancy in children and young adults. However, prognostic biomarkers and efficacious targeted treatments for metastatic disease remain lacking. Using an immunoproteomic approach, we discovered that autoantibodies against the cell-cycle kinase inhibitor p27 (KIP1, CDKN1B) were elevated in plasma of high-risk osteosarcoma patients. Using a large cohort of serum samples from osteosarcoma patients (n = 233), we validated that a higher level of the p27 autoantibody significantly correlated with poor overall and event-free survival (P < 0.05). Immunohistochemical analysis also showed that p27 was mislocalized to the cytoplasm in the majority of osteosarcoma cases and in highly metastatic osteosarcoma cell lines. We demonstrated that ectopic expression of cytoplasmic p27 promoted migration and invasion of osteosarcoma cells, whereas shRNA-mediated gene silencing suppressed these effects. In addition, mutations at the p27 phosphorylation sites S10 or T198, but not T157, abolished the migratory and invasive phenotypes. Furthermore, the development of pulmonary metastases increased in mice injected with cells expressing cytoplasmic p27 compared with an empty vector control. Collectively, our findings support further investigation of p27 as a potential prognostic biomarker and therapeutic target in osteosarcoma cases exhibiting aberrant p27 subcellular localization. Cancer Res; 76(13); 4002-11. ©2016 AACR.
Collapse
Affiliation(s)
- Yiting Li
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas. Dan L. Duncan Cancer Center Baylor College of Medicine, Houston, Texas. Texas Children's Hematology and Oncology Centers, Texas Children's Hospital, Houston, Texas
| | - Manjula Nakka
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas. Texas Children's Hematology and Oncology Centers, Texas Children's Hospital, Houston, Texas
| | - Aaron J Kelly
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas. Program of Structural and Computational Biology and Molecular Biophysics, Baylor College of Medicine, Houston, Texas
| | - Ching C Lau
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas. Dan L. Duncan Cancer Center Baylor College of Medicine, Houston, Texas. Texas Children's Hematology and Oncology Centers, Texas Children's Hospital, Houston, Texas. Program of Structural and Computational Biology and Molecular Biophysics, Baylor College of Medicine, Houston, Texas
| | - Mark Krailo
- Children's Oncology Group, Monrovia, California. Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Donald A Barkauskas
- Children's Oncology Group, Monrovia, California. Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - John M Hicks
- Department of Pathology, Baylor College of Medicine, Houston, Texas
| | - Tsz-Kwong Man
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas. Dan L. Duncan Cancer Center Baylor College of Medicine, Houston, Texas. Texas Children's Hematology and Oncology Centers, Texas Children's Hospital, Houston, Texas. Program of Structural and Computational Biology and Molecular Biophysics, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
31
|
Schlick B, Massoner P, Lueking A, Charoentong P, Blattner M, Schaefer G, Marquart K, Theek C, Amersdorfer P, Zielinski D, Kirchner M, Trajanoski Z, Rubin MA, Müllner S, Schulz-Knappe P, Klocker H. Serum Autoantibodies in Chronic Prostate Inflammation in Prostate Cancer Patients. PLoS One 2016; 11:e0147739. [PMID: 26863016 PMCID: PMC4749310 DOI: 10.1371/journal.pone.0147739] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 01/07/2016] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Chronic inflammation is frequently observed on histological analysis of malignant and non-malignant prostate specimens. It is a suspected supporting factor for prostate diseases and their progression and a main cause of false positive PSA tests in cancer screening. We hypothesized that inflammation induces autoantibodies, which may be useful biomarkers. We aimed to identify and validate prostate inflammation associated serum autoantibodies in prostate cancer patients and evaluate the expression of corresponding autoantigens. METHODS Radical prostatectomy specimens of prostate cancer patients (N = 70) were classified into high and low inflammation groups according to the amount of tissue infiltrating lymphocytes. The corresponding pre-surgery blood serum samples were scrutinized for autoantibodies using a low-density protein array. Selected autoantigens were identified in prostate tissue and their expression pattern analyzed by immunohistochemistry and qPCR. The identified autoantibody profile was cross-checked in an independent sample set (N = 63) using the Luminex-bead protein array technology. RESULTS Protein array screening identified 165 autoantibodies differentially abundant in the serum of high compared to low inflammation patients. The expression pattern of three corresponding antigens were established in benign and cancer tissue by immunohistochemistry and qPCR: SPAST (Spastin), STX18 (Syntaxin 18) and SPOP (speckle-type POZ protein). Of these, SPAST was significantly increased in prostate tissue with high inflammation. All three autoantigens were differentially expressed in primary and/or castration resistant prostate tumors when analyzed in an inflammation-independent tissue microarray. Cross-validation of the inflammation autoantibody profile on an independent sample set using a Luminex-bead protein array, retrieved 51 of the significantly discriminating autoantibodies. Three autoantibodies were significantly upregulated in both screens, MUT, RAB11B and CSRP2 (p>0.05), two, SPOP and ZNF671, close to statistical significance (p = 0.051 and 0.076). CONCLUSIONS We provide evidence of an inflammation-specific autoantibody profile and confirm the expression of corresponding autoantigens in prostate tissue. This supports evaluation of autoantibodies as non-invasive markers for prostate inflammation.
