1
|
Hua Y, Jiang P, Dai C, Li M. Extracellular vesicle autoantibodies. J Autoimmun 2024; 149:103322. [PMID: 39341173 DOI: 10.1016/j.jaut.2024.103322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 09/30/2024]
Abstract
Autoantibodies are immunoglobulin proteins produced by autoreactive B cells responding to self-antigens. Extracellular vesicles (EVs) are membranous structures released by almost all types of cells and extensively distributed in various biological fluids. Studies have indicated that EVs loaded with self-antigens not only play important roles in antigen presentation and autoantibody production but can also form functional immune complexes with autoantibodies (termed EV autoantibodies). While numerous papers have summarized the production and function of pathogenic autoantibodies in diseases, especially autoimmune diseases, reviews on EV autoantibodies are rare. In this review, we outline the existing knowledge about EVs, autoantibodies, and EV antigens, highlighting the formation of EV autoantibodies and their functions in autoimmune diseases and cancers. In conclusion, EV autoantibodies may be involved in the occurrence of disease(s) and also serve as potential non-invasive markers that could help in the diagnosis and/or prognosis of disease. Additional studies designed to define in more detail the molecular characteristics of EV autoantibodies and their contribution to disease are recommended.
Collapse
Affiliation(s)
- Yan Hua
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230031, China; Department of Laboratory Medicine, Anhui Provincial Cancer Hospital, Hefei, Anhui, 230031, China; Core Unit of National Clinical Research Center for Laboratory Medicine of China, Hefei, Anhui, 230001, China
| | - Panpan Jiang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230031, China; Department of Laboratory Medicine, Anhui Provincial Cancer Hospital, Hefei, Anhui, 230031, China
| | - Chunyang Dai
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230031, China; Department of Laboratory Medicine, Anhui Provincial Cancer Hospital, Hefei, Anhui, 230031, China; Core Unit of National Clinical Research Center for Laboratory Medicine of China, Hefei, Anhui, 230001, China
| | - Ming Li
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230031, China; Department of Laboratory Medicine, Anhui Provincial Cancer Hospital, Hefei, Anhui, 230031, China; Core Unit of National Clinical Research Center for Laboratory Medicine of China, Hefei, Anhui, 230001, China.
| |
Collapse
|
2
|
Strum S, Andersen MH, Svane IM, Siu LL, Weber JS. State-Of-The-Art Advancements on Cancer Vaccines and Biomarkers. Am Soc Clin Oncol Educ Book 2024; 44:e438592. [PMID: 38669611 DOI: 10.1200/edbk_438592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
The origins of cancer vaccines date back to the 1800s. Since then, there have been significant efforts to generate vaccines against solid and hematologic malignancies using a variety of platforms. To date, these efforts have generally been met with minimal success. However, in the era of improved methods and technological advancements, supported by compelling preclinical and clinical data, a wave of renewed interest in the field offers the promise of discovering field-changing paradigms in the management of established and resected disease using cancer vaccines. These include novel approaches to personalized neoantigen vaccine development, as well as innovative immune-modulatory vaccines (IMVs) that facilitate activation of antiregulatory T cells to limit immunosuppression caused by regulatory immune cells. This article will introduce some of the limitations that have affected cancer vaccine development over the past several decades, followed by an introduction to the latest advancements in neoantigen vaccine and IMV therapy, and then conclude with a discussion of some of the newest technologies and progress that are occurring across the cancer vaccine space. Cancer vaccines are among the most promising frontiers for breakthrough innovations and strategies poised to make a measurable impact in the ongoing fight against cancer.
