151
|
Mitoma H, Manto M, Hampe CS. Immune-mediated Cerebellar Ataxias: Practical Guidelines and Therapeutic Challenges. Curr Neuropharmacol 2019; 17:33-58. [PMID: 30221603 PMCID: PMC6341499 DOI: 10.2174/1570159x16666180917105033] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 07/06/2018] [Accepted: 09/03/2018] [Indexed: 12/11/2022] Open
Abstract
Immune-mediated cerebellar ataxias (IMCAs), a clinical entity reported for the first time in the 1980s, include gluten ataxia (GA), paraneoplastic cerebellar degenerations (PCDs), antiglutamate decarboxylase 65 (GAD) antibody-associated cerebellar ataxia, post-infectious cerebellitis, and opsoclonus myoclonus syndrome (OMS). These IMCAs share common features with regard to therapeutic approaches. When certain factors trigger immune processes, elimination of the antigen( s) becomes a priority: e.g., gluten-free diet in GA and surgical excision of the primary tumor in PCDs. Furthermore, various immunotherapeutic modalities (e.g., steroids, immunoglobulins, plasmapheresis, immunosuppressants, rituximab) should be considered alone or in combination to prevent the progression of the IMCAs. There is no evidence of significant differences in terms of response and prognosis among the various types of immunotherapies. Treatment introduced at an early stage, when CAs or cerebellar atrophy is mild, is associated with better prognosis. Preservation of the "cerebellar reserve" is necessary for the improvement of CAs and resilience of the cerebellar networks. In this regard, we emphasize the therapeutic principle of "Time is Cerebellum" in IMCAs.
Collapse
Affiliation(s)
- Hiroshi Mitoma
- Address correspondence to this author at the Medical Education Promotion Center, Tokyo Medical University, Tokyo, Japan;, E-mail:
| | | | | |
Collapse
|
152
|
Baldin AV, Grishina AN, Korolev DO, Kuznetsova EB, Golovastova MO, Kalpinskiy AS, Alekseev BY, Kaprin AD, Zinchenko DV, Savvateeva LV, Varshavsky VA, Zernii EY, Vinarov AZ, Bazhin AV, Philippov PP, Zamyatnin AA. Autoantibody against arrestin-1 as a potential biomarker of renal cell carcinoma. Biochimie 2018; 157:26-37. [PMID: 30389514 DOI: 10.1016/j.biochi.2018.10.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 10/25/2018] [Indexed: 12/28/2022]
Abstract
Renal cell carcinoma (RCC) is the second-most common uronephrological cancer. In the absence of specific symptoms, early diagnosis of RCC is challenging. Monitoring of the aberrant expression of tumour-associated antigens (TAAs) and related autoantibody response is considered as a novel approach of RCC diagnostics. The aim of this study was to examine the aberrant expression of arrestin-1 in renal tumours, to investigate the possible epigenetic mechanism underlying arrestin-1 expression, and to assess the frequency of anti-arrestin-1 autoantibody response. Immunohistochemistry was used to assess the presence of arrestin-1 in primary tumours and metastases of 39 patients with RCC and renal oncocytoma. Bisulfite sequencing was employed to analyse the methylation status of the promoter of the SAG gene encoding arrestin-1. Western blot analysis was performed to detect autoantibodies against arrestin-1 in serum samples of 36 RCC and oncocytoma patients. Arrestin-1 was found to be expressed in RCC (58.7% of cases) and renal oncocytoma (90% of cases) cells, while being absent in healthy kidney. The expression of arrestin-1 in RCC metastases was more prominent than in primary tumours. Hypomethylation of the SAG gene promoter is unlikely to be the mechanism for the aberrant expression of arrestin-1. Autoantibodies against arrestin-1 were detected in sera of 75% of RCC patients. Taken together, our findings suggest employment of autoantibody against arrestin-1 as biomarker of RCC.
Collapse
Affiliation(s)
- Alexey V Baldin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991, Moscow, Russia.
| | - Alena N Grishina
- Anatomic Pathology Department, Sechenov First Moscow State Medical University, 119991, Moscow, Russia.
| | - Dmitry O Korolev
- Institute of Uronephrology and Human Reproductive Health, Sechenov First Moscow State Medical University, 119991, Moscow, Russia
| | - Ekaterina B Kuznetsova
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991, Moscow, Russia; Research Centre for Medical Genetics, 115522, Moscow, Russia.
| | - Marina O Golovastova
- Department of Cell Signalling, Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991, Moscow, Russia
| | - Alexey S Kalpinskiy
- P.A. Hertzen Moscow Oncology Research Center, National Medical Research Center of Radiology, 125284, Moscow, Russia
| | - Boris Y Alekseev
- P.A. Hertzen Moscow Oncology Research Center, National Medical Research Center of Radiology, 125284, Moscow, Russia
| | - Andrey D Kaprin
- P.A. Hertzen Moscow Oncology Research Center, National Medical Research Center of Radiology, 125284, Moscow, Russia.
| | - Dmitry V Zinchenko
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Pushchino, Moscow Region, 142290 Russia.
| | - Lyudmila V Savvateeva
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991, Moscow, Russia
| | - Vladimir A Varshavsky
- Anatomic Pathology Department, Sechenov First Moscow State Medical University, 119991, Moscow, Russia
| | - Evgeni Yu Zernii
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991, Moscow, Russia; Department of Cell Signalling, Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991, Moscow, Russia.
| | - Andrey Z Vinarov
- Institute of Uronephrology and Human Reproductive Health, Sechenov First Moscow State Medical University, 119991, Moscow, Russia.
| | - Alexandr V Bazhin
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany.
| | - Pavel P Philippov
- Department of Cell Signalling, Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991, Moscow, Russia
| | - Andrey A Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991, Moscow, Russia; Department of Cell Signalling, Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991, Moscow, Russia.
| |
Collapse
|
153
|
Music M, Prassas I, Diamandis EP. Optimizing cancer immunotherapy: Is it time for personalized predictive biomarkers? Crit Rev Clin Lab Sci 2018; 55:466-479. [PMID: 30277835 DOI: 10.1080/10408363.2018.1499706] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cancer immunotherapy, a treatment that selectively augments a patient's anti-tumor immune response, is a breakthrough advancement in personalized medicine. A subset of cancer patients undergoing immunotherapy have displayed robust and long-lasting therapeutic responses. Currently, the spotlight is on the use of blocking antibodies against the T-cell checkpoint molecules, cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and programed cell death-1 (PD-1)/programed death-ligand 1 (PD-L1), which have been effectively used to combat many cancers types. Despite the overall enthusiasm, immune checkpoint blockade inhibitors suffer from significant limitations such as high cost, serious toxicity in a substantial proportion of patients, and a response rate as low as 10%-40% in some clinical trials. Consequently, there is an urgent and unmet medical need for companion biomarkers that could both predict the response of individual patients to these therapies, and provide the means for precise monitoring of their therapeutic outcome. In this era of precision medicine, predictive biomarkers are a hot commodity because they can effectively separate responders from non-responders, and spare non-responders from serious therapy-related toxicity. Emerging predictive biomarkers for immune checkpoint blockade are: PD-L1 expression, increased amounts of tumor-infiltrating lymphocytes, increased mutational load and mismatch repair deficiency. Other well-studied biomarkers include inflammatory infiltrate, absolute lymphocyte count and lactate dehydrogenase levels. We review recent progress on predictive cancer biomarkers in immunotherapy, with a special emphasis on serum autoantibodies that have the potential to be personalized for optimal clinical outcomes.
Collapse
Affiliation(s)
- Milena Music
- a Department of Laboratory Medicine and Pathobiology , University of Toronto , Toronto , Canada
| | - Ioannis Prassas
- b Department of Pathology and Laboratory Medicine , Mount Sinai Hospital , Toronto , Canada
| | - Eleftherios P Diamandis
- a Department of Laboratory Medicine and Pathobiology , University of Toronto , Toronto , Canada.,b Department of Pathology and Laboratory Medicine , Mount Sinai Hospital , Toronto , Canada.,c Department of Clinical Biochemistry , University Health Network , Toronto , Canada.,d Lunenfeld-Tanenbaum Research Institute , Mount Sinai Hospital , Toronto , Canada
| |
Collapse
|
154
|
ARORA RICHA, SAXENA SONAL, SHRIVASTAVA SAMEER, SINGH PIYUSHKUMAR, HUSSAIN SHAHID, KUMAR MANOJ, JENA SUBASCHANDRA, KUMAWAT MANOJ, SINGH RAJKUMARJAMES, GOGOI PURNIMA, SAHOO MONALISA, KUMAR NAVEEN. Genetic characterization and phylogenetic analysis of triose phosphate isomerase (TPI) gene amplified from a case of canine mammary tumour showing TPI over-expression and high titre anti-TPI antibodies. THE INDIAN JOURNAL OF ANIMAL SCIENCES 2018. [DOI: 10.56093/ijans.v88i9.83549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
155
|
B cells and antibody production in melanoma. Mamm Genome 2018; 29:790-805. [DOI: 10.1007/s00335-018-9778-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 08/24/2018] [Indexed: 01/12/2023]
|
156
|
Betteridge ZE, Priest L, Cooper RG, McHugh NJ, Blackhall F, Lamb JA. Investigation of myositis and scleroderma specific autoantibodies in patients with lung cancer. Arthritis Res Ther 2018; 20:176. [PMID: 30092841 PMCID: PMC6085683 DOI: 10.1186/s13075-018-1678-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 07/17/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The close temporal association between onset of some connective tissue diseases and cancer suggests a paraneoplastic association. Adult patients with scleroderma with anti-RNA polymerase III autoantibodies and adult patients with dermatomyositis with anti-transcriptional intermediary factor 1 (anti-TIF1) or anti-nuclear matrix protein 2 (anti-NXP2) autoantibodies have a significantly increased risk of developing cancer. Autoantibodies may serve as biomarkers for early detection of cancer and also could be relevant for prediction of responses to immune therapies. We aimed to test whether myositis and scleroderma specific or associated autoantibodies are detectable in individuals with lung cancer. METHODS Serum from 60 Caucasian patients with lung cancer (30 with small cell lung cancer, 30 with non-small cell lung cancer) was screened for myositis and scleroderma specific and associated autoantibodies by radiolabelled immunoprecipitation. RESULTS Anti-TIF1, anti-NXP2 or anti-RNA polymerase III autoantibodies were not detected in any of the 60 patients with lung cancer. Anti-glycyl-transfer RNA (tRNA) synthetase (anti-EJ) autoantibodies were detected in one patient with non-small cell lung cancer. No other known myositis or scleroderma autoantibodies were identified. CONCLUSIONS Myositis and scleroderma specific autoantibodies, including anti-TIF1, anti-NXP2 and anti-RNA polymerase III, are rare in patients with lung cancer without an autoimmune disease. We report here the first case of anti-EJ autoantibodies being detected in a patient with lung cancer without clinical or radiographic evidence of the anti-synthetase syndrome.