Collapse
Affiliation(s)
- Bettina Schlick
- Division of Experimental Urology, Dept. of Urology, Medical University of Innsbruck, Innsbruck, Austria
- ONCOTYROL, Center for Personalized Cancer Medicine, Innsbruck, Austria
| | - Petra Massoner
- Division of Experimental Urology, Dept. of Urology, Medical University of Innsbruck, Innsbruck, Austria
- ONCOTYROL, Center for Personalized Cancer Medicine, Innsbruck, Austria
| | | | | | - Mirjam Blattner
- Department of Pathology and Laboratory Medicine, Institute of Precision Medicine, Weill Medical College of Cornell University, New York, NY, United States of America
| | - Georg Schaefer
- ONCOTYROL, Center for Personalized Cancer Medicine, Innsbruck, Austria
- Department of Pathology, Medical University of Innsbruck, Innsbruck, Austria
| | | | | | | | | | | | - Zlatko Trajanoski
- Division of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Mark A. Rubin
- Department of Pathology and Laboratory Medicine, Institute of Precision Medicine, Weill Medical College of Cornell University, New York, NY, United States of America
| | | | | | - Helmut Klocker
- Division of Experimental Urology, Dept. of Urology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
32
|
Schummer M, Thorpe J, Giraldez M, Bergan L, Tewari M, Urban N. Evaluating Serum Markers for Hormone Receptor-Negative Breast Cancer. PLoS One 2015; 10:e0142911. [PMID: 26565788 PMCID: PMC4643893 DOI: 10.1371/journal.pone.0142911] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 10/27/2015] [Indexed: 12/27/2022] Open
Abstract
INTRODUCTION Breast cancer is the most frequently diagnosed cancer and the leading cause of cancer death in females worldwide. Death rates have been declining, largely as a result of early detection through mammography and improved treatment, but mammographic screening is controversial because of over-diagnosis of breast disease that might not require treatment, and under-diagnosis of cancer in women with dense breasts. Breast cancer screening could be improved by pairing mammography with a tumor circulating marker, of which there are currently none. Given genomic similarities between the basal breast cancer subtype and serous ovarian cancer, and given our success in identifying circulating markers for ovarian cancer, we investigated the performance in hormone receptor-negative breast cancer detection of both previously identified ovarian serum markers and circulating markers associated with transcripts that were differentially expressed in breast cancer tissue compared to healthy breast tissue from reduction mammaplasties. METHODS We evaluated a total of 15 analytes (13 proteins, 1 miRNA, 1 autoantibody) in sera drawn at or before breast cancer surgery from 43 breast cancer cases (28 triple-negative-TN-and 15 hormone receptor-negative-HRN-/ HER2-positive) and 87 matched controls. RESULTS In the analysis of our whole cohort of breast cancer cases, autoantibodies to TP53 performed significantly better than the other selected 14 analytes showing 25.6% and 34.9% sensitivity at 95% and 90% specificity respectively with AUC: 0.7 (p<0.001). The subset of 28 TN cancers showed very similar results. We observed no correlation between anti-TP53 and the 14 other markers; however, anti-TP53 expression correlated with Body-Mass-Index. It did not correlate with tumor size, positive lymph nodes, tumor stage, the presence of metastases or recurrence. CONCLUSION None of the 13 serum proteins nor miRNA 135b identified women with HRN or TN breast cancer. TP53 autoantibodies identified women with HRN breast cancer and may have potential for early detection, confirming earlier reports. TP53 autoantibodies are long lasting in serum but may be affected by storage duration. Autoantibodies to TP53 might correlate with Body-Mass-Index.
Collapse
Affiliation(s)
- Michèl Schummer
- Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, Washington, United States of America
| | - Jason Thorpe
- Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, Washington, United States of America
| | - Maria Giraldez
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Lindsay Bergan
- Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, Washington, United States of America
| | - Muneesh Tewari
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, Michigan, United States of America
- Divisions of Hematology/Oncology and Molecular Medicine and Genetics, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, United States of America
- Center for Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Nicole Urban
- Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, Washington, United States of America
| |
Collapse
|
33
|
Syed P, Gupta S, Choudhary S, Pandala NG, Atak A, Richharia A, K P M, Zhu H, Epari S, Noronha SB, Moiyadi A, Srivastava S. Autoantibody Profiling of Glioma Serum Samples to Identify Biomarkers Using Human Proteome Arrays. Sci Rep 2015; 5:13895. [PMID: 26370624 PMCID: PMC4570193 DOI: 10.1038/srep13895] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Accepted: 06/23/2015] [Indexed: 12/13/2022] Open
Abstract
The heterogeneity and poor prognosis associated with gliomas, makes biomarker identification imperative. Here, we report autoantibody signatures across various grades of glioma serum samples and sub-categories of glioblastoma multiforme using Human Proteome chips containing ~17000 full-length human proteins. The deduced sets of classifier proteins helped to distinguish Grade II, III and IV samples from the healthy subjects with 88, 89 and 94% sensitivity and 87, 100 and 73% specificity, respectively. Proteins namely, SNX1, EYA1, PQBP1 and IGHG1 showed dysregulation across various grades. Sub-classes of GBM, based on its proximity to the sub-ventricular zone, have been reported to have different prognostic outcomes. To this end, we identified dysregulation of NEDD9, a protein involved in cell migration, with probable prognostic potential. Another subcategory of patients where the IDH1 gene is mutated, are known to have better prognosis as compared to patients carrying the wild type gene. On a comparison of these two cohorts, we found STUB1 and YWHAH proteins dysregulated in Grade II glioma patients. In addition to common pathways associated with tumourigenesis, we found enrichment of immunoregulatory and cytoskeletal remodelling pathways, emphasizing the need to explore biochemical alterations arising due to autoimmune responses in glioma.