Collapse
Affiliation(s)
- Scott Strum
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada
| | - Mads Hald Andersen
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
| | - Lillian L Siu
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada
| | - Jeffrey S Weber
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY
| |
Collapse
|
3
|
Soldin I, Pereira N. Autoimmunity and Cancer: Two Stations on the Same Continuum. Cureus 2024; 16:e54317. [PMID: 38496074 PMCID: PMC10944658 DOI: 10.7759/cureus.54317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/16/2024] [Indexed: 03/19/2024] Open
Abstract
INTRODUCTION Autoimmunity has been associated with different types of cancer, including hematological malignancies like lymphomas, and solid tumors. Additionally, the potential role of medication-induced immunosuppression should be considered. AIM Our study aimed to investigate the relationship between autoimmunity and the development of cancer, as well as the impact of immunosuppressive drugs on increasing cancer risk. METHODS The study sample was composed of patients who developed cancer after the administration of biological agents for the treatment of autoimmune disorders. Selected patients were treated in our hospital between 1st January 2011 and 31st December 2021 and followed up in internal medicine, gastroenterology, or dermatology consult. From 434 patients with autoimmune diseases using biological agents, only 20 developed cancer, which was our final study sample. The data analysis was performed using the IQVIR package version 2.0.2 (IQVIA, Durham, NC). A p-value of <0.05 was considered statistically significant. RESULTS We found a significant correlation between long-term corticosteroid therapy and an increased risk of cancer. However, the effect of biological therapies on cancer risk was not statistically significant. It's worth noting that our sample size was small, so we cannot extrapolate these findings. CONCLUSIONS Physicians need to be aware that treating autoimmune diseases with immunosuppressive therapies may contribute to the development of cancer. Further research is needed to determine the impact of such treatments on cancer prognosis.
Collapse
Affiliation(s)
- Inês Soldin
- Medical Oncology, Instituto Português de Oncologia do Porto Francisco Gentil, Porto, PRT
| | - Nídia Pereira
- Internal Medicine, Hospital Pedro Hispano, Matosinhos, PRT
| |
Collapse
|
4
|
Wang K, Qiu C, Xing M, Li M, Wang B, Ye H, Shi J, Dai L, Wang X, Wang P. Association of elevated autoantibody to high expression of GNAS in hepatocellular carcinoma. Heliyon 2023; 9:e22627. [PMID: 38107305 PMCID: PMC10724561 DOI: 10.1016/j.heliyon.2023.e22627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 12/19/2023] Open
Abstract
Purpose This study was based on hepatocellular carcinoma (HCC) patients of early-stage to explore the diagnostic capability and possible production causes of anti-GNAS autoantibody. Methods We evaluated the frequency of anti-GNAS autoantibody in sera from patients with early-stage HCC by enzyme-linked immunosorbent assay (ELISA) and the expression of GNAS protein in early-stage HCC tissues by immunohistochemistry. Western blotting (WB) and real-time polymerase chain reaction (RT-PCR) were utilized to examine the expressions of GNAS protein and mRNA in cell lines. GEO and International Cancer Genome Consortium (ICGC) databases were inquired to explore mRNA expression and mutation of GNAS in HCC tissues. Results The positive rates of anti-GNAS autoantibody in HCC patients at clinical stage I (78.1 %) and clinical stage II (57.1 %) were all significantly higher than that in healthy control (20 %). There was also a significant difference in GNAS protein expression between HCC and its adjacent normal liver tissues. The results from WB and RT-PCR showed a significant difference at the mRNA level but no statistical difference at the protein level between HCC and normal liver cell lines. The difference in mRNA level between HCC and adjacent normal liver tissues was verified to be significant. Furthermore, the ICGC database demonstrated a 10.6 % mutation frequency for GNAS in HCC patients. Conclusion The coordination of elevated anti-GNAS autoantibody, high expression of GNAS in the mRNA and protein levels in HCC, and high frequency of GNAS mutation indicates that anti-GNAS autoantibody may be used as an early indicator of HCC.
Collapse
Affiliation(s)
- Keyan Wang
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
| | - Cuipeng Qiu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
- Department of Epidemiology and Health Statistics & Henan Key Laboratory for Tumor Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Mengtao Xing
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China
| | - Miao Li
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Bofei Wang
- Department of Epidemiology and Health Statistics & Henan Key Laboratory for Tumor Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Hua Ye
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
- Department of Epidemiology and Health Statistics & Henan Key Laboratory for Tumor Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Jianxiang Shi
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
| | - Liping Dai
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
| | - Xiao Wang
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
| | - Peng Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, China
- Department of Epidemiology and Health Statistics & Henan Key Laboratory for Tumor Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou 450001, Henan, China
| |
Collapse
|
5
|
Sorda JA, Barreyro FJ, Rojas G, Greco DA, Paes A, Avagnina A, Daruich J, Ballerga EG. Icteric Variant of Stauffer Syndrome as a Paraneoplastic Manifestation of Type 1 Papillary Renal Cell Carcinoma. ACG Case Rep J 2023; 10:e01111. [PMID: 37539377 PMCID: PMC10396334 DOI: 10.14309/crj.0000000000001111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 06/30/2023] [Indexed: 08/05/2023] Open
Abstract
Intrahepatic cholestasis as a paraneoplastic manifestation was first described by Dr. Maurice H. Stauffer in 1961. This paraneoplastic manifestation was primarily associated with renal cell carcinoma characterized by abnormal liver enzymes without hepatic metastasis. Stauffer syndrome is classified into 2 types: classical and jaundice variants. Indeed, the jaundice variant is extremely rare and only described in 13 published cases. We report a case of intrahepatic cholestasis associated with a type 1 papillary renal cell carcinoma with complete resolution after surgical treatment.