Collapse
Affiliation(s)
- Zoe E Betteridge
- Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| | - Lynsey Priest
- Division of Molecular and Clinical Cancer Sciences, University of Manchester, Manchester, UK
| | - Robert G Cooper
- MRC/ARUK Centre for Integrated Research into Musculoskeletal Ageing, University of Liverpool, Liverpool, UK
| | - Neil J McHugh
- Department of Pharmacy and Pharmacology, University of Bath, Bath, UK.,Royal National Hospital for Rheumatic Diseases, Royal United Hospitals Foundation Trust, Bath, UK
| | - Fiona Blackhall
- Division of Molecular and Clinical Cancer Sciences, University of Manchester, Manchester, UK.,CRUK Lung Cancer Centre of Excellence, The Christie NHS Foundation Trust, Wilmslow Road, Manchester, UK
| | - Janine A Lamb
- Centre for Epidemiology, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK.
| |
Collapse
|
157
|
A diagnostic autoantibody signature for primary cutaneous melanoma. Oncotarget 2018; 9:30539-30551. [PMID: 30093967 PMCID: PMC6078131 DOI: 10.18632/oncotarget.25669] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 06/04/2018] [Indexed: 01/16/2023] Open
Abstract
Melanoma is an aggressive form of skin cancer that is curable by surgical excision in the majority of cases, if detected at an early stage. To improve early stage melanoma detection, the development of a highly sensitive diagnostic test is of utmost importance. Here we aimed to identify antibodies to a panel of tumour associated antigens that can differentiate primary melanoma patients and healthy individuals. A total of 245 sera from primary melanoma patients and healthy volunteers were screened against a high-throughput microarray platform containing 1627 functional proteins. Following rigorous statistical analysis, we identified a combination of 10 autoantibody biomarkers that, as a panel, displays a sensitivity of 79%, specificity of 84% and an AUC of 0.828 for primary melanoma detection. This melanoma autoantibody signature may prove valuable for the development of a diagnostic blood test for routine population screening that, when used in conjunction with current melanoma diagnostic techniques, could improve the early diagnosis of this malignancy and ultimately decrease the mortality rate of patients.
Collapse
|
158
|
Autoantibodies as Potential Biomarkers in Breast Cancer. BIOSENSORS-BASEL 2018; 8:bios8030067. [PMID: 30011807 PMCID: PMC6163859 DOI: 10.3390/bios8030067] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 07/11/2018] [Accepted: 07/11/2018] [Indexed: 12/13/2022]
Abstract
Breast cancer is a major cause of mortality in women; however, technologies for early stage screening and diagnosis (e.g., mammography and other imaging technologies) are not optimal for the accurate detection of cancer. This creates demand for a more effective diagnostic means to replace or be complementary to existing technologies for early discovery of breast cancer. Cancer neoantigens could reflect tumorigenesis, but they are hardly detectable at the early stage. Autoantibodies, however, are biologically amplified and hence may be measurable early on, making them promising biomarkers to discriminate breast cancer from healthy tissue accurately. In this review, we summarized the recent findings of breast cancer specific antigens and autoantibodies, which may be useful in early detection, disease stratification, and monitoring of treatment responses of breast cancer.
Collapse
|
159
|
Hwang HM, Heo CK, Lee HJ, Kwak SS, Lim WH, Yoo JS, Yu DY, Lim KJ, Kim JY, Cho EW. Identification of anti-SF3B1 autoantibody as a diagnostic marker in patients with hepatocellular carcinoma. J Transl Med 2018; 16:177. [PMID: 29954402 PMCID: PMC6025833 DOI: 10.1186/s12967-018-1546-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 06/12/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Tumor-associated (TA) autoantibodies, which are generated by the immune system upon the recognition of abnormal TA antigens, are promising biomarkers for the early detection of tumors. In order to detect autoantibody biomarkers effectively, antibody-specific epitopes in the diagnostic test should maintain the specific conformations that are as close as possible to those presenting in the body. However, when using patients' serum as a source of TA autoantibodies the characterization of the autoantibody-specific epitope is not easy due to the limited amount of patient-derived serum. METHODS To overcome these limits, we constructed a B cell hybridoma pool derived from a hepatocellular carcinoma (HCC) model HBx-transgenic mouse and characterized autoantibodies derived from them as tumor biomarkers. Their target antigens were identified by mass spectrometry and the correlations with HCC were examined. With the assumption that TA autoantibodies generated in the tumor mouse model are induced in human cancer patients, the enzyme-linked immunosorbent assays (ELISA) based on the characteristics of mouse TA autoantibodies were developed for the detection of autoantibody biomarkers in human serum. To mimic natural antigenic structures, the specific epitopes against autoantibodies were screened from the phage display cyclic random heptapeptide library, and the streptavidin antigens fused with the specific epitopes were used as coating antigens. RESULTS In this study, one of HCC-associated autoantibodies derived from HBx-transgenic mouse, XC24, was characterized. Its target antigen was identified as splicing factor 3b subunit 1 (SF3B1) and the high expression of SF3B1 was confirmed in HCC tissues. The specific peptide epitopes against XC24 were selected and, among them, XC24p11 cyclic peptide (-CDATPPRLC-) was used as an epitope of anti-SF3B1 autoantibody ELISA. With this epitope, we could effectively distinguish between serum samples from HCC patients (n = 102) and healthy subjects (n = 85) with 73.53% sensitivity and 91.76% specificity (AUC = 0.8731). Moreover, the simultaneous detection of anti-XC24p11 epitope autoantibody and AFP enhanced the efficiency of HCC diagnosis with 87.25% sensitivity and 90.59% specificity (AUC = 0.9081). CONCLUSIONS ELISA using XC24p11 peptide epitope that reacts against anti-SF3B1 autoantibody can be used as a novel test to enhance the diagnostic efficiency of HCC.
Collapse
Affiliation(s)
- Hai-Min Hwang
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141 South Korea
- Department of Microbiology and Molecular Biology, College of Bioscience and Biotechnology, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134 South Korea
| | - Chang-Kyu Heo
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141 South Korea
- Department of Microbiology and Molecular Biology, College of Bioscience and Biotechnology, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134 South Korea
| | - Hye Jung Lee
- Proteometech Inc., 1101 Wooree Venture Town, 466 Gangseo-ro, Gangseo-gu, Seoul, 07573 South Korea
- Graduate Program for Nanomedical Science, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722 South Korea
| | - Sang-Seob Kwak
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141 South Korea
- Department of Functional Genomics, University of Science and Technology, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141 South Korea
| | - Won-Hee Lim
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141 South Korea
- Department of Functional Genomics, University of Science and Technology, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141 South Korea
| | - Jong-Shin Yoo
- Biomedical Omics Group, Korea Basic Science Institute, 162 YeonGuDanji-ro, Ochang-eup, Cheongju, Chungbuk 28119 South Korea
| | - Dae-Yuel Yu
- Disease Model Research Laboratory, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141 South Korea
| | - Kook Jin Lim
- Proteometech Inc., 1101 Wooree Venture Town, 466 Gangseo-ro, Gangseo-gu, Seoul, 07573 South Korea
- Graduate Program for Nanomedical Science, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722 South Korea
| | - Jeong-Yoon Kim
- Department of Microbiology and Molecular Biology, College of Bioscience and Biotechnology, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134 South Korea
| | - Eun-Wie Cho
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141 South Korea
- Department of Functional Genomics, University of Science and Technology, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141 South Korea
| |
Collapse
|
160
|
Pashov A, Hernandez Puente CV, Ibrahim SM, Monzavi-Karbassi B, Makhoul I, Kieber-Emmons T. Thinking Cancer. Monoclon Antib Immunodiagn Immunother 2018; 37:117-125. [PMID: 29939836 DOI: 10.1089/mab.2018.0014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Evolutionary theories are necessarily invoked for understanding cancer development at the level of species, at the level of cells and tissues, and for developing effective therapies. It is crucial to view cancer in a Darwinian light, where the differential survival of individual cells is based on heritable variations. In the process of this somatic evolution, multicellularity controls are overridden by cancer cells, which become increasingly autonomous. Ecological epigenetics also helps understand how rogue cells that have basically the same DNA as their normal cell counterpart overcome the tissue homeostasis. As we struggle to wrap our minds around the complexity of these phenomena, we apply often times anthropomorphic terms, such as subversion, hijacking, or hacking, to describe especially the most complex among them-the interaction of tumors with the immune system. In this commentary we highlight examples of the anthropomorphic thinking of cancer and try to put into context the relative meaning of terms and the mechanisms that are oftentimes invoked to justify those terms.