Collapse
Affiliation(s)
- Parvez Syed
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Shabarni Gupta
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Saket Choudhary
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Narendra Goud Pandala
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Apurva Atak
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Annie Richharia
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Manubhai K P
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Heng Zhu
- Department of Pharmacology and Molecular Sciences/High-Throughput Biology Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sridhar Epari
- Department of Pathology, Tata Memorial Centre, Mumbai 400 012, India
| | - Santosh B Noronha
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Aliasgar Moiyadi
- Department of Neurosurgery, Tata Memorial Centre, Mumbai 400 012, India
| | - Sanjeeva Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| |
Collapse
|
34
|
Chapoval AI, Legutki JB, Stafford P, Trebukhov AV, Johnston SA, Shoikhet YN, Lazarev AF. Immunosignature: Serum Antibody Profiling for Cancer Diagnostics. Asian Pac J Cancer Prev 2015; 16:4833-7. [DOI: 10.7314/apjcp.2015.16.12.4833] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
35
|
Hong Y, Huang J. Autoantibodies against tumor-associated antigens for detection of hepatocellular carcinoma. World J Hepatol 2015; 7:1581-1585. [PMID: 26085917 PMCID: PMC4462696 DOI: 10.4254/wjh.v7.i11.1581] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 01/30/2015] [Accepted: 04/16/2015] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common tumors worldwide. The survival rate after the onset of symptoms is generally less than one year for the late presentation of HCC, and reliable tools for early diagnosis are lacking. Therefore, novel biomarkers for the early detection of HCC are urgently required. Recent studies show that the abnormal release of proteins by tumor cells can elicit humoral immune responses to self-antigens called tumor-associated antigens (TAAs). The corresponding autoantibodies can be detected before the clinical diagnosis of cancer. Therefore, there is growing interest in using serum autoantibodies as cancer biomarkers. In this review, we focus on the advances in research on autoantibodies against TAAs as serum biomarker for detection of HCC, the mechanism of the production of TAAs, and the association of autoantibodies with patients' clinical characteristics.
Collapse
Affiliation(s)
- Yu Hong
- Yu Hong, Jian Huang, Liver Research Center, Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Jian Huang
- Yu Hong, Jian Huang, Liver Research Center, Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| |
Collapse
|
36
|
Immune-Signatures for Lung Cancer Diagnostics: Evaluation of Protein Microarray Data Normalization Strategies. MICROARRAYS 2015; 4:162-87. [PMID: 27600218 PMCID: PMC4996396 DOI: 10.3390/microarrays4020162] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 03/23/2015] [Accepted: 03/25/2015] [Indexed: 01/27/2023]
Abstract
New minimal invasive diagnostic methods for early detection of lung cancer are urgently needed. It is known that the immune system responds to tumors with production of tumor-autoantibodies. Protein microarrays are a suitable highly multiplexed platform for identification of autoantibody signatures against tumor-associated antigens (TAA). These microarrays can be probed using 0.1 mg immunoglobulin G (IgG), purified from 10 µL of plasma. We used a microarray comprising recombinant proteins derived from 15,417 cDNA clones for the screening of 100 lung cancer samples, including 25 samples of each main histological entity of lung cancer, and 100 controls. Since this number of samples cannot be processed at once, the resulting data showed non-biological variances due to “batch effects”. Our aim was to evaluate quantile normalization, “distance-weighted discrimination” (DWD), and “ComBat” for their effectiveness in data pre-processing for elucidating diagnostic immune-signatures. “ComBat” data adjustment outperformed the other methods and allowed us to identify classifiers for all lung cancer cases versus controls and small-cell, squamous cell, large-cell, and adenocarcinoma of the lung with an accuracy of 85%, 94%, 96%, 92%, and 83% (sensitivity of 0.85, 0.92, 0.96, 0.88, 0.83; specificity of 0.85, 0.96, 0.96, 0.96, 0.83), respectively. These promising data would be the basis for further validation using targeted autoantibody tests.
Collapse
|
37
|
Mohamed A, Ayman A, Deniece J, Wang T, Kovach C, Siddiqui MT, Cohen C. P62/Ubiquitin IHC Expression Correlated with Clinicopathologic Parameters and Outcome in Gastrointestinal Carcinomas. Front Oncol 2015; 5:70. [PMID: 25870850 PMCID: PMC4378280 DOI: 10.3389/fonc.2015.00070] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 03/09/2015] [Indexed: 11/30/2022] Open
Abstract
P62 and ubiquitin are small regulatory proteins demonstrated to have implications in the prognosis and survival of various malignancies including: hepatocellular, breast, ovarian, and some gastrointestinal carcinomas. Several trials studied the link of their activity to the extrinsic apoptosis pathway and showed that their autophagy modification has a critical stand point in tumorigenesis. These findings explain their vital role in controlling the process of cell death and survival. It has been shown recently that p62 and ubiquitin overexpression in different types of cancers, such as triple negative breast and ovarian cancers, have directly correlated with incidence of distant metastases. We aim to evaluate p62/ubiquitin expression in gastrointestinal carcinomas of gastric, colonic, and pancreatic origin, and correlate with annotated clinicopathologic data. In gastric carcinoma (61), positive p62 nuclear expression was noted in 57% and cytoplasmic in 61%, while positive ubiquitin was nuclear expressed in 68.8%, and cytoplasmic in 29.5%. In colon carcinoma (45), positive p62 nuclear expression was noted in 29% and cytoplasmic in 71%, while positive ubiquitin was nuclear in 58% and cytoplasmic in 44%. In pancreatic cancer (18), positive p62 nuclear expression was noted in 78% and cytoplasmic in 56%, while positive ubiquitin was nuclear in 83% and cytoplasmic in 72%. Normal gastric (6), colon (4), and pancreatic (4) tissues were negative for both P62 and ubiquitin (nuclear and cytoplasmic staining <20%). Ubiquitin high expression was associated with more lymph node metastases in colon (4.14 vs 1.70, P = 0.04), and pancreatic adenocarcinomas (3.07 vs 0.33, P = 0.03). Also, ubiquitin high expression was associated with worse pancreatic adenocarcinoma overall survival (1.37 vs 2.26 mos, P = 0.04). In addition, gastric cancer patients with high p62 expression tend to have more poorly differentiated grade when compared to those with low expression (21 vs 17, P = 0.04) but less lymph node metastases (2.77 vs 5.73, P = 0.01). P62 and ubiquitin expression did not correlate with other clinicopathologic parameters in gastric, colon or pancreatic denocarcinomas. The results suggest that p62 and ubiquitin are highly expressed in gastric, colonic, and pancreatic carcinomas. High ubiquitin expression was noted to have an impact on number of lymph node metastases in patients with colon and pancreatic adenocarcinomas, but on overall survival only in patients with pancreatic adenocarcinoma. Also, P62 high expression is correlated with poor differentiation, but less lymph node metastases, in gastric carcinoma.