Collapse
Affiliation(s)
- Juan Antonio Sorda
- Department of Gastroenterology and Hepatology, University Hospital “José de San Martín”, Faculty of Medicine, University of Buenos Aires, Argentina
| | - Fernando Javier Barreyro
- Laboratory of Molecular Biotechnology (BIOTECMOL), Biotechnology Institute of Misiones (INBIOMIS), National University of Misiones, National Scientific and Technical Research Council (CONICET), Argentina
| | - German Rojas
- Department of Gastroenterology and Hepatology, University Hospital “José de San Martín”, Faculty of Medicine, University of Buenos Aires, Argentina
| | - Daniel Alejandro Greco
- Department of Pathology, University Hospital “José de San Martín,” Faculty of Medicine, University of Buenos Aires, Argentina
| | - Andrea Paes
- Department of Pathology, University Hospital “José de San Martín,” Faculty of Medicine, University of Buenos Aires, Argentina
| | - Alejandra Avagnina
- Department of Pathology, University Hospital “José de San Martín,” Faculty of Medicine, University of Buenos Aires, Argentina
| | - Jorge Daruich
- Department of Gastroenterology and Hepatology, University Hospital “José de San Martín”, Faculty of Medicine, University of Buenos Aires, Argentina
| | - Esteban González Ballerga
- Department of Gastroenterology and Hepatology, University Hospital “José de San Martín”, Faculty of Medicine, University of Buenos Aires, Argentina
| |
Collapse
|
6
|
Shin H, Kim Y, Jon S. Nanovaccine Displaying Immunodominant T Cell Epitopes of Fibroblast Activation Protein Is Effective Against Desmoplastic Tumors. ACS NANO 2023. [PMID: 37184372 DOI: 10.1021/acsnano.3c00764] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Cancer-associated fibroblasts (CAFs), which are dominant cell types in the tumor microenvironment (TME), support tumor growth by secreting cytokines and forming an extracellular matrix (ECM) that hampers the penetration of chemical and biological therapeutics within the tumor and thereby limits their therapeutic efficacy. Here, we report a cancer nanovaccine targeting fibroblast activation protein α (FAP)-expressing CAFs as a potential pan-tumor vaccine. We predicted immunodominant FAP-specific epitope peptides in silico and selected two candidate peptides after in vitro and in vivo screening for immunogenicity and antitumor efficacy. Next, we developed a nanoparticle-based vaccine that displays the two selected epitope peptides on the surface of lipid nanoparticles encapsulating CpG adjuvant (FAPPEP-SLNPs). Immunization with one of two FAPPEP-SLNP nanovaccines led to considerable growth inhibition of various tumors, including desmoplastic tumors, by depleting FAP+ CAFs and thereby reducing ECM production in the TME while causing little appreciable adverse effects. Furthermore, when combined with a chemotherapeutic drug, the FAPPEP-SLNP nanovaccine increased drug accumulation and resulted in a synergistic antitumor efficacy far better than that of each corresponding monotherapy. These findings suggest that our FAPPEP-SLNP nanovaccine has potential for use as an "off-the-shelf" pan-tumor vaccine applicable to a variety of tumors and may be a suitable platform for use in various combination therapies.