Collapse
Affiliation(s)
- Anastas Pashov
- 1 Stephan Angelov Institute of Microbiology , Bulgarian Academy of Sciences, Sofia, Bulgaria
| | | | | | - Behjatolah Monzavi-Karbassi
- 3 Department of Pathology, University of Arkansas for Medical Sciences , Little Rock, Arkansas
- 4 Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences , Little Rock, Arkansas
| | - Issam Makhoul
- 4 Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences , Little Rock, Arkansas
- 5 Department of Medicine, University of Arkansas for Medical Sciences , Little Rock, Arkansas
| | - Thomas Kieber-Emmons
- 3 Department of Pathology, University of Arkansas for Medical Sciences , Little Rock, Arkansas
- 4 Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences , Little Rock, Arkansas
| |
Collapse
|
161
|
Zhang JB, Cao M, Chen J, Ye SR, Xie K, He X, Ma XL, Zhang J, Yie SM. Serum anti-TOPO48 autoantibody as a biomarker for early diagnosis and prognosis in patients with esophageal squamous cell carcinoma. Clin Res Hepatol Gastroenterol 2018; 42:276-284. [PMID: 29170084 DOI: 10.1016/j.clinre.2017.09.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Revised: 09/03/2017] [Accepted: 09/21/2017] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND AIM We previously reported a novel tumor associated antigen (TTA) with molecular weight around 48kDa that is a fragment derived from human DNA-topoiomerase I (TOP1). We termed the novel TAA as TOPO48 and termed autoantibody against the TAA as anti-TOPO48 autoantibody. The aim of this study is to further investigate the clinical applications of the autoantibody in patients with esophageal squamous cell carcinomas (ESCC). METHODS Serum levels of the anti-TOPO48 autoantibody in 112 ESCC patients, 112 age- and gender-matched healthy controls and 75 patients with esophageal benign tumors were determined by using a specific anti-TOPO48 autoantibody ELISA. Then, we statistically evaluated its clinical significance. RESULTS We found that serum anti-TOPO48 autoantibody levels in ESCC patients were significantly higher than that in healthy controls and benign tumor patients (P=0.001). The percentage of sera with a positive level of anti-TOPO48 autoantibody in early stages was significantly higher than that in advanced stages of the cancer patients when the maximum level of healthy control sera was taken as a cut-off value (P=0.001). The area under ROC curve was 0.863 (95% CI=0.797-0.928) for healthy controls vs. early stage ESCC. In addition, patients with positive anti-TOPO48 autoantibody had significantly higher survival rate and longer survival time than that with negative anti-TOPO48 autoantibody in cancer patients (P=0.038, 0.025 and 0.047 for all stages, early stage and advanced stage, respectively). CONCLUSIONS Our results suggest that anti-TOPO48 autoantibody may be a potentially useful biomarker for early diagnosis and prognosis of ESCC.
Collapse
Affiliation(s)
- Jian-Bo Zhang
- Core Laboratory, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospitals, 610072 Chengdu, Sichuan, PR, China
| | - Mei Cao
- Core Laboratory, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospitals, 610072 Chengdu, Sichuan, PR, China
| | - Jie Chen
- Core Laboratory, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospitals, 610072 Chengdu, Sichuan, PR, China
| | - Shang-Rong Ye
- Chengdu Cancer Bioengineering Research Institute, 610075 Chengdu, Sichuan PR, China
| | - Ke Xie
- Department of Oncology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospitals, 610072 Chengdu, Sichuan, PR, China
| | - Xu He
- Core Laboratory, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospitals, 610072 Chengdu, Sichuan, PR, China
| | - Xiao-Li Ma
- Chengdu Cancer Bioengineering Research Institute, 610075 Chengdu, Sichuan PR, China
| | - Jia Zhang
- Chengdu Cancer Bioengineering Research Institute, 610075 Chengdu, Sichuan PR, China
| | - Shang-Mian Yie
- Core Laboratory, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospitals, 610072 Chengdu, Sichuan, PR, China; Chengdu Cancer Bioengineering Research Institute, 610075 Chengdu, Sichuan PR, China.
| |
Collapse
|
162
|
Stadler S, Singh VK, Knörr F, Damm-Welk C, Woessmann W. Immune Response against ALK in Children with ALK-Positive Anaplastic Large Cell Lymphoma. Cancers (Basel) 2018; 10:cancers10040114. [PMID: 29642597 PMCID: PMC5923369 DOI: 10.3390/cancers10040114] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/05/2018] [Accepted: 04/07/2018] [Indexed: 12/26/2022] Open
Abstract
Patients with anaplastic lymphoma kinase (ALK)-positive anaplastic large cell lymphoma (ALCL) mount a humoral and cellular immune response against ALK. More than 90% of children and adolescents with ALK-positive ALCL have detectable anti-ALK antibodies in serum or plasma, and the antibody titer inversely correlates with the risk of relapse. ALK-specific CD8 and CD4 T cell responses have been described in patients with ALK-positive ALCL. Vaccination with ALK DNA led to protection against lymphoma growth in a murine model. Collectively, these data suggest that the ALK-specific immune response is involved in the control of the disease. The characteristics of the humoral and cellular immune response against ALK as well as tumor immune escape mechanisms have been increasingly investigated. However, tumor and host factors contributing to the individual immune response against ALK are still largely unknown. Depending on the individual strength of the immune response and its determinants, individualized immunological approaches might be appropriate for the consolidation of ALCL patients. Strategies such as ALK vaccination could be effective for those with a pre-existing anti-tumor immunity, while an allogeneic blood stem cell transplantation or check-point inhibition could be effective for others.
Collapse
Affiliation(s)
- Serena Stadler
- Department of Pediatric Hematology and Oncology, Justus-Liebig University, D-35392 Giessen, Germany.
| | - Vijay Kumar Singh
- Department of Pediatric Hematology and Oncology, Justus-Liebig University, D-35392 Giessen, Germany.
| | - Fabian Knörr
- Department of Pediatric Hematology and Oncology, Justus-Liebig University, D-35392 Giessen, Germany.
| | - Christine Damm-Welk
- Department of Pediatric Hematology and Oncology, Justus-Liebig University, D-35392 Giessen, Germany.
| | - Wilhelm Woessmann
- Department of Pediatric Hematology and Oncology, Justus-Liebig University, D-35392 Giessen, Germany.
| |
Collapse
|
163
|
Fortner RT, Schock H, Le Cornet C, Hüsing A, Vitonis AF, Johnson TS, Fichorova RN, Fashemi T, Yamamoto HS, Tjønneland A, Hansen L, Overvad K, Boutron-Ruault MC, Kvaskoff M, Severi G, Boeing H, Trichopoulou A, Papatesta EM, Vecchia CL, Palli D, Sieri S, Tumino R, Sacerdote C, Mattiello A, Onland-Moret NC, Peeters PH, Bueno-de-Mesquita H, Weiderpass E, Quirós JR, Duell EJ, Sánchez MJ, Navarro C, Ardanaz E, Larrañaga N, Nodin B, Jirström K, Idahl A, Lundin E, Khaw KT, Travis RC, Gunter M, Johansson M, Dossus L, Merritt MA, Riboli E, Terry KL, Cramer DW, Kaaks R. Ovarian cancer early detection by circulating CA125 in the context of anti-CA125 autoantibody levels: Results from the EPIC cohort. Int J Cancer 2018; 142:1355-1360. [PMID: 29159934 PMCID: PMC5805613 DOI: 10.1002/ijc.31164] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 09/25/2017] [Accepted: 10/26/2017] [Indexed: 01/12/2023]
Abstract
CA125 is the best ovarian cancer early detection marker to date; however, sensitivity is limited and complementary markers are required to improve discrimination between ovarian cancer cases and non-cases. Anti-CA125 autoantibodies are observed in circulation. Our objective was to evaluate whether these antibodies (1) can serve as early detection markers, providing evidence of an immune response to a developing tumor, and (2) modify the discriminatory capacity of CA125 by either masking CA125 levels (resulting in lower discrimination) or acting synergistically to improve discrimination between cases and non-cases. We investigated these objectives using a nested case-control study within the European Prospective Investigation into Cancer and Nutrition cohort (EPIC) including 250 cases diagnosed within 4 years of blood collection and up to four matched controls. Circulating CA125 antigen and antibody levels were quantified using an electrochemiluminescence assay. Adjusted areas under the curve (aAUCs) by 2-year lag-time intervals were calculated using conditional logistic regression calibrated toward the absolute risk estimates from a pre-existing epidemiological risk model as an offset-variable. Anti-CA125 levels alone did not discriminate cases from controls. For cases diagnosed <2 years after blood collection, discrimination by CA125 antigen was suggestively higher with higher anti-CA125 levels (aAUC, highest antibody tertile: 0.84 [0.76-0.92]; lowest tertile: 0.76 [0.67-0.86]; phet = 0.06). We provide the first evidence of potentially synergistic discrimination effects of CA125 and anti-CA125 antibodies in ovarian early detection. If these findings are replicated, evaluating CA125 in the context of its antibody may improve ovarian cancer early detection.