Collapse
Affiliation(s)
- Amr Mohamed
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine , Atlanta, GA , USA ; Department of Medicine, Morehouse School of Medicine , Atlanta, GA , USA
| | - Alkhoder Ayman
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine , Atlanta, GA , USA
| | - Johnson Deniece
- Department of Medicine, Morehouse School of Medicine , Atlanta, GA , USA
| | - Tengteng Wang
- Department of Epidemiology, Rollins School of Public Health, Emory University , Atlanta, GA , USA
| | - Charles Kovach
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine , Atlanta, GA , USA
| | - Momin T Siddiqui
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine , Atlanta, GA , USA
| | - Cynthia Cohen
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine , Atlanta, GA , USA
| |
Collapse
|
38
|
Hong Y, Long J, Li H, Chen S, Liu Q, Zhang B, He X, Wang Y, Li H, Li Y, Zhang T, Lu C, Yan H, Zhang M, Li Q, Cao B, Bai Z, Wang J, Zhang Z, Zhu S, Zheng J, Ou X, Ma H, Jia J, You H, Wang S, Huang J. An Analysis of Immunoreactive Signatures in Early Stage Hepatocellular Carcinoma. EBioMedicine 2015; 2:438-46. [PMID: 26137588 PMCID: PMC4486196 DOI: 10.1016/j.ebiom.2015.03.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 03/10/2015] [Accepted: 03/12/2015] [Indexed: 01/14/2023] Open
Abstract
Background Hepatocellular carcinoma (HCC) is prevalent worldwide and early diagnosis of HCC is critical for effective treatment and optimal prognosis. Methods Serum was screened first by immunoproteomic analysis for HCC-related tumor associated antigens (TAAs). Selected TAAs were clinically evaluated retrospectively in patients with HCC, liver cirrhosis, chronic hepatitis and healthy controls. Levels of autoantibody to the selected TAAs were measured by protein microarrays containing protein antigens of the candidate TAAs. Analyses were done by using receiver operating characteristics (ROC) to calculate diagnostic accuracy. Findings Twenty-two candidate TAAs were assessed by protein microarray analysis in 914 participants with serum α-fetoprotein (AFP) available. Twelve candidate TAAs were statistically different in signal intensity between HCC and controls. Among them, CENPF, HSP60 and IMP-2 showed AUC (area under the curve) values of 0.826, 0.764 and 0.796 respectively for early HCC. The highest prevalence of autoantibody positivity was observed in HCC cases with BCLC tumor stage A, well-differentiated histology and Child-Pugh grade C. Specifically, 73.6% or 79.3% cases of early HCC with negative AFP were positive for autoantibody to CENPF or HSP60. Interpretation Tumor-associated autoimmune reactions may be triggered by early stage HCCs. Measurement of serum autoantibody to TAAs may be complementary to AFP measurements and improve diagnosis of early HCC. Tumor-associated autoimmune reaction may be triggered by early stage HCCs, and TAAs may be potential marker for early HCC. Measurement of autoantibody to TAAs may be complementary to AFP measurements and improve diagnosis of early HCC. Generation of autoantibody to CENPF may result from autoimmune reaction in response to overexpression of CENPF in tumor cell.