Collapse
Affiliation(s)
- Hocheol Shin
- Department of Biological Sciences, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon 34141, Republic of Korea
- Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon 34141, Republic of Korea
| | - Yujin Kim
- Department of Biological Sciences, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon 34141, Republic of Korea
- Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon 34141, Republic of Korea
| | - Sangyong Jon
- Department of Biological Sciences, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon 34141, Republic of Korea
- Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon 34141, Republic of Korea
| |
Collapse
|
7
|
Zhu QY. Bioinformatics analysis of the pathogenic link between Epstein-Barr virus infection, systemic lupus erythematosus and diffuse large B cell lymphoma. Sci Rep 2023; 13:6310. [PMID: 37072474 PMCID: PMC10113247 DOI: 10.1038/s41598-023-33585-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 04/15/2023] [Indexed: 05/03/2023] Open
Abstract
Epstein-Barr virus (EBV) is a risk factor for diffuse large B-cell lymphoma (DLBCL) and systemic lupus erythematosus (SLE). While prior research has suggested a potential correlation between SLE and DLBCL, the molecular mechanisms remain unclear. The present study aimed to explore the contribution of EBV infection to the pathogenesis of DLBCL in the individuals with SLE using bioinformatics approaches. The Gene Expression Omnibus database was used to compile the gene expression profiles of EBV-infected B cells (GSE49628), SLE (GSE61635), and DLBCL (GSE32018). Altogether, 72 shared common differentially expressed genes (DEGs) were extracted and enrichment analysis of the shared genes showed that p53 signaling pathway was a common feature of the pathophysiology. Six hub genes were selected using protein-protein interaction (PPI) network analysis, including CDK1, KIF23, NEK2, TOP2A, NEIL3 and DEPDC1, which showed preferable diagnostic values for SLE and DLBCL and involved in immune cell infiltration and immune responses regulation. Finally, TF-gene and miRNA-gene regulatory networks and 10 potential drugs molecule were predicted. Our study revealed the potential molecular mechanisms by which EBV infection contribute to the susceptibility of DLBCL in SLE patients for the first time and identified future biomarkers and therapeutic targets for SLE and DLBCL.
Collapse
Affiliation(s)
- Qian-Ying Zhu
- Department of Laboratory Medicine, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518003, People's Republic of China.
| |
Collapse
|
8
|
Geng Q, Cao X, Fan D, Wang Q, Wang X, Zhang M, Zhao L, Jiao Y, Deng T, Liu H, Zhou J, Lou Y, Liang J, Xiao C. Potential medicinal value of N6-methyladenosine in autoimmune diseases and tumours. Br J Pharmacol 2023. [PMID: 36624563 DOI: 10.1111/bph.16030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/23/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Autoimmune diseases (ADs) are closely related to malignant tumours. On the one hand, ADs can increase the incidence of tumours; on the other hand, malignant tumours can cause rheumatic disease-like manifestations. With the increasing depth of analysis into the mechanism of N6 -methyladenosine (m6A) modification, it has been found that changes in m6A-related modification enzymes are closely related to the occurrence and development of ADs and malignant tumours. In this review, we explore the pathogenesis of ADs and tumours based on m6A modification. According to systematic assessment of the similarities between ADs and tumours, m6A may represent a common target of both diseases. At present, most of the drugs targeting m6A are in the research and development stage, not in clinical trials. Therefore, advancing the development of drugs targeting m6A is of great significance for both the combined treatment of ADs and malignant tumours and improving the quality of life and prognosis of patients.
Collapse
Affiliation(s)
- Qishun Geng
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiaoxue Cao
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Danping Fan
- Beijing Key Laboratory of Research of Chinese Medicine on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qiong Wang
- China-Japan Friendship Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Xing Wang
- China-Japan Friendship Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Mengxiao Zhang
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Lu Zhao
- China-Japan Friendship Hospital, Capital Medical University, Beijing, China
| | - Yi Jiao
- China-Japan Friendship Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Tingting Deng
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Honglin Liu
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Jing Zhou
- Department of Physiology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China
| | - Yanni Lou
- Oncology Department of Integrated Traditional Chinese and Western Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Jing Liang
- Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing, China
| | - Cheng Xiao
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
9
|
Juanes‐Velasco P, García‐Vaquero ML, Landeira‐Viñuela A, Lopez‐Campos JL, Marín C, Lecrevisse Q, Arias‐Hidalgo C, Montalvillo E, Góngora R, Hernández Á, Fuentes M. Systematic evaluation of plasma signaling cascades by functional proteomics approaches: SARS-CoV-2 infection as model. Proteomics Clin Appl 2022; 16:e2100100. [PMID: 36168869 PMCID: PMC9537801 DOI: 10.1002/prca.202100100] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 09/09/2022] [Accepted: 09/21/2022] [Indexed: 12/30/2022]
Abstract
PURPOSE Acute phase reactants (APRs) play a critical role in inflammation. The difference in their physiological functions or the different dynamic ranges of these proteins in plasma makes it difficult to detect them simultaneously and to use several of these proteins as a tool in clinical practice. EXPERIMENTAL DESIGN A novel multiplex assay has been designed and optimized to carry out a high-throughput and simultaneous screening of APRs, allowing the detection of each of them at the same time and in their corresponding dynamic range. RESULTS Using Sars-CoV-2 infection as a model, it has been possible to profile different patterns of acute phase proteins that vary significantly between healthy and infected patients. In addition, severity profiles (acute respiratory distress syndrome and sepsis) have been established. CONCLUSIONS AND CLINICAL RELEVANCE Differential profiles in acute phase proteins can serve as a diagnostic and prognostic tool, among patient stratification. The design of this new platform for their simultaneous detection paves the way for them to be more extensive use in clinical practice.