Collapse
Affiliation(s)
- Renée T. Fortner
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Helena Schock
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Charlotte Le Cornet
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anika Hüsing
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Allison F. Vitonis
- Ob/Gyn Epidemiology Center, Brigham and Women's Hospital, Boston, Massachusetts
| | - Theron S. Johnson
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Raina N. Fichorova
- Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Boston, Massachusetts
- Laboratory of Genital Tract Biology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Titilayo Fashemi
- Laboratory of Genital Tract Biology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Hidemi S. Yamamoto
- Laboratory of Genital Tract Biology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Anne Tjønneland
- Unit of Diet, Genes and Environment, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Louise Hansen
- Unit of Diet, Genes and Environment, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Kim Overvad
- Department of Public Health, Section for Epidemiology, Aarhus University, Aarhus, Denmark
| | - Marie-Christine Boutron-Ruault
- INSERM, Centre for Research in Epidemiology and Population Health (CESP), U1018, Nutrition, Hormones and Women’s Health team, F-94805, Villejuif, France
- Université Paris Sud, UMRS 1018, F-94805, Villejuif, France
- Gustave Roussy, F-94805, Villejuif, France
| | - Marina Kvaskoff
- INSERM, Centre for Research in Epidemiology and Population Health (CESP), U1018, Nutrition, Hormones and Women’s Health team, F-94805, Villejuif, France
- Université Paris Sud, UMRS 1018, F-94805, Villejuif, France
- Gustave Roussy, F-94805, Villejuif, France
| | - Gianluca Severi
- INSERM, Centre for Research in Epidemiology and Population Health (CESP), U1018, Nutrition, Hormones and Women’s Health team, F-94805, Villejuif, France
- Université Paris Sud, UMRS 1018, F-94805, Villejuif, France
- Gustave Roussy, F-94805, Villejuif, France
- Human Genetics Foundation (HuGeF), Torino, Italy
| | - Heiner Boeing
- Department of Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Antonia Trichopoulou
- Hellenic Health Foundation, Athens, Greece
- WHO Collaborating Center for Nutrition and Health, Unit of Nutritional Epidemiology and Nutrition in Public Health, Dept. of Hygiene, Epidemiology and Medical Statistics, School of Medicine, National and Kapodistrian University of Athens, Greece
| | | | - Carlo La Vecchia
- Hellenic Health Foundation, Athens, Greece
- Dept. of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Domenico Palli
- Cancer Risk Factors and Life-Style Epidemiology Unit, Cancer Research and Prevention Institute – ISPO, Florence, Italy
| | - Sabina Sieri
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Rosario Tumino
- Cancer Registry and Histopathology Unit, "Civic - M.P-Arezzo" Hospital, ASP Ragusa, Italy
| | - Carlotta Sacerdote
- Unit of Cancer Epidemiology, Città della Salute e della Scienza University-Hospital and Center for Cancer Prevention (CPO), Turin, Italy
| | - Amalia Mattiello
- Dipartimeno di Medicina Clinica e Chirurgia, Federico II University, Naples, Italy
| | - N. Charlotte Onland-Moret
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Netherlands
| | - Petra H. Peeters
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Netherlands
- MRC-PHE Centre for Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College, London, UK
| | - H.B(as). Bueno-de-Mesquita
- Department for Determinants of Chronic Diseases (DCD), National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
- Department of Epidemiology and Biostatistics, The School of Public Health, Imperial College London, London, United Kingdom
| | - Elisabete Weiderpass
- Department of Community Medicine, Faculty of Health Sciences, University of Tromsø, The Arctic University of Norway, Tromsø, Norway
- Department of Research, Cancer Registry of Norway, Institute of Population-Based Cancer Research, Oslo, Norway
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Genetic Epidemiology Group, Folkhälsan Research Center, Helsinki, Finland
| | | | - Eric J Duell
- Cancer Epidemiology Research Program, Bellvitge Biomedical Research Institute (IDIBELL), Catalan Institute of Oncology (ICO), Barcelona, Spain
| | - Maria-Jose Sánchez
- Escuela Andaluza de Salud Pública. Instituto de Investigación Biosanitaria ibs.GRANADA. Hospitales Universitarios de Granada/Universidad de Granada, Granada, Spain
- CIBER de Epidemiología y Salud Pública (CIBERESP), Spain
| | - Carmen Navarro
- CIBER de Epidemiología y Salud Pública (CIBERESP), Spain
- Department of Epidemiology, Murcia Regional Health Council, IMIB-Arrixaca, Murcia, Spain
| | - Eva Ardanaz
- CIBER de Epidemiología y Salud Pública (CIBERESP), Spain
- Navarra Public Health Institute, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Nerea Larrañaga
- CIBER de Epidemiología y Salud Pública (CIBERESP), Spain
- Public Health Division of Gipuzkoa, Regional Government of the Basque Country, Spain
| | - Björn Nodin
- Department of Clinical Sciences Lund, Division of Oncology and Pathology, Lund University, Lund, Sweden
| | - Karin Jirström
- Department of Clinical Sciences Lund, Division of Oncology and Pathology, Lund University, Lund, Sweden
| | - Annika Idahl
- Department of Clinical Sciences, Obstetrics and Gynecology, Umeå University, Umeå, Sweden
- Department of Public Health and Clinical Medicine, Nutritional Research, Umeå University, Umeå, Sweden
| | - Eva Lundin
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Kay-Tee Khaw
- Cancer Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Ruth C. Travis
- Cancer Epidemiology Unit, Nuffield Department of Population Health University of Oxford, Oxford UK
| | - Marc Gunter
- International Agency for Research on Cancer, Lyon, France
| | | | - Laure Dossus
- International Agency for Research on Cancer, Lyon, France
| | - Melissa A. Merritt
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, United Kingdom
| | - Elio Riboli
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, United Kingdom
| | - Kathryn L. Terry
- Ob/Gyn Epidemiology Center, Brigham and Women's Hospital, Boston, Massachusetts
- Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Boston, Massachusetts
| | - Daniel W. Cramer
- Ob/Gyn Epidemiology Center, Brigham and Women's Hospital, Boston, Massachusetts
- Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Boston, Massachusetts
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
164
|
Wang YH, Shen XD. Human immunodeficiency virus infection and mortality risk among lung cancer patients: A systematic review and meta-analysis. Medicine (Baltimore) 2018; 97:e0361. [PMID: 29642182 PMCID: PMC5908612 DOI: 10.1097/md.0000000000010361] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Previous studies have suggested that patients with human immunodeficiency virus (HIV) infection are at higher risk of lung cancer, but the impact of HIV infection on the risk of mortality among lung cancer patients is still unclear. We conducted a systematic review and meta-analysis to clarify the association between HIV infection and mortality risk among lung cancer patients. METHODS PubMed and Embase databases were searched to identify studies assessing the association between HIV infection and mortality risk among lung cancer patients. Only studies reporting adjusted relative risk (RR) of mortality among lung cancer patients with HIV infection were included. Meta-analysis of random-effect model was utilized to calculate the pooled RR with 95% confidence interval (CI). RESULTS Twelve cohort studies were finally included. Compared with lung cancer patients without HIV infection, the pooled RR of mortality among lung cancer patients with HIV infection was 1.48 (95% CI, 1.22-1.78, P < .001; I = 88.6%). After excluding 2 studies with low quality, HIV infection was still significantly associated with an elevated risk of mortality among lung cancer patients (RR = 1.51, 95% CI, 1.25-1.82, P < .001; I = 89.8%). Sensitivity analysis showed that the statistical significance of the pooled RR was not changed by excluding any one study. CONCLUSION The outcomes from the meta-analysis provide strong evidence for the elevated risk of mortality among lung cancer patients with HIV infection, and HIV infection is an important prognostic factor in lung cancer patients.
Collapse
Affiliation(s)
- Yue-Hua Wang
- Department of Respiration, Jinhua People's Hospital, Jinhua
| | - Xiang-Di Shen
- Faculty of Basic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
165
|
Da Gama Duarte J, Parakh S, Andrews MC, Woods K, Pasam A, Tutuka C, Ostrouska S, Blackburn JM, Behren A, Cebon J. Autoantibodies May Predict Immune-Related Toxicity: Results from a Phase I Study of Intralesional Bacillus Calmette-Guérin followed by Ipilimumab in Patients with Advanced Metastatic Melanoma. Front Immunol 2018; 9:411. [PMID: 29552014 PMCID: PMC5840202 DOI: 10.3389/fimmu.2018.00411] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 02/14/2018] [Indexed: 11/13/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of advanced melanoma. The first ICI to demonstrate clinical benefit, ipilimumab, targets cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4); however, the long-term overall survival is just 22%. More than 40 years ago intralesional (IL) bacillus Calmette-Guérin (BCG), a living attenuated strain of Mycobacterium bovis, was found to induce tumor regression by stimulating cell-mediated immunity following a localized and self-limiting infection. We evaluated these two immune stimulants in combination with melanoma with the aim of developing a more effective immunotherapy and to assess toxicity. In this phase I study, patients with histologically confirmed stage III/IV metastatic melanoma received IL BCG injection followed by up to four cycles of intravenous ipilimumab (anti-CTLA-4) (ClinicalTrials.gov number NCT01838200). The trial was discontinued following treatment of the first five patients as the two patients receiving the escalation dose of BCG developed high-grade immune-related adverse events (irAEs) typical of ipilimumab monotherapy. These irAEs were characterized in both patients by profound increases in the repertoire of autoantibodies directed against both self- and cancer antigens. Interestingly, the induced autoantibodies were detected at time points that preceded the development of symptomatic toxicity. There was no overlap in the antigen specificity between patients and no evidence of clinical responses. Efforts to increase response rates through the use of novel immunotherapeutic combinations may be associated with higher rates of irAEs, thus the imperative to identify biomarkers of toxicity remains strong. While the small patient numbers in this trial do not allow for any conclusive evidence of predictive biomarkers, the observed changes warrant further examination of autoantibody repertoires in larger patient cohorts at risk of developing irAEs during their course of treatment. In summary, dose escalation of IL BCG followed by ipilimumab therapy was not well tolerated in advanced melanoma patients and showed no evidence of clinical benefit. Measuring autoantibody responses may provide early means for identifying patients at risk from developing severe irAEs during cancer immunotherapy.
Collapse
Affiliation(s)
- Jessica Da Gama Duarte
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia.,School of Cancer Medicine, La Trobe University, Heidelberg, VIC, Australia.,Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Melbourne, VIC, Australia
| | - Sagun Parakh
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia.,School of Cancer Medicine, La Trobe University, Heidelberg, VIC, Australia.,Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Melbourne, VIC, Australia
| | - Miles C Andrews
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia.,School of Cancer Medicine, La Trobe University, Heidelberg, VIC, Australia.,Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Melbourne, VIC, Australia.,MD Anderson Cancer Center, University of Texas, Houston, TX, United States
| | - Katherine Woods
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia.,School of Cancer Medicine, La Trobe University, Heidelberg, VIC, Australia.,Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Melbourne, VIC, Australia
| | - Anupama Pasam
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia.,School of Cancer Medicine, La Trobe University, Heidelberg, VIC, Australia.,Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Melbourne, VIC, Australia
| | - Candani Tutuka
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia.,School of Cancer Medicine, La Trobe University, Heidelberg, VIC, Australia.,Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Melbourne, VIC, Australia
| | - Simone Ostrouska
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia.,School of Cancer Medicine, La Trobe University, Heidelberg, VIC, Australia
| | - Jonathan M Blackburn
- Department of Integrative Biomedical Sciences and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,Sengenics Corporation, Singapore, Singapore
| | - Andreas Behren
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia.,School of Cancer Medicine, La Trobe University, Heidelberg, VIC, Australia.,Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Melbourne, VIC, Australia
| | - Jonathan Cebon
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia.,School of Cancer Medicine, La Trobe University, Heidelberg, VIC, Australia.,Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Melbourne, VIC, Australia
| |
Collapse
|
166
|
Villaseñor R, Kuennecke B, Ozmen L, Ammann M, Kugler C, Grüninger F, Loetscher H, Freskgård PO, Collin L. Region-specific permeability of the blood-brain barrier upon pericyte loss. J Cereb Blood Flow Metab 2017; 37:3683-3694. [PMID: 28273726 PMCID: PMC5718326 DOI: 10.1177/0271678x17697340] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The blood-brain barrier (BBB) regulates differing needs of the various brain regions by controlling transport of blood-borne components from the neurovascular circulation into the brain parenchyma. The mechanisms underlying region-specific transport across the BBB are not completely understood. Previous work showed that pericytes are key regulators of BBB function. Here we investigated whether pericytes influence BBB permeability in a region-specific manner by analysing the regional permeability of the BBB in the pdgf-b ret/ret mouse model of pericyte depletion. We show that BBB permeability is heterogeneous in pdgf-b ret/ret mice, being significantly higher in the cortex, striatum and hippocampus compared to the interbrain and midbrain. However, we show that this regional heterogeneity in BBB permeability is not explained by local differences in pericyte coverage. Region-specific differences in permeability were not associated with disruption of tight junctions but may result from changes in transcytosis across brain endothelial cells. Our data show that certain brain regions are able to maintain low BBB permeability despite substantial pericyte loss and suggest that additional, locally-acting mechanisms may contribute to control of transport.