Collapse
Affiliation(s)
- Yu Hong
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing, China ; National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Jiang Long
- Department of Oncology Minimally Invasive Interventional Radiology, Beijing You-an Hospital, Capital Medical University, Beijing, China
| | - Hai Li
- Department of Hepatopancreatobiliary and Splenic Medicine, Affiliated Hospital of Medical College of Chinese People's Armed Police Force, Tianjin, China
| | - Shuhong Chen
- Biotechnology Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Qiqi Liu
- Biotechnology Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Bei Zhang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing, China ; National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Xiaomin He
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing, China ; National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Yan Wang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing, China ; National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Hongyi Li
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing, China ; National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Yimei Li
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing, China ; National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Tao Zhang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing, China ; National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Chenzhen Lu
- Department of Hepatopancreatobiliary and Splenic Medicine, Affiliated Hospital of Medical College of Chinese People's Armed Police Force, Tianjin, China
| | - Hao Yan
- Biotechnology Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Minli Zhang
- Biotechnology Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Qing Li
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Bangwei Cao
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhigang Bai
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jin Wang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhongtao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Shengtao Zhu
- Department of Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jiasheng Zheng
- Department of Oncology Minimally Invasive Interventional Radiology, Beijing You-an Hospital, Capital Medical University, Beijing, China
| | - Xiaojuan Ou
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing, China ; National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Hong Ma
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing, China ; National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Jidong Jia
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing, China ; National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Hong You
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing, China ; National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Shengqi Wang
- Biotechnology Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Jian Huang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing, China ; National Clinical Research Center of Digestive Diseases, Beijing, China
| |
Collapse
|
39
|
Abstract
In addition to their historical role, autoantibodies appear promising as biomarkers to facilitate diagnosis, improve patient outcome and decrease mortality in cancer. Autoantibodies may also be useful in the identification of subjects at risk for cancer, that is, those bearing premalignant changes. Numerous studies have demonstrated that cancer serum contains a variety of autoantibodies that react with autologous cellular antigens, that is, tumor-associated antigens. Interestingly, some of these antigens are involved in signal transduction, cell cycle regulation, cell proliferation, and apoptosis. As such, identification of these molecules has additional importance for development of novel anticancer drugs and vaccines. This review focuses on the use of autoantibodies in breast cancer, a major public health problem. We also address the need for additional research to validate this approach in cancer diagnostics and therapeutics in general.
Collapse
|
40
|
Use of autoantibodies to detect the onset of breast cancer. J Immunol Res 2014; 2014:574981. [PMID: 25143958 PMCID: PMC4131063 DOI: 10.1155/2014/574981] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 04/28/2014] [Indexed: 12/12/2022] Open
Abstract
The widespread use of screening mammography has resulted in increased detection of early-stage breast disease, particularly for in situ carcinoma and early-stage breast cancer. However, the majority of women with abnormalities noted on screening mammograms are not diagnosed with cancer because of several factors, including radiologist assessment, patient age, breast density, malpractice concerns, and quality control procedures. Although magnetic resonance imaging is a highly sensitive detection tool that has become standard for women at very high risk of developing breast cancer, it lacks sufficient specificity and costeffectiveness for use as a general screening tool. Therefore, there is an important need to improve screening and diagnosis of early-invasive and noninvasive tumors, that is, in situ carcinoma. The great potential for molecular tools to improve breast cancer outcomes based on early diagnosis has driven the search for diagnostic biomarkers. Identification of tumor-specific markers capable of eliciting an immune response in the early stages of tumor development seems to provide an effective approach for early diagnosis. The aim of this review is to describe several autoantibodies identified during breast cancer diagnosis. We will focus on these molecules highlighted in the past two years and discuss the potential future use of autoantibodies as biomarkers of early-stage breast cancer.
Collapse
|
41
|
Liu X, Ye H, Li L, Li W, Zhang Y, Zhang JY. Humoral autoimmune responses to insulin-like growth factor II mRNA-binding proteins IMP1 and p62/IMP2 in ovarian cancer. J Immunol Res 2014; 2014:326593. [PMID: 24872956 PMCID: PMC4020369 DOI: 10.1155/2014/326593] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 03/23/2014] [Accepted: 04/10/2014] [Indexed: 01/07/2023] Open
Abstract
Ovarian cancer is one of the leading causes of cancer-related deaths among women. There is an urgent need of better approaches for the identification of appropriate biomarkers in the early detection of ovarian cancer. The aim of this study was to elucidate the significance of autoantibodies against insulin-like growth factor II mRNA-binding proteins (IMPs) in patients with ovarian cancer. In this study, autoantibody responses to two members (IMP1 and p62/IMP2) of IMPs were evaluated by enzyme-linked immunosorbent assay (ELISA), western blotting, and indirect immunofluorescence assay in sera from patients with ovarian cancer and normal human individuals. The results have demonstrated that both IMP1 and p62/IMP2 can induce relatively higher frequency of autoantibody responses in patients with ovarian cancer (26.5% and 29.4%) compared to normal individuals (P<0.01). Our preliminary data suggest that IMP1 and p62/IMP2 can stimulate autoimmune responses in ovarian cancer, and anti-IMP1 and anti-p62/IMP2 autoantibodies could be used as potential biomarkers in immunodiagnosis of ovarian cancer.