Collapse
Affiliation(s)
- Pablo Juanes‐Velasco
- Department of Medicine and Cytometry General Service‐Nucleus, CIBERONCCancer Research Centre (IBMCC/CSIC/USAL/IBSAL)SalamancaSpain
| | - Marina L. García‐Vaquero
- Department of Medicine and Cytometry General Service‐Nucleus, CIBERONCCancer Research Centre (IBMCC/CSIC/USAL/IBSAL)SalamancaSpain
| | - Alicia Landeira‐Viñuela
- Department of Medicine and Cytometry General Service‐Nucleus, CIBERONCCancer Research Centre (IBMCC/CSIC/USAL/IBSAL)SalamancaSpain
| | - José Luis Lopez‐Campos
- Unidad Médico‐Quirúrgica de Enfermedades Respiratorias. Instituto de Biomedicina de Sevilla (IBiS)Hospital Universitario Virgen del Rocío/Universidad de SevillaSpain,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES)Instituto de Salud Carlos IIIMadridSpain
| | - Carmen Marín
- Instituto de Biomedicina de Sevilla (IBiS)Hospital Universitario Virgen del RocíoSevillaSpain
| | - Quentin Lecrevisse
- Department of Medicine and Cytometry General Service‐Nucleus, CIBERONCCancer Research Centre (IBMCC/CSIC/USAL/IBSAL)SalamancaSpain
| | - Carlota Arias‐Hidalgo
- Department of Medicine and Cytometry General Service‐Nucleus, CIBERONCCancer Research Centre (IBMCC/CSIC/USAL/IBSAL)SalamancaSpain
| | - Enrique Montalvillo
- Department of Medicine and Cytometry General Service‐Nucleus, CIBERONCCancer Research Centre (IBMCC/CSIC/USAL/IBSAL)SalamancaSpain
| | - Rafael Góngora
- Department of Medicine and Cytometry General Service‐Nucleus, CIBERONCCancer Research Centre (IBMCC/CSIC/USAL/IBSAL)SalamancaSpain
| | - Ángela‐Patricia Hernández
- Department of Medicine and Cytometry General Service‐Nucleus, CIBERONCCancer Research Centre (IBMCC/CSIC/USAL/IBSAL)SalamancaSpain,Department of Pharmaceutical Sciences: Organic Chemistry; Faculty of PharmacyUniversity of Salamanca, CIETUS, IBSALSalamanca37007Spain
| | - Manuel Fuentes
- Department of Medicine and Cytometry General Service‐Nucleus, CIBERONCCancer Research Centre (IBMCC/CSIC/USAL/IBSAL)SalamancaSpain,Proteomics UnitCancer Research Centre (IBMCC/CSIC/USAL/IBSAL)Salamanca37007Spain
| |
Collapse
|
10
|
Effective Combination Immunotherapy with Oncolytic Adenovirus and Anti-PD-1 for Treatment of Human and Murine Ovarian Cancers. Diseases 2022; 10:diseases10030052. [PMID: 35997357 PMCID: PMC9396998 DOI: 10.3390/diseases10030052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/29/2022] [Accepted: 08/04/2022] [Indexed: 12/15/2022] Open
Abstract
Ovarian cancer (OvCa) is one of the most common gynecological cancers and has the highest mortality in this category. Tumors are often detected late, and unfortunately over 70% of OvCa patients experience relapse after first-line treatments. OvCa has shown low response rates to immune checkpoint inhibitor (ICI) treatments, thus leaving room for improvement. We have shown that oncolytic adenoviral therapy with Ad5/3-E2F-d24-hTNFa-IRES-hIL2 (aka. TILT-123) is promising for single-agent treatment of cancer, but also for sensitizing tumors for T-cell dependent immunotherapy approaches, such as ICI treatments. Therefore, this study set out to determine the effect of inhibition of the immune checkpoint inhibitors (ICI), in the context of TILT-123 therapy of OvCa. We show that simultaneous treatment of patient derived samples with TILT-123 and ICIs anti-PD-1 or anti-PD-L1 efficiently reduced overall viability. The combinations induced T cell activation, T cells expressed activation markers more often, and the treatment caused positive microenvironment changes, measured by flow cytometric assays. Furthermore, in an immunocompetent in vivo C57BL/6NHsda mouse model, tumor growth was hindered, when treated with TILT-123, ICI or both. Taken together, this study provides a rationale for using TILT-123 virotherapy in combination with TILT-123 and immune checkpoint inhibitors together in an ovarian cancer OvCa clinical trial.