Collapse
Affiliation(s)
- Roberto Villaseñor
- 1 Roche Pharma Research and Early Development (pRED), Neurodegeneration and Regeneration, Roche Innovation Center Basel, Switzerland
| | - Basil Kuennecke
- 2 Roche Pharma Research and Early Development (pRED), Translational Medicine Neuroscience & Biomarkers, Roche Innovation Center Basel, Switzerland
| | - Laurence Ozmen
- 1 Roche Pharma Research and Early Development (pRED), Neurodegeneration and Regeneration, Roche Innovation Center Basel, Switzerland
| | - Michelle Ammann
- 1 Roche Pharma Research and Early Development (pRED), Neurodegeneration and Regeneration, Roche Innovation Center Basel, Switzerland
| | - Christof Kugler
- 1 Roche Pharma Research and Early Development (pRED), Neurodegeneration and Regeneration, Roche Innovation Center Basel, Switzerland
| | - Fiona Grüninger
- 1 Roche Pharma Research and Early Development (pRED), Neurodegeneration and Regeneration, Roche Innovation Center Basel, Switzerland
| | - Hansruedi Loetscher
- 1 Roche Pharma Research and Early Development (pRED), Neurodegeneration and Regeneration, Roche Innovation Center Basel, Switzerland
| | - Per-Ola Freskgård
- 1 Roche Pharma Research and Early Development (pRED), Neurodegeneration and Regeneration, Roche Innovation Center Basel, Switzerland
| | - Ludovic Collin
- 1 Roche Pharma Research and Early Development (pRED), Neurodegeneration and Regeneration, Roche Innovation Center Basel, Switzerland
| |
Collapse
|
167
|
Wilson AL, Moffitt LR, Duffield N, Rainczuk A, Jobling TW, Plebanski M, Stephens AN. Autoantibodies against HSF1 and CCDC155 as Biomarkers of Early-Stage, High-Grade Serous Ovarian Cancer. Cancer Epidemiol Biomarkers Prev 2017; 27:183-192. [PMID: 29141850 DOI: 10.1158/1055-9965.epi-17-0752] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 10/23/2017] [Accepted: 11/09/2017] [Indexed: 11/16/2022] Open
Abstract
Background: Tumor-directed circulating autoantibodies (AAb) are a well-established feature of many solid tumor types, and are often observed prior to clinical disease manifestation. As such, they may provide a good indicator of early disease development. We have conducted a pilot study to identify novel AAbs as markers of early-stage HGSOCs.Methods: A rare cohort of patients with early (FIGO stage Ia-c) HGSOCs for IgG, IgA, and IgM-mediated AAb reactivity using high-content protein arrays (containing 9,184 individual proteins). AAb reactivity against selected antigens was validated by ELISA in a second, independent cohort of individual patients.Results: A total of 184 antigens were differentially detected in early-stage HGSOC patients compared with all other patient groups assessed. Among the six most highly detected "early-stage" antigens, anti-IgA AAbs against HSF1 and anti-IgG AAbs CCDC155 (KASH5; nesprin 5) were significantly elevated in patients with early-stage malignancy. Receiver operating characteristic (ROC) analysis suggested that AAbs against HSF1 provided better detection of early-stage malignancy than CA125 alone. Combined measurement of anti-HSF1, anti-CCDC155, and CA125 also improved efficacy at higher sensitivity.Conclusions: The combined measurement of anti-HSF1, anti-CCDC155, and CA125 may be useful for early-stage HGSOC detection.Impact: This is the first study to specifically identify AAbs associated with early-stage HGSOC. The presence and high frequency of specific AAbs in early-stage cancer patients warrants a larger scale examination to define their value for early disease detection at primary diagnosis and/or recurrence. Cancer Epidemiol Biomarkers Prev; 27(2); 183-92. ©2017 AACR.
Collapse
Affiliation(s)
- Amy L Wilson
- Department of Molecular and Translational Sciences, Monash University, Victoria, Australia.,Centre for Cancer Research, Hudson Institute of Medical Research, Victoria, Australia
| | - Laura R Moffitt
- Department of Molecular and Translational Sciences, Monash University, Victoria, Australia.,Centre for Cancer Research, Hudson Institute of Medical Research, Victoria, Australia
| | - Nadine Duffield
- Department of Molecular and Translational Sciences, Monash University, Victoria, Australia.,Centre for Cancer Research, Hudson Institute of Medical Research, Victoria, Australia
| | - Adam Rainczuk
- Department of Molecular and Translational Sciences, Monash University, Victoria, Australia.,Centre for Cancer Research, Hudson Institute of Medical Research, Victoria, Australia
| | - Tom W Jobling
- Obstetrics and Gynaecology, Monash Medical Centre, Clayton, Victoria, Australia.,Epworth Research Institute, Epworth HealthCare, Richmond, Victoria, Australia
| | - Magdalena Plebanski
- Department of Immunology and Pathology, Monash University, Melbourne, Australia.,School of Health and Biomedical Sciences, RMIT, Bundoora, Victoria, Australia
| | - Andrew N Stephens
- Department of Molecular and Translational Sciences, Monash University, Victoria, Australia. .,Centre for Cancer Research, Hudson Institute of Medical Research, Victoria, Australia.,Epworth Research Institute, Epworth HealthCare, Richmond, Victoria, Australia
| |
Collapse
|
168
|
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
|
169
|
Pazhouhandeh M, Samiee F, Boniadi T, Khedmat AF, Vahedi E, Mirdamadi M, Sigari N, Siadat SD, Vaziri F, Fateh A, Ajorloo F, Tafsiri E, Ghanei M, Mahboudi F, Rahimi Jamnani F. Comparative Network Analysis of Patients with Non-Small Cell Lung Cancer and Smokers for Representing Potential Therapeutic Targets. Sci Rep 2017; 7:13812. [PMID: 29062084 PMCID: PMC5653836 DOI: 10.1038/s41598-017-14195-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 10/06/2017] [Indexed: 02/08/2023] Open
Abstract
Cigarette smoking is the leading cause of lung cancer worldwide. In this study, we evaluated the serum autoantibody (AAb) repertoires of non-small cell lung cancer (NSCLC) patients and smokers (SM), leading to the identification of overactivated pathways and hubs involved in the pathogenesis of NSCLC. Surface- and solution-phase biopanning were performed on immunoglobulin G purified from the sera of NSCLC and SM groups. In total, 20 NSCLC- and 12 SM-specific peptides were detected, which were used to generate NSCLC and SM protein datasets. NSCLC- and SM-related proteins were visualized using STRING and Gephi, and their modules were analyzed using Enrichr. By integrating the overrepresented pathways such as pathways in cancer, epithelial growth factor receptor, c-Met, interleukin-4 (IL-4) and IL-6 signaling pathways, along with a set of proteins (e.g. phospholipase D (PLD), IL-4 receptor, IL-17 receptor, laminins, collagens, and mucins) into the PLD pathway and inflammatory cytokines network as the most critical events in both groups, two super networks were made to elucidate new aspects of NSCLC pathogenesis and to determine the influence of cigarette smoking on tumour formation. Taken together, assessment of the AAb repertoires using a systems biology approach can delineate the hidden events involved in various disorders.
Collapse
Affiliation(s)
| | - Fatemeh Samiee
- Department of Microbial Biotechnology, Islamic Azad University, Pharmaceutical Sciences Branch, Tehran, Iran
| | - Tahereh Boniadi
- Department of Microbial Biotechnology, Islamic Azad University, Pharmaceutical Sciences Branch, Tehran, Iran
| | - Abbas Fadaei Khedmat
- Department of Pulmonology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ensieh Vahedi
- Chemical Injuries Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mahsa Mirdamadi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Naseh Sigari
- Internal Medicine Department, Medical Faculty, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Seyed Davar Siadat
- Human Antibody Lab, Innovation Center, Pasteur Institute of Iran, Tehran, Iran
- Microbiology Research Center, Department of Mycobacteriology and Pulmonary Research Pasteur Institute of Iran, Tehran, Iran
| | - Farzam Vaziri
- Human Antibody Lab, Innovation Center, Pasteur Institute of Iran, Tehran, Iran
- Microbiology Research Center, Department of Mycobacteriology and Pulmonary Research Pasteur Institute of Iran, Tehran, Iran
| | - Abolfazl Fateh
- Human Antibody Lab, Innovation Center, Pasteur Institute of Iran, Tehran, Iran
- Microbiology Research Center, Department of Mycobacteriology and Pulmonary Research Pasteur Institute of Iran, Tehran, Iran
| | - Faezeh Ajorloo
- Department of Biology, Faculty of Science, Islamic Azad University, East Tehran Branch, Tehran, Iran
| | - Elham Tafsiri
- Molecular Medicine Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Mostafa Ghanei
- Chemical Injuries Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | | | - Fatemeh Rahimi Jamnani
- Human Antibody Lab, Innovation Center, Pasteur Institute of Iran, Tehran, Iran.