Collapse
Affiliation(s)
- Xinxin Liu
- Center for Tumor Biotherapy, The First Affiliated Hospital & College of Public Health, Zhengzhou University, Zhengzhou, Henan 450052, China
- Department of Biological Sciences & Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA
| | - Hua Ye
- Center for Tumor Biotherapy, The First Affiliated Hospital & College of Public Health, Zhengzhou University, Zhengzhou, Henan 450052, China
- Department of Biological Sciences & Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA
| | - Liuxia Li
- Center for Tumor Biotherapy, The First Affiliated Hospital & College of Public Health, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Wenjie Li
- Center for Tumor Biotherapy, The First Affiliated Hospital & College of Public Health, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yi Zhang
- Center for Tumor Biotherapy, The First Affiliated Hospital & College of Public Health, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Jian-Ying Zhang
- Center for Tumor Biotherapy, The First Affiliated Hospital & College of Public Health, Zhengzhou University, Zhengzhou, Henan 450052, China
- Department of Biological Sciences & Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA
| |
Collapse
|
42
|
Jia J, Wang W, Meng W, Ding M, Ma S, Wang X. Development of a multiplex autoantibody test for detection of lung cancer. PLoS One 2014; 9:e95444. [PMID: 24755629 PMCID: PMC3995760 DOI: 10.1371/journal.pone.0095444] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2013] [Accepted: 03/27/2014] [Indexed: 12/11/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths for both men and women. Early diagnosis of lung cancer has a 5-year survival rate of 48.8%, however, nearly 35% of stage I patients relapses after surgical resection, thus portending a poor prognosis. Therefore, detecting lung cancer in early stage and further identifying the high-risk patients would allow the opportunity to provide adjuvant therapy and possibly increase survival. There is considerable evidence that the immune system produces an autoantibody response to neoplastic cells. The detection of such autoantibodies has been shown to have diagnostic and prognostic value. Here we took advantage of the high-throughput Luminex technique to multiplex a total of 14 tumor-associated autoantigens to detect the autoantibody from the patients sera. The 14 antigens were expressed by in vitro transcription/translation system with HaloTag at N-terminus. The fusion proteins were then covalently immobilized onto the Luminex microspheres conjugated by the halo-link ligand, thus eliminating the protein purification procedure. Sera samples from cancer patients and healthy controls were interacted with the microsphere-antigen complex to measure the autoantibodies. We have developed a quick multiplex detection system for measuring autoantibody signature from patient sera with minimal cross-reaction. A panel of seven autoantibody biomarkers has generated an AUC>80% in distinguishing the lung cancers from healthy controls. This study is the first report by combining Luminex platform and HaloTag technology to detect humoral immune response in cancer patients. Due to the flexibility of the Luminex technology, this approach can be applied to others conditions such as infectious, neurological, and metabolic diseases. One can envision that this multiplex Luminex system as well as the panel of seven biomarkers could be used to screen the high-risk population with subsequent CT test based on the blood test result.
Collapse
Affiliation(s)
- Jing Jia
- Center for Molecular Medicine, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang, P. R. China
| | - Wenzhe Wang
- Center for Molecular Medicine, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang, P. R. China
| | - Wen Meng
- Hangzhou First People’s Hospital, Hangzhou, Zhejiang, P. R. China
| | - Mingjian Ding
- Center for Molecular Medicine, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang, P. R. China
| | - Shenglin Ma
- Institute of Lung Cancer, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang, P. R. China
- Hangzhou First People’s Hospital, Hangzhou, Zhejiang, P. R. China
- * E-mail: (XW); (SM)
| | - Xiaoju Wang
- Center for Molecular Medicine, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang, P. R. China
- Institute of Lung Cancer, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang, P. R. China
- * E-mail: (XW); (SM)
| |
Collapse
|
43
|
Preferential autoimmune response in prostate cancer to cyclin B1 in a panel of tumor-associated antigens. J Immunol Res 2014; 2014:827827. [PMID: 24860838 PMCID: PMC4016862 DOI: 10.1155/2014/827827] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 03/28/2014] [Indexed: 12/22/2022] Open
Abstract
Previous studies have demonstrated that sera from patients with prostate cancer (PCa) contain autoantibodies that react with tumor-associated antigens (TAAs). Autoantibodies to cyclin B1 and fourteen other TAAs were detected by enzyme-linked immunosorbent assay (ELISA) and Western blotting in 464 sera from patients with PCa, benign prostatic hyperplasia (BPH), and other controls. Autoantibodies to cyclin B1 were detected in 31.0% of sera from randomly selected patients with PCa versus 4.8% in sera with BPH. In the further analysis, 31.4% of sera from PCa patients at the early stage contained anti-cyclin B1 autoantibody, and even 29.4% of patients who had normal prostate-specific antigen (PSA) levels in their serum samples were observed anti-cyclin B1 positive. The cumulative positive rate of autoantibodies against seven selected TAAs (cyclin B1, survivin, p53, DFS70/LEDGFp75, RalA, MDM2, and NPM1) in PCa reached 80.5%, significantly higher than that in normal control sera. In summary, autoantibody to cyclin B1 might be a potential biomarker for the immunodiagnosis of early stage PCa, especially useful in patients with normal PSA level. This study further supports the hypothesis that a customized TAA array can be used for enhancing anti-TAA autoantibody detection, and it may constitute a promising and powerful tool for immunodiagnosis of PCa.
Collapse
|
44
|
Evans RL, Pottala JV, Egland KA. Classifying patients for breast cancer by detection of autoantibodies against a panel of conformation-carrying antigens. Cancer Prev Res (Phila) 2014; 7:545-55. [PMID: 24641868 DOI: 10.1158/1940-6207.capr-13-0416] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Patients with breast cancer elicit an autoantibody response against cancer proteins, which reflects and amplifies the cellular changes associated with tumorigenesis. Detection of autoantibodies in plasma may provide a minimally invasive mechanism for early detection of breast cancer. To identify cancer proteins that elicit a humoral response, we generated a cDNA library enriched for breast cancer genes that encode membrane and secreted proteins, which are more likely to induce an antibody response compared with intracellular proteins. To generate conformation-carrying antigens that are efficiently recognized by patients' antibodies, a eukaryotic expression strategy was established. Plasma from 200 patients with breast cancer and 200 age-matched healthy controls were measured for autoantibody activity against 20 different antigens designed to have conformational epitopes using ELISA. A conditional logistic regression model was used to select a combination of autoantibody responses against the 20 different antigens to classify patients with breast cancer from healthy controls. The best combination included ANGPTL4, DKK1, GAL1, MUC1, GFRA1, GRN, and LRRC15; however, autoantibody responses against GFRA1, GRN, and LRRC15 were inversely correlated with breast cancer. When the autoantibody responses against the 7 antigens were added to the base model, including age, BMI, race and current smoking status, the assay had the following diagnostic capabilities: c-stat (95% CI), 0.82 (0.78-0.86); sensitivity, 73%; specificity, 76%; and positive likelihood ratio (95% CI), 3.04 (2.34-3.94). The model was calibrated across risk deciles (Hosmer-Lemeshow, P = 0.13) and performed well in specific subtypes of breast cancer including estrogen receptor positive, HER-2 positive, invasive, in situ and tumor sizes >1 cm.