Collapse
|
11
|
Fransen MF. Immunological Responses to Cancer Therapy. Int J Mol Sci 2022; 23:ijms23136989. [PMID: 35805986 PMCID: PMC9267108 DOI: 10.3390/ijms23136989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 06/16/2022] [Indexed: 02/04/2023] Open
Affiliation(s)
- Marieke F Fransen
- Department of Pulmonary Medicine, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam UMC, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
12
|
Aran A, Garrigós L, Curigliano G, Cortés J, Martí M. Evaluation of the TCR Repertoire as a Predictive and Prognostic Biomarker in Cancer: Diversity or Clonality? Cancers (Basel) 2022; 14:cancers14071771. [PMID: 35406543 PMCID: PMC8996954 DOI: 10.3390/cancers14071771] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/22/2022] [Accepted: 03/29/2022] [Indexed: 02/06/2023] Open
Abstract
Simple Summary The TCR is the T cell antigen receptor, and it is responsible of the T cell activation, through the HLA-antigen complex recognition. Studying the TCR repertoire in patients with cancer can help to better understand the anti-tumoural responses and it has been suggested to have predictive and or/prognostic values, both for the disease and in response to treatments. The aim of this review is to summarize TCR repertoire studies performed in patients with cancer found in the literature, thoroughly analyse the different factors that can be involved in shaping the TCR repertoire, and draw the current conclusions in this field, especially focusing on whether the TCR diversity—or its opposite, the clonality—can be used as predictors or prognostic biomarkers of the disease. Abstract T cells play a vital role in the anti-tumoural response, and the presence of tumour-infiltrating lymphocytes has shown to be directly correlated with a good prognosis in several cancer types. Nevertheless, some patients presenting tumour-infiltrating lymphocytes do not have favourable outcomes. The TCR determines the specificities of T cells, so the analysis of the TCR repertoire has been recently considered to be a potential biomarker for patients’ progression and response to therapies with immune checkpoint inhibitors. The TCR repertoire is one of the multiple elements comprising the immune system and is conditioned by several factors, including tissue type, tumour mutational burden, and patients’ immunogenetics. Its study is crucial to understanding the anti-tumoural response, how to beneficially modulate the immune response with current or new treatments, and how to better predict the prognosis. Here, we present a critical review including essential studies on TCR repertoire conducted in patients with cancer with the aim to draw the current conclusions and try to elucidate whether it is better to encounter higher clonality with few TCRs at higher frequencies, or higher diversity with many different TCRs at lower frequencies.
Collapse
Affiliation(s)
- Andrea Aran
- Immunology Unit, Department of Cell Biology, Physiology and Immunology, Institut de Biotecnologia I Biomedicina (IBB), Universitat Autònoma de Barcelona (UAB), 08193 Bellaterra, Spain;
| | - Laia Garrigós
- International Breast Cancer Center (IBCC), 08017 Barcelona, Spain; (L.G.); (J.C.)
| | - Giuseppe Curigliano
- Division of Early Drug Development, European Institute of Oncology, IRCCS, 20141 Milano, Italy;
- Department of Oncology and Hemato-Oncology, University of Milano, 20122 Milano, Italy
| | - Javier Cortés
- International Breast Cancer Center (IBCC), 08017 Barcelona, Spain; (L.G.); (J.C.)