- Microbiology Research Center, Department of Mycobacteriology and Pulmonary Research Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
170
|
The roles and applications of autoantibodies in progression, diagnosis, treatment and prognosis of human malignant tumours. Autoimmun Rev 2017; 16:1270-1281. [PMID: 29042252 DOI: 10.1016/j.autrev.2017.10.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 09/06/2017] [Indexed: 02/07/2023]
Abstract
The existence of autoantibodies towards an individual's own proteins or nucleic acids has been established for more than 100years, and for a long period, these autoantibodies have been believed to be closely associated with autoimmune diseases. However, in recent years, researchers have become more interested in the role and application of autoantibodies in progression, diagnosis, treatment and prognosis of human malignant tumours. Over the past few decades, numerous epidemiological studies have shown that the risk of certain cancers is significantly altered (increased or decreased) in patients with autoimmune diseases, which suggests that autoantibodies may play either promoting or suppressing roles in cancer progression. The idea that autoantibodies are directly involved in tumour progression gains special support by the findings that some antibodies secreted by a variety of cancer cells can promote their proliferation and metastasis. Because the cancer cells generate cell antigenic changes (neoantigens), which trigger the immune system to produce autoantibodies, serum autoantibodies against tumour-associated antigens have been established as a novel type of cancer biomarkers and have been extensively studied in different types of cancer. The autoantibodies as biomarkers in cancer diagnosis are not only more sensitive and specific than antigens, but also could appear before clinical evidences of the tumours, thus disclosing them. The observations that cancer risk is lower in patients with some autoimmune diseases suggest that certain autoantibodies may be protective from certain cancers. Moreover, the presence of autoantibodies in healthy individuals implies that it could be safe to employ autoantibodies to treat cancer. Of note, an autoantibodies derived from lupus murine model received much attention due to their selective cytotoxicity for malignant tumour cell without harming normal ones. These studies showed the therapeutic value of autoantibodies in cancer. In this review, we revisited the pathological or protective role of autoantibodies in cancer progression, summarize the application of autoantibodies in cancer diagnosis and prognosis, and discuss the value of autoantibodies in cancer therapy. The studies established to date suggest that autoantibodies not only regulate cancer progression but also promise to be valuable instruments in oncological diagnosis and therapy.
Collapse
|
171
|
Mitoma H, Manto M, Hampe CS. Immune-mediated cerebellar ataxias: from bench to bedside. CEREBELLUM & ATAXIAS 2017; 4:16. [PMID: 28944066 PMCID: PMC5609024 DOI: 10.1186/s40673-017-0073-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 09/07/2017] [Indexed: 02/08/2023]
Abstract
The cerebellum is a vulnerable target of autoimmunity in the CNS. The category of immune-mediated cerebellar ataxias (IMCAs) was recently established, and includes in particular paraneoplastic cerebellar degenerations (PCDs), gluten ataxia (GA) and anti-GAD65 antibody (Ab) associated-CA, all characterized by the presence of autoantibodies. The significance of onconeuronal autoantibodies remains uncertain in some cases. The pathogenic role of anti-GAD65Ab has been established both in vitro and in vivo, but a consensus has not been reached yet. Recent studies of anti-GAD65 Ab-associated CA have clarified that (1) autoantibodies are generally polyclonal and elicit pathogenic effects related to epitope specificity, and (2) the clinical course can be divided into two phases: a phase of functional disorder followed by cell death. These features provide the rationale for prompt diagnosis and therapeutic strategies. The concept “Time is brain” has been completely underestimated in the field of immune ataxias. We now put forward the concept “Time is cerebellum” to underline the importance of very early therapeutic strategies in order to prevent or stop the loss of neurons and synapses. The diagnosis of IMCAs should depend not only on Ab testing, but rather on a rapid and comprehensive assessment of the clinical/immune profile. Treatment should be applied during the period of preserved cerebellar reserve, and should encompass early removal of the conditions (such as remote primary tumors) or diseases that trigger the autoimmunity, followed by the combinations of various immunotherapies.
Collapse
Affiliation(s)
- Hiroshi Mitoma
- Tokyo Medical University, Medical Education Promotion Center, 6-7-1 Nishi-Shinjyuku, Shinjyuku-ku, Tokyo, 160-0023 Japan
| | - Mario Manto
- Unité d'Etude du Mouvement (UEM), FNRS, ULB-Erasme, 1070 Bruxelles, Belgium.,Service des Neurosciences, University of Mons, 7000 Mons, Belgium
| | | |
Collapse
|
172
|
A systems medicine approach for finding target proteins affecting treatment outcomes in patients with non-Hodgkin lymphoma. PLoS One 2017; 12:e0183969. [PMID: 28892521 PMCID: PMC5593188 DOI: 10.1371/journal.pone.0183969] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Accepted: 08/15/2017] [Indexed: 02/07/2023] Open
Abstract
Autoantibody profiling with a systems medicine approach can help identify critical dysregulated signaling pathways (SPs) in cancers. In this way, immunoglobulins G (IgG) purified from the serum samples of 92 healthy controls, 10 pre-treated (PR) non-Hodgkin lymphoma (NHL) patients, and 20 NHL patients who underwent chemotherapy (PS) were screened with a phage-displayed random peptide library. Protein-protein interaction networks of the PR and PS groups were analyzed and visualized by Gephi. The results indicated AXIN2, SENP2, TOP2A, FZD6, NLK, HDAC2, HDAC1, and EHMT2, in addition to CAMK2A, PLCG1, PLCG2, GRM5, GRIN2B, GRIN2D, CACNA2D3, and SPTAN1 as hubs in 11 and 7 modules of PR and PS networks, respectively. PR- and PS-specific hubs were evaluated in the Kyoto Encyclopedia of Genes and Genomes (KEGG) and Reactome databases. The PR-specific hubs were involved in Wnt SP, signaling by Notch1 in cancer, telomere maintenance, and transcriptional misregulation. In contrast, glutamate receptor SP, Fc receptor-related pathways, growth factors-related SPs, and Wnt SP were statistically significant enriched pathways, based on the pathway analysis of PS hubs. The results revealed that the most PR-specific proteins were associated with events involved in tumor development, while chemotherapy in the PS group was associated with side effects of drugs and/or cancer recurrence. As the findings demonstrated, PR- and PS-specific proteins in this study can be promising therapeutic targets in future studies.
Collapse
|
173
|
Ishigaki H, Maeda T, Inoue H, Akagi T, Sasamura T, Ishida H, Inubushi T, Okahara J, Shiina T, Nakayama M, Itoh Y, Ogasawara K. Transplantation of iPS-Derived Tumor Cells with a Homozygous MHC Haplotype Induces GRP94 Antibody Production in MHC-Matched Macaques. Cancer Res 2017; 77:6001-6010. [PMID: 28882998 DOI: 10.1158/0008-5472.can-17-0775] [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: 03/21/2017] [Revised: 07/24/2017] [Accepted: 08/31/2017] [Indexed: 11/16/2022]
Abstract
Immune surveillance is a critical component of the antitumor response in vivo, yet the specific components of the immune system involved in this regulatory response remain unclear. In this study, we demonstrate that autoantibodies can mitigate tumor growth in vitro and in vivo We generated two cancer cell lines, embryonal carcinoma and glioblastoma cell lines, from monkey-induced pluripotent stem cells (iPSC) carrying a homozygous haplotype of major histocompatibility complex (MHC, Mafa in Macaca fascicularis). To establish a monkey cancer model, we transplanted these cells into monkeys carrying the matched Mafa haplotype in one of the chromosomes. Neither Mafa-homozygous cancer cell line grew in monkeys carrying the matched Mafa haplotype heterozygously. We detected in the plasma of these monkeys an IgG autoantibody against GRP94, a heat shock protein. Injection of the plasma prevented growth of the tumor cells in immunodeficient mice, whereas plasma IgG depleted of GRP94 IgG exhibited reduced killing activity against cancer cells in vitro These results indicate that humoral immunity, including autoantibodies against GRP94, plays a role in cancer immune surveillance. Cancer Res; 77(21); 6001-10. ©2017 AACR.
Collapse
Affiliation(s)
- Hirohito Ishigaki
- Division of Pathology and Disease Regulation, Department of Pathology, Shiga University of Medical Science, Otsu, Shiga, Japan.
| | - Toshinaga Maeda
- Central Research Laboratory, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Hirokazu Inoue
- Division of Microbiology and Infectious Diseases, Department of Pathology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | | | - Takako Sasamura
- Division of Pathology and Disease Regulation, Department of Pathology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Hideaki Ishida
- Division of Pathology and Disease Regulation, Department of Pathology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Toshiro Inubushi
- Biomedical MR Science Center, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Junko Okahara
- Central Institute for Experimental Animals, Kawasaki, Kanagawa, Japan
| | - Takashi Shiina
- Department of Molecular Life Science, Division of Basic Medical Science and Molecular Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Misako Nakayama
- Division of Pathology and Disease Regulation, Department of Pathology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Yasushi Itoh
- Division of Pathology and Disease Regulation, Department of Pathology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Kazumasa Ogasawara
- Division of Pathology and Disease Regulation, Department of Pathology, Shiga University of Medical Science, Otsu, Shiga, Japan
| |
Collapse
|
174
|
Blandin Knight S, Crosbie PA, Balata H, Chudziak J, Hussell T, Dive C. Progress and prospects of early detection in lung cancer. Open Biol 2017; 7:170070. [PMID: 28878044 PMCID: PMC5627048 DOI: 10.1098/rsob.170070] [Citation(s) in RCA: 513] [Impact Index Per Article: 64.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 07/27/2017] [Indexed: 12/14/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related death in the world. It is broadly divided into small cell (SCLC, approx. 15% cases) and non-small cell lung cancer (NSCLC, approx. 85% cases). The main histological subtypes of NSCLC are adenocarcinoma and squamous cell carcinoma, with the presence of specific DNA mutations allowing further molecular stratification. If identified at an early stage, surgical resection of NSCLC offers a favourable prognosis, with published case series reporting 5-year survival rates of up to 70% for small, localized tumours (stage I). However, most patients (approx. 75%) have advanced disease at the time of diagnosis (stage III/IV) and despite significant developments in the oncological management of late stage lung cancer over recent years, survival remains poor. In 2014, the UK Office for National Statistics reported that patients diagnosed with distant metastatic disease (stage IV) had a 1-year survival rate of just 15-19% compared with 81-85% for stage I.