Collapse
Affiliation(s)
- Rick L Evans
- 2301 East 60th Street North, Sioux Falls, SD 57104.
| | | | | |
Collapse
|
45
|
Circulating autoantibodies to LGALS3BP: a novel biomarker for cancer. DISEASE MARKERS 2013; 35:747-52. [PMID: 24347795 PMCID: PMC3850626 DOI: 10.1155/2013/214595] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 10/11/2013] [Accepted: 10/11/2013] [Indexed: 12/24/2022]
Abstract
Purpose. Circulating autoantibodies have been extensively investigated as possible markers for early diagnosis of cancer. The present study was carried out to investigate whether anti-LGALS3BP IgG autoantibodies could be classified as a biomarker for malignant tumors. Methods. An in-house developed enzyme-linked immunosorbent assay was used to detect autoantibodies to LGALS3BP in sera from 71 patients with various types of cancers and 54 healthy subjects matched by age and gender. Results. Patients with cancer have significant higher circulating levels of anti-LGALS3BP antibodies as compared to control subjects (P < 0.001). The test has a sensitivity of 33% and a specificity of 98%. Conclusions. Anti-LGALS3BP IgG autoantibodies are a promising biomarker for malignant tumors and could play a role in the development of a multimarker assay for the early detection of cancer.
Collapse
|
46
|
Ostendorff HP, Awad A, Braunschweiger KI, Liu Z, Wan Z, Rothschild KJ, Lim MJ. Multiplexed VeraCode bead-based serological immunoassay for colorectal cancer. J Immunol Methods 2013; 400-401:58-69. [PMID: 24161315 DOI: 10.1016/j.jim.2013.09.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 09/13/2013] [Accepted: 09/17/2013] [Indexed: 02/06/2023]
Abstract
Colorectal cancer (CRC) is the second leading cause of cancer deaths in the US and Western world. Despite increased screening and advances in treatment, the mortality rate (ca. 50,000/year) and high national health-care burden for CRC are likely to remain high unless an effective non-invasive screening test for CRC is instituted for a large segment of the population. Blood-based protein biomarkers hold great promise for early disease diagnosis and personalized medicine; yet robust and reproducible multiplexing platforms and methodologies have lagged behind their genomic counterparts. Here, we report the development of a novel, multiplexed, hybrid immunoassay for CRC that is formatted on barcoded VeraCode™ micro-beads, which have until now only been used for genomic assays. The method combines a sandwich immunoassay format for detection of serum protein biomarkers with an antigen assay for autoantibody detection. The serum protein biomarkers CEA and GDF15 as well as autoantibodies to the p53 tumor associated antigen (TAA) were used to exemplify the method. This multiplex biomarker panel was configured to run on Illumina's holographically barcoded VeraCode™ micro-bead platform, which is capable of measuring hundreds of analytes simultaneously in a single well from small volumes of blood (<50 μL) using a 96-well industry standard microtiter plate. This novel use of the VeraCode™ micro-bead platform translates into a potentially low volume, high throughput, multiplexed assay for CRC, for the purposes of biomarker validation, as well as patient screening, diagnostics and prognostics. In an evaluation of a 186 patient sera training set (CRC and normal), we obtained a diagnostic sensitivity of 54% and a specificity of 98%. We anticipate that by expanding and refining the biomarkers in this initial panel, and performing more extensive clinical validations, such an assay could ultimately provide a basis for CRC population screening to complement the more invasive, expensive and low throughput (but highly sensitive and specific) colonoscopy.
Collapse
|
47
|
Gao H, Zheng Z, Mao Y, Wang W, Qiao Y, Zhou L, Liu F, He H, Zhao X. Identification of tumor antigens that elicit a humoral immune response in the sera of Chinese esophageal squamous cell carcinoma patients by modified serological proteome analysis. Cancer Lett 2013; 344:54-61. [PMID: 24157810 DOI: 10.1016/j.canlet.2013.10.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 10/11/2013] [Accepted: 10/14/2013] [Indexed: 12/01/2022]
Abstract
Our aim was to identify novel tumor-associated antigens from the esophageal squamous cell carcinoma (ESCC) cell line EC0156, and related autoantibodies in sera from patients with ESCC. We used modified serological proteome analysis, involving one- and two-dimensional electrophoresis, Western blot, and MALDI-TOF/TOF-MS to identify 6 ESCC-associated antigens. From these, 105 kDa heat shock protein (HSP105) and triosephosphate isomerase (TIM) were further evaluated and we determined they could induce autoantibody responses in ESCC sera and are highly expressed in ESCC tissues. Anti-HSP105 and anti-TIM autoantibodies were found in 39.1% (18/46) and 34.8% (16/46) of patients with ESCC, respectively, but only in two controls. A receiver operating characteristic curve constructed with HSP105 and TIM gave a sensitivity of 54.3% and 95% (38/40) specificity in discriminating ESCC from matched controls. Interestingly, we found that autoantibodies against TIM in ESCC serum mainly reacted with glycosylated but not deglycosylated TIM. The preliminary results suggest the potential utility of screening autoantibodies in sera for use as biomarkers for cancer diagnosis.