- Medica Scientia Innovation Research (MedSIR), 08018 Barcelona, Spain
- Medica Scientia Innovation Research (MedSIR), Ridgewood, NJ 07450, USA
- Department of Medicine, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, 28670 Madrid, Spain
| | - Mercè Martí
- Immunology Unit, Department of Cell Biology, Physiology and Immunology, Institut de Biotecnologia I Biomedicina (IBB), Universitat Autònoma de Barcelona (UAB), 08193 Bellaterra, Spain;
- Correspondence: ; Tel.: +34-935812409
| |
Collapse
|
13
|
Juanes-Velasco P, Landeira-Viñuela A, García-Vaquero ML, Lecrevisse Q, Herrero R, Ferruelo A, Góngora R, Corrales F, Rivas JDL, Lorente JA, Hernández ÁP, Fuentes M. SARS-CoV-2 Infection Triggers Auto-Immune Response in ARDS. Front Immunol 2022; 13:732197. [PMID: 35154090 PMCID: PMC8831226 DOI: 10.3389/fimmu.2022.732197] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 01/06/2022] [Indexed: 12/15/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a severe pulmonary disease, which is one of the major complications in COVID-19 patients. Dysregulation of the immune system and imbalances in cytokine release and immune cell activation are involved in SARS-CoV-2 infection. Here, the inflammatory, antigen, and auto-immune profile of patients presenting COVID-19-associated severe ARDS has been analyzed using functional proteomics approaches. Both, innate and humoral responses have been characterized through acute-phase protein network and auto-antibody signature. Severity and sepsis by SARS-CoV-2 emerged to be correlated with auto-immune profiles of patients and define their clinical progression, which could provide novel perspectives in therapeutics development and biomarkers of COVID-19 patients. Humoral response in COVID-19 patients’ profile separates with significant differences patients with or without ARDS. Furthermore, we found that this profile can be correlated with COVID-19 severity and results more common in elderly patients.
Collapse
Affiliation(s)
- Pablo Juanes-Velasco
- Department of Medicine and Cytometry General Service-Nucleus, CIBERONC, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), Salamanca, Spain
| | - Alicia Landeira-Viñuela
- Department of Medicine and Cytometry General Service-Nucleus, CIBERONC, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), Salamanca, Spain
| | - Marina L García-Vaquero
- Department of Medicine and Cytometry General Service-Nucleus, CIBERONC, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), Salamanca, Spain
| | - Quentin Lecrevisse
- Department of Medicine and Cytometry General Service-Nucleus, CIBERONC, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), Salamanca, Spain
| | - Raquel Herrero
- Department of Critical Care Medicine, Hospital Universitario de Getafe, Madrid, Spain.,CIBER de Enfermedades Respiratorias, Instituto de Investigación Carlos III, Madrid, Spain.,Fundación de Investigación Biomédica del Hospital Universitario de Getafe, Madrid, Spain
| | - Antonio Ferruelo
- Fundación de Investigación Biomédica del Hospital Universitario de Getafe, Madrid, Spain
| | - Rafael Góngora
- Department of Medicine and Cytometry General Service-Nucleus, CIBERONC, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), Salamanca, Spain
| | - Fernando Corrales
- Functional Proteomics Laboratory, National Center for Biotechnology, Consejo Superior de Investigaciones Científicas, Madrid, Spain.,PROTEORED-ISCIII, Red Nacional de Laboratorios de Proteomica-ISCIII, Madrid, Spain
| | - Javier De Las Rivas
- Bioinformatics and Functional Genomics Group, Cancer Research Center (IBMCC, CSIC/USAL/IBSAL), Consejo Superior de Investigaciones Científicas & University of Salamanca, Salamanca, Spain
| | - Jose A Lorente
- Department of Critical Care Medicine, Hospital Universitario de Getafe, Madrid, Spain.,CIBER de Enfermedades Respiratorias, Instituto de Investigación Carlos III, Madrid, Spain.,Fundación de Investigación Biomédica del Hospital Universitario de Getafe, Madrid, Spain.,Universidad Europea de Madrid, Madrid, Spain
| | - Ángela-Patricia Hernández
- Department of Medicine and Cytometry General Service-Nucleus, CIBERONC, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), Salamanca, Spain
| | - Manuel Fuentes
- Department of Medicine and Cytometry General Service-Nucleus, CIBERONC, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), Salamanca, Spain.,PROTEORED-ISCIII, Red Nacional de Laboratorios de Proteomica-ISCIII, Madrid, Spain.,Proteomics Unit, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), Salamanca, Spain
| |
Collapse
|