Collapse
MESH Headings
- Adenocarcinoma/diagnostic imaging
- Adenocarcinoma/genetics
- Adenocarcinoma/mortality
- Adenocarcinoma/surgery
- Adenocarcinoma of Lung
- Biomarkers, Tumor/blood
- Biomarkers, Tumor/genetics
- Bronchoscopy/methods
- Carcinoma, Non-Small-Cell Lung/diagnostic imaging
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/mortality
- Carcinoma, Non-Small-Cell Lung/surgery
- Carcinoma, Squamous Cell/diagnostic imaging
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/mortality
- Carcinoma, Squamous Cell/surgery
- Circulating Tumor DNA/blood
- Circulating Tumor DNA/genetics
- Early Detection of Cancer/methods
- Humans
- Liquid Biopsy/methods
- Lung Neoplasms/diagnostic imaging
- Lung Neoplasms/genetics
- Lung Neoplasms/mortality
- Lung Neoplasms/surgery
- Neoplasm Staging
- Neoplastic Cells, Circulating/metabolism
- Neoplastic Cells, Circulating/pathology
- Prognosis
- Radiography
- Small Cell Lung Carcinoma/diagnostic imaging
- Small Cell Lung Carcinoma/genetics
- Small Cell Lung Carcinoma/mortality
- Small Cell Lung Carcinoma/surgery
- Survival Analysis
Collapse
Affiliation(s)
- Sean Blandin Knight
- North West Lung Centre, University Hospital South Manchester, Manchester, UK
| | - Phil A Crosbie
- North West Lung Centre, University Hospital South Manchester, Manchester, UK
- Cancer Research UK Lung Cancer Centre of Excellence at Manchester and University College London, UK
| | - Haval Balata
- North West Lung Centre, University Hospital South Manchester, Manchester, UK
| | - Jakub Chudziak
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK
| | - Tracy Hussell
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, UK
| | - Caroline Dive
- Cancer Research UK Lung Cancer Centre of Excellence at Manchester and University College London, UK
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK
| |
Collapse
|
175
|
Nakajima K, Nangia-Makker P, Hogan V, Raz A. Cancer Self-Defense: An Immune Stealth. Cancer Res 2017; 77:5441-5444. [PMID: 28838888 DOI: 10.1158/0008-5472.can-17-1324] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 07/05/2017] [Accepted: 08/04/2017] [Indexed: 12/16/2022]
Abstract
The hurdles in realizing successful cancer immunotherapy stem from the fact that cancer patients are either refractory to immune response and/or develop resistance. Here, we propose that these phenomena are due, in part, to the deployment/secretion of a "decoy flare," for example, anomalous cancer-associated antigens by the tumor cells. The cancer secretome, which resembles the parent cell make-up, is composed of soluble macromolecules (proteins, glycans, lipids, DNAs, RNAs, etc.) and insoluble vesicles (exosomes), thus hindering cancer detection/recognition by immunotherapeutic agents, resulting in a "cancer-stealth" effect. Immunotherapy, or any treatment that relies on antigens' expression/function, could be improved by the understanding of the properties of the cancer secretome, as its clinical evaluation may change the therapeutic landscape. Cancer Res; 77(20); 5441-4. ©2017 AACR.
Collapse
Affiliation(s)
- Kosei Nakajima
- Department of Oncology, Karmanos Cancer Institute, School of Medicine, Wayne State University, Detroit, Michigan
| | - Pratima Nangia-Makker
- Department of Oncology, Karmanos Cancer Institute, School of Medicine, Wayne State University, Detroit, Michigan
| | - Victor Hogan
- Department of Oncology, Karmanos Cancer Institute, School of Medicine, Wayne State University, Detroit, Michigan
| | - Avraham Raz
- Department of Oncology, Karmanos Cancer Institute, School of Medicine, Wayne State University, Detroit, Michigan. .,Department of Pathology, Karmanos Cancer Institute, School of Medicine, Wayne State University, Detroit, Michigan
| |
Collapse
|
176
|
Fortner RT, Damms-Machado A, Kaaks R. Systematic review: Tumor-associated antigen autoantibodies and ovarian cancer early detection. Gynecol Oncol 2017; 147:465-480. [PMID: 28800944 DOI: 10.1016/j.ygyno.2017.07.138] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 07/20/2017] [Accepted: 07/24/2017] [Indexed: 12/20/2022]
Abstract
OBJECTIVES Tumor-associated autoantibodies (AAbs), produced as an immune response to tumor-associated antigens (TAAs), are a novel pathway of early detection markers. METHODS We conducted a systematic review on AAbs and ovarian cancer to summarize the diagnostic performance of individual AAbs and AAb panels. A total of 29 studies including 85 AAbs were included; 27 of the studies were conducted in prevalent cases and cancer-free controls and 2 investigations included pre-diagnosis samples. The majority of studies were hypothesis-driven, evaluating AAbs to target TAAs; 10 studies used screening approaches such as serological expression cloning (SEREX) and nucleic acid-programmable protein arrays (NAPPA). RESULTS The highest sensitivities for individual AAbs were reported for RhoGDI-AAbs (89.5%) and TUBA1C-AAbs (89%); however, specificity levels were relatively low (80% and 75%, respectively). High sensitivities at high specificities were reported for HOXA7-AAbs for detection of moderately differentiated ovarian tumors (66.7% sensitivity at 100% specificity) and IL8-AAbs in stage I-II ovarian cancer (65.5% sensitivity at 98% specificity). A panel of 11 AAbs (ICAM3, CTAG2, p53, STYXL1, PVR, POMC, NUDT11, TRIM39, UHMK1, KSR1, and NXF3) provided 45% sensitivity at 98% specificity for serous ovarian cancer, when at least 2 AAbs were above a threshold of 95% specificity. Twelve of the AAbs identified in this review were investigated in more than one study. Data on diagnostic discrimination by tumor histology and stage at diagnosis are sparse. Limited data suggest select AAb markers improve diagnostic discrimination when combined with markers such as CA125 and HE4. CONCLUSIONS AAbs for ovarian cancer early detection is an emerging area, and large-scale, prospective investigations considering histology and stage are required for discovery and validation. However, data to date suggests panels of AAbs may eventually reach sufficient diagnostic discrimination to allow earlier detection of disease as a complement to existing markers and transvaginal ultrasound.
Collapse
Affiliation(s)
| | - Antje Damms-Machado
- Division of Cancer Epidemiology, German Cancer Research Center (DFKZ), Heidelberg, Germany
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, German Cancer Research Center (DFKZ), Heidelberg, Germany.
| |
Collapse
|
177
|
Gupta S, Mukherjee S, Syed P, Pandala NG, Choudhary S, Singh VA, Singh N, Zhu H, Epari S, Noronha SB, Moiyadi A, Srivastava S. Evaluation of autoantibody signatures in meningioma patients using human proteome arrays. Oncotarget 2017; 8:58443-58456. [PMID: 28938569 PMCID: PMC5601665 DOI: 10.18632/oncotarget.16997] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 03/11/2017] [Indexed: 12/26/2022] Open
Abstract
Meningiomas are one of the most common tumors of the Central nervous system (CNS). This study aims to identify the autoantibody biomarkers in meningiomas using high-density human proteome arrays (~17,000 full-length recombinant human proteins). Screening of sera from 15 unaffected healthy individuals, 10 individuals with meningioma grade I and 5 with meningioma grade II was performed. This comprehensive proteomics based investigation revealed the dysregulation of 489 and 104 proteins in grades I and II of meningioma, respectively, along with the enrichment of several signalling pathways, which might play a crucial role in the manifestation of the disease. Autoantibody targets like IGHG4, CRYM, EFCAB2, STAT6, HDAC7A and CCNB1 were significantly dysregulated across both the grades. Further, we compared this to the tissue proteome and gene expression profile from GEO database. Previously reported upregulated proteins from meningioma tissue-based proteomics obtained from high-resolution mass spectrometry demonstrated an aggravated autoimmune response, emphasizing the clinical relevance of these targets. Some of these targets like SELENBP1 were tested for their presence in tumor tissue using immunoblotting. In the light of highly invasive diagnostic modalities employed to diagnose CNS tumors like meningioma, these autoantibody markers offer a minimally invasive diagnostic platform which could be pursued further for clinical translation.
Collapse
Affiliation(s)
- Shabarni Gupta
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Shuvolina Mukherjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Parvez Syed
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India.,Department of Biochemistry/Biotechnology, University of Turku, Turun yliopisto, Finland
| | - Narendra Goud Pandala
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Saket Choudhary
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, India.,Molecular and Computational Biology, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - Vedita Anand Singh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Namrata Singh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, 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, India
| | - Santosh B Noronha
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, India
| | | | - Sanjeeva Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| |
Collapse
|
178
|
Macdonald IK, Parsy-Kowalska CB, Chapman CJ. Autoantibodies: Opportunities for Early Cancer Detection. Trends Cancer 2017; 3:198-213. [PMID: 28718432 DOI: 10.1016/j.trecan.2017.02.003] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 02/06/2017] [Accepted: 02/07/2017] [Indexed: 12/18/2022]
Abstract
Cancer cells can induce an immunological response resulting in the production of tumor-associated (TA) autoantibodies. These serum immunobiomarkers have been detected for a range of cancers at an early stage before the development of clinical symptoms. Their measurement is minimally invasive and cost effective using established technologies. TA autoantibodies are present in a clinically significant number of individuals and could supplement current screening modalities to aid early diagnosis of high-risk populations and assist the clinical management of patients. Here we review their production, discovery, and validation as biomarkers for cancer and their current and future potential as clinical tools.
Collapse
|
179
|
Abstract
Autoimmunity and cancer have a multifarious epidemiology. Often, it is because of an impaired genome, culminating in functional aberrations in the human system. Systemic lupus erythematosus (SLE) is a heterogeneous complex disease which ensues due to the failure of the immune system to distinguish between self and non-self antigens, thus producing autoantibodies against DNA, RNA and proteins. Cancer, the other side of the same coin, results from an excessive proliferation of cells that evade immune regulation as a result of incompetent defense by T-cells, B-cells and macrophages. Recent findings have indicated that lupus autoantibodies could be used as an effective weapon to kill cancerous cells. This is an attempt to take an account of malicious 'lupus autoantibodies' and their role in neutralizing cancerous cells which may help in enhancing the survival rate of cancer patients, hence, killers can be saviors.