Collapse
Affiliation(s)
- Hongjun Gao
- Clinical Laboratory of China Meitan General Hospital, Beijing, PR China
| | - Zhaoxu Zheng
- Department of Abdominal Surgery, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Yousheng Mao
- Department of Thoracic Surgery, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Wei Wang
- Department of Thoracic Surgery, Navy General Hospital of Chinese PLA, Beijing, PR China
| | - Yuanyuan Qiao
- Center for Basic Medical Science, Navy General Hospital of Chinese PLA, Beijing, PR China
| | - Lanping Zhou
- State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Fang Liu
- State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Hongzhi He
- State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Xiaohang Zhao
- Center for Basic Medical Science, Navy General Hospital of Chinese PLA, Beijing, PR China; State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China.
| |
Collapse
|
48
|
Whittemore K, Sykes K. A microarray method for identifying tumor antigens by screening a tumor cDNA expression library against cancer sera. Hum Vaccin Immunother 2013; 9:2178-88. [PMID: 23851590 PMCID: PMC3906402 DOI: 10.4161/hv.25634] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 06/18/2013] [Accepted: 07/04/2013] [Indexed: 11/19/2022] Open
Abstract
The immune system responds to tumor cells. The challenge has been how to effectively use these responses to treat or protect against cancer. Toward the goal of developing a cancer vaccine, we are pursuing methodologies for the discovery and testing of useful antigens. We present an array-based approach for discovering these B cell antigens by directly screening for specific host-sera reactivity to lysates from tumor-derived cDNA expression libraries. Several cancer-specific antigens were identified, and these are currently being validated as potential candidates.
Collapse
Affiliation(s)
- Kurt Whittemore
- Center for Innovations in Medicine; The Biodesign Institute; Arizona State University; Tempe, AZ USA
- Biological Design Program; Arizona State University; Tempe, AZ USA
| | - Kathryn Sykes
- Center for Innovations in Medicine; The Biodesign Institute; Arizona State University; Tempe, AZ USA
- HealthTell, Inc.; Chandler, AZ USA
| |
Collapse
|
49
|
Zaenker P, Ziman MR. Serologic autoantibodies as diagnostic cancer biomarkers--a review. Cancer Epidemiol Biomarkers Prev 2013; 22:2161-81. [PMID: 24057574 DOI: 10.1158/1055-9965.epi-13-0621] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Current diagnostic techniques used for the early detection of cancers are successful but subject to detection bias. A recent focus lies in the development of more accurate diagnostic tools. An increase in serologic autoantibody levels has been shown to precede the development of cancer disease symptoms. Therefore, autoantibody levels in patient blood serum have been proposed as diagnostic biomarkers for early-stage diagnosis of cancers. Their clinical application has, however, been hindered by low sensitivity, specificity, and low predictive value scores. These scores have been shown to improve when panels of multiple diagnostic autoantibody biomarkers are used. A five-marker biomarker panel has been shown to increase the sensitivity of prostate cancer diagnosis to 95% as compared with 12.2% for prostate-specific antigen alone. New potential biomarker panels were also discovered for lung, colon, and stomach cancer diagnosis with sensitivity of 76%, 65.4%, and 50.8%, respectively. Studies in breast and liver cancer, however, seem to favor single markers, namely α-2-HS-glycoprotein and des-γ-carboxyprothrombin with sensitivities of 79% and 89% for the early detection of the cancers. The aim of this review is to discuss the relevance of autoantibodies in cancer diagnosis and to outline the current methodologies used in the detection of autoantibodies. The review concludes with a discussion of the autoantibodies currently used in the diagnosis of cancers of the prostate, breast, lung, colon, stomach, and liver. A discussion of the potential future use of autoantibodies as diagnostic cancer biomarkers is also included in this review.
Collapse
Affiliation(s)
- Pauline Zaenker
- Authors' Affiliations: School of Medical Sciences, Edith Cowan University, Perth; and Department of Pathology and Laboratory Medicine, The University of Western Australia, Crawley, Western Australia, Australia
| | | |
Collapse
|
50
|
Zhang Y, Yang D, Weng L, Wang L. Early lung cancer diagnosis by biosensors. Int J Mol Sci 2013; 14:15479-509. [PMID: 23892596 PMCID: PMC3759869 DOI: 10.3390/ijms140815479] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Revised: 06/29/2013] [Accepted: 07/04/2013] [Indexed: 12/20/2022] Open
Abstract
Lung cancer causes an extreme threat to human health, and the mortality rate due to lung cancer has not decreased during the last decade. Prognosis or early diagnosis could help reduce the mortality rate. If microRNA and tumor-associated antigens (TAAs), as well as the corresponding autoantibodies, can be detected prior to clinical diagnosis, such high sensitivity of biosensors makes the early diagnosis and prognosis of cancer realizable. This review provides an overview of tumor-associated biomarker identifying methods and the biosensor technology available today. Laboratorial researches utilizing biosensors for early lung cancer diagnosis will be highlighted.
Collapse
Affiliation(s)
- Yuqian Zhang
- Key Laboratory for Organic Electronics & Information Displays (KLOEID) and Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, China; E-Mails: (Y.Z.); (D.Y.)
| | - Dongliang Yang
- Key Laboratory for Organic Electronics & Information Displays (KLOEID) and Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, China; E-Mails: (Y.Z.); (D.Y.)
| | - Lixing Weng
- College of Geography and Biological Information, Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, China
| | - Lianhui Wang
- Key Laboratory for Organic Electronics & Information Displays (KLOEID) and Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, China; E-Mails: (Y.Z.); (D.Y.)
| |
Collapse
|