Collapse
Affiliation(s)
- G Singh
- 1 Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Gautam Buddha Nagar, Greater Noida, Uttar Pradesh, India
| | - A K Agarwal
- 2 Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - J Prosek
- 2 Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - M S K Jayadev
- 1 Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Gautam Buddha Nagar, Greater Noida, Uttar Pradesh, India
| | - A Singh
- 1 Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Gautam Buddha Nagar, Greater Noida, Uttar Pradesh, India
| |
Collapse
|
180
|
Rastogi A, Ali A, Tan SH, Banerjee S, Chen Y, Cullen J, Xavier CP, Mohamed AA, Ravindranath L, Srivastav J, Young D, Sesterhenn IA, Kagan J, Srivastava S, McLeod DG, Rosner IL, Petrovics G, Dobi A, Srivastava S, Srinivasan A. Autoantibodies against oncogenic ERG protein in prostate cancer: potential use in diagnosis and prognosis in a panel with C-MYC, AMACR and HERV-K Gag. Genes Cancer 2017; 7:394-413. [PMID: 28191285 PMCID: PMC5302040 DOI: 10.18632/genesandcancer.126] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Overdiagnosis and overtreatment of prostate cancer (CaP) is attributable to widespread reliance on PSA screening in the US. This has prompted us and others to search for improved biomarkers for CaP, to facilitate early detection and disease stratification. In this regard, autoantibodies (AAbs) against tumor antigens could serve as potential candidates for diagnosis and prognosis of CaP. Towards this, our goals were: i) To investigate whether AAbs against ERG oncoprotein (overexpressed in 25-50% of Caucasian American and African American CaP) are present in the sera of CaP patients; ii) To evaluate an AAb panel to enhance CaP detection. The results using an enzyme-linked immunosorbent assay (ELISA) showed that anti-ERG AAbs are present in a significantly higher proportion in the sera of CaP patients compared to healthy controls (p = 0.0001). Furthermore, a panel of AAbs against ERG, AMACR and human endogenous retrovirus-K Gag successfully differentiated CaP patient sera from healthy controls (AUC = 0.791). These results demonstrate for the first time that anti-ERG AAbs are present in the sera of CaP patients. In addition, the data also suggest that AAbs against ERG together with AMACR and HERV-K Gag may be a useful panel of biomarkers for diagnosis and prognosis of CaP.
Collapse
Affiliation(s)
- Anshu Rastogi
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Amina Ali
- Urology Service, Department of Surgery, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Shyh-Han Tan
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Sreedatta Banerjee
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Yongmei Chen
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Jennifer Cullen
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Charles P Xavier
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Ahmed A Mohamed
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Lakshmi Ravindranath
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Jigisha Srivastav
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Denise Young
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | | | - Jacob Kagan
- Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, USA
| | - Sudhir Srivastava
- Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, USA
| | - David G McLeod
- Urology Service, Department of Surgery, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Inger L Rosner
- Urology Service, Department of Surgery, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Gyorgy Petrovics
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Albert Dobi
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Shiv Srivastava
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Alagarsamy Srinivasan
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| |
Collapse
|
181
|
Gupta S, Manubhai KP, Mukherjee S, Srivastava S. Serum Profiling for Identification of Autoantibody Signatures in Diseases Using Protein Microarrays. Methods Mol Biol 2017; 1619:303-315. [PMID: 28674893 DOI: 10.1007/978-1-4939-7057-5_21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Protein microarrays are platforms for studying protein-protein interactions and identifying disease-related self-antigens/autoantigens, which elicit an immune response in a high-throughput format. Protein arrays have been extensively used over the past two decades for several clinical applications. By using this platform, serum containing autoantibodies against potential self-antigens can be screened on proteome-wide arrays, harboring a large repertoire of full-length human proteins. Identification of such autoantigens can help deducing early diagnostic, as well as, prognostic markers in case of malignancies, autoimmune disorders, and other systemic diseases. Here, we provide an overview of the protein microarray technology along with details of an established method to study autoantibody profiles from patient sera.
Collapse
Affiliation(s)
- Shabarni Gupta
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, 400076, India
| | - K P Manubhai
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, 400076, India
| | - Shuvolina Mukherjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, 400076, India
| | - Sanjeeva Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, 400076, India.
| |
Collapse
|
182
|
Polyspecificity of Anti-lipid A Antibodies and Its Relevance to the Development of Autoimmunity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 966:181-202. [PMID: 28887790 DOI: 10.1007/5584_2017_94] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The process of natural selection favours germ-line gene segments that encode CDRs that have the ability to recognize a range of structurally related antigens. This presents an immunological advantage to the host, as it can confer protection against a common pathogen and still cope with new or changing antigens. Cross-reactive and polyspecific antibodies also play a central role in autoimmune responses, and a link has been shown to exist between auto-reactive B cells and certain bacterial infections. Bacterial DNA, lipids, and carbohydrates have been implicated in the progression of autoimmune diseases such as systemic lupus erythematosus. As well, reports of anti-lipid A antibody polyspecificity towards single-stranded DNA together with the observed sequence homology amongst isolated auto- and anti-lipid A antibodies has prompted further study of this phenomenon. Though the lipid A epitope appears cryptic during Gram-negative bacterial infection, there have been several reported instances of lipid A-specific antibodies isolated from human sera, some of which have exhibited polyspecificity for single stranded DNA. In such cases, the breakdown of negative selection through polyspecificity can have the unfortunate consequence of autoimmune disease. This review summarizes current knowledge regarding such antibodies and emphasizes the features of S1-15, A6, and S55-5, anti-lipid A antibodies whose structures were recently determined by X-ray crystallography.
Collapse
|
183
|
Garranzo-Asensio M, Guzman-Aranguez A, Povés C, Fernández-Aceñero MJ, Torrente-Rodríguez RM, Ruiz-Valdepeñas Montiel V, Domínguez G, Frutos LS, Rodríguez N, Villalba M, Pingarrón JM, Campuzano S, Barderas R. Toward Liquid Biopsy: Determination of the Humoral Immune Response in Cancer Patients Using HaloTag Fusion Protein-Modified Electrochemical Bioplatforms. Anal Chem 2016; 88:12339-12345. [PMID: 28193070 DOI: 10.1021/acs.analchem.6b03526] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- María Garranzo-Asensio
- Departamento
de Bioquímica y Biología Molecular IV, Facultad de Óptica
y Optometría, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| | - Ana Guzman-Aranguez
- Departamento
de Bioquímica y Biología Molecular IV, Facultad de Óptica
y Optometría, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| | - Carmen Povés
- Gastroenterology
Unit, Hospital Universitario Clínico San Carlos, E-28040 Madrid, Spain
| | | | - Rebeca M. Torrente-Rodríguez
- Departamento
de Química Analítica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Víctor Ruiz-Valdepeñas Montiel
- Departamento
de Química Analítica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Gemma Domínguez
- Departamento de Medicina, Facultad de Medicina, Instituto de Investigaciones Biomédicas “Alberto Sols”, CSIC-UAM, E-28029 Madrid, Spain
| | - Luis San Frutos
- Gynecology
and Obstetrics Department, Hospital Puerta de Hierro, E-28222 Majadahonda, Spain
| | - Nuria Rodríguez
- Medical
Oncology Department, Hospital Universitario La Paz, E-28046 Madrid, Spain
| | - Mayte Villalba
- Departamento
de Bioquímica y Biología Molecular I Facultad de Ciencias
Químicas, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| | - José M. Pingarrón
- Departamento
de Química Analítica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Susana Campuzano
- Departamento
de Química Analítica, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Rodrigo Barderas
- Departamento
de Bioquímica y Biología Molecular I Facultad de Ciencias
Químicas, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| |
Collapse
|
184
|
A protein fragment derived from DNA-topoisomerase I as a novel tumour-associated antigen for the detection of early stage carcinoma. Br J Cancer 2016; 115:1555-1564. [PMID: 27875523 PMCID: PMC5155364 DOI: 10.1038/bjc.2016.369] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 09/07/2016] [Accepted: 10/08/2016] [Indexed: 12/19/2022] Open
Abstract
Background: The production of autoantibodies against tumour-associated antigens (TAAs) is believed to reflect greater immunologic reactivity in cancer patients and enhanced immune surveillance for cancer cells. Over the past few decades, a number of different TAAs and their corresponding autoantibodies have been investigated. However, positive frequency of autoantibody detection in cancer patients has been relatively low. Here we describe a novel TAA that was a fragment derived from human DNA-topoiomerase I and an autoantibody against the novel TAA with relatively high positive frequency in the sera of early-stage non-small-cell lung cancer (NSCLC), gastric cancer (GC), colorectal cancer (CRC) and oesophageal squamous cell carcinoma (ESCC). Methods: Serologic enzyme-linked immunosorbent assay (ELISA) and western blot were used to discover a novel TAA with a molecular weight of 48 kDa separated by ion exchange chromatography. Autoantibody ELISA, immnohistochemistry and immunofluorescent staining, recombinant protein cloning/expression and western blot were used to identify the novel TAA. The association of the autoantibody against the novel TAA with early-stage carcinoma was explored by screening 203 stage I/II patients and 170 stage III/IV patients with NSCLC, GC, CRC or ESCC. Results: We identified the novel TAA as a fragment derived from human DNA-topoiomerase I (TOP1). We found that the novel TAA induced specific autoantibodies with a high prevalence that ranged from 58 to 72% in some of the most common types of cancer. We observed that the immune response against the novel TAA was associated with early stage ESCC, GC, CRC and NSCLC. Conclusions: The findings in this study suggest that the autoantibody against the novel TAA may be a potential biomarker for use in the early detection and diagnosis of cancer.
Collapse
|
185
|
Atak A, Mukherjee S, Jain R, Gupta S, Singh VA, Gahoi N, K P M, Srivastava S. Protein microarray applications: Autoantibody detection and posttranslational modification. Proteomics 2016; 16:2557-2569. [PMID: 27452627 DOI: 10.1002/pmic.201600104] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 07/09/2016] [Accepted: 07/19/2016] [Indexed: 12/18/2022]
Abstract
The discovery of DNA microarrays was a major milestone in genomics; however, it could not adequately predict the structure or dynamics of underlying protein entities, which are the ultimate effector molecules in a cell. Protein microarrays allow simultaneous study of thousands of proteins/peptides, and various advancements in array technologies have made this platform suitable for several diagnostic and functional studies. Antibody arrays enable researchers to quantify the abundance of target proteins in biological fluids and assess PTMs by using the antibodies. Protein microarrays have been used to assess protein-protein interactions, protein-ligand interactions, and autoantibody profiling in various disease conditions. Here, we summarize different microarray platforms with focus on its biological and clinical applications in autoantibody profiling and PTM studies. We also enumerate the potential of tissue microarrays to validate findings from protein arrays as well as other approaches, highlighting their significance in proteomics.
Collapse
Affiliation(s)
- Apurva Atak
- Proteomics Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Shuvolina Mukherjee
- Proteomics Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Rekha Jain
- Proteomics Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Shabarni Gupta
- Proteomics Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Vedita Anand Singh
- Proteomics Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Nikita Gahoi
- Proteomics Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Manubhai K P
- Proteomics Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Sanjeeva Srivastava
- Proteomics Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India.
| |
Collapse
|