1
|
Felberg A, Bieńkowski M, Stokowy T, Myszczyński K, Polakiewicz Z, Kitowska K, Sądej R, Mohlin F, Kuźniewska A, Kowalska D, Stasiłojć G, Jongerius I, Spaapen R, Mesa-Guzman M, Montuenga LM, Blom AM, Pio R, Okrój M. Elevated expression of complement factor I in lung cancer cells associates with shorter survival-Potentially via non-canonical mechanism. Transl Res 2024; 269:1-13. [PMID: 38395390 DOI: 10.1016/j.trsl.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 01/27/2024] [Accepted: 02/20/2024] [Indexed: 02/25/2024]
Abstract
While numerous membrane-bound complement inhibitors protect the body's cells from innate immunity's autoaggression, soluble inhibitors like complement factor I (FI) are rarely produced outside the liver. Previously, we reported the expression of FI in non-small cell lung cancer (NSCLC) cell lines. Now, we assessed the content of FI in cancer biopsies from lung cancer patients and associated the results with clinicopathological characteristics and clinical outcomes. Immunohistochemical staining intensity did not correlate with age, smoking status, tumor size, stage, differentiation grade, and T cell infiltrates, but was associated with progression-free survival (PFS), overall survival (OS) and disease-specific survival (DSS). Multivariate Cox analysis of low vs. high FI content revealed HR 0.55, 95 % CI 0.32-0.95, p=0.031 for PFS, HR 0.51, 95 % CI 0.25-1.02, p=0.055 for OS, and HR 0.32, 95 % CI 0.12-0.84, p=0.021 for DSS. Unfavorable prognosis might stem from the non-canonical role of FI, as the staining pattern did not correlate with C4d - the product of FI-supported degradation of active complement component C4b. To elucidate that, we engineered three human NSCLC cell lines naturally expressing FI with CRISPR/Cas9 technology, and compared the transcriptome of FI-deficient and FI-sufficient clones in each cell line. RNA sequencing revealed differentially expressed genes engaged in intracellular signaling pathways controlling proliferation, apoptosis, and responsiveness to growth factors. Moreover, in vitro colony-formation assays showed that FI-deficient cells formed smaller foci than FI-sufficient NSCLC cells, but their size increased when purified FI protein was added to the medium. We postulate that a non-canonical activity of FI influences cellular physiology and contributes to the poor prognosis of lung cancer patients.
Collapse
Affiliation(s)
- Anna Felberg
- Department of Cell Biology and Immunology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Dębinki 1 street, 80-211 Gdańsk, Poland
| | | | - Tomasz Stokowy
- Scientific Computing Group, IT Division, University of Bergen, Norway
| | - Kamil Myszczyński
- Centre of Biostatistics and Bioinformatics Analysis, Medical University of Gdańsk, Poland
| | - Zuzanna Polakiewicz
- Department of Molecular Enzymology and Oncology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Poland
| | - Kamila Kitowska
- Department of Molecular Enzymology and Oncology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Poland
| | - Rafał Sądej
- Department of Molecular Enzymology and Oncology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Poland
| | - Frida Mohlin
- Department of Translational Medicine, Lund University, Sweden
| | - Alicja Kuźniewska
- Department of Cell Biology and Immunology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Dębinki 1 street, 80-211 Gdańsk, Poland
| | - Daria Kowalska
- Department of Cell Biology and Immunology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Dębinki 1 street, 80-211 Gdańsk, Poland
| | - Grzegorz Stasiłojć
- Department of Cell Biology and Immunology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Dębinki 1 street, 80-211 Gdańsk, Poland
| | - Ilse Jongerius
- Department of Immunopathology, Sanquin Research, Amsterdam and Landsteiner Laboratory, Amsterdam University Medical Center, University of Amsterdam, The Netherlands; Emma Children's Hospital, Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Amsterdam University Medical Center, The Netherlands
| | - Robbert Spaapen
- Emma Children's Hospital, Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Amsterdam University Medical Center, The Netherlands
| | - Miguel Mesa-Guzman
- Department of Thoracic Surgery, Clinica Universidad de Navarra, Pamplona, Spain
| | - Luis M Montuenga
- Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain; Program in Solid Tumors, Cima Universidad de Navarra, Cancer Center Clinica Universidad de Navarra (CCUN), Pamplona, Spain; Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain; Instituto de Investigación Sanitaria de Navarra (IdisNa), Pamplona, Spain
| | - Anna M Blom
- Department of Translational Medicine, Lund University, Sweden
| | - Ruben Pio
- Program in Solid Tumors, Cima Universidad de Navarra, Cancer Center Clinica Universidad de Navarra (CCUN), Pamplona, Spain; Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain; Instituto de Investigación Sanitaria de Navarra (IdisNa), Pamplona, Spain; Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Marcin Okrój
- Department of Cell Biology and Immunology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Dębinki 1 street, 80-211 Gdańsk, Poland.
| |
Collapse
|
2
|
Fantini M, Tsang KY, Arlen PM. Generation of the therapeutic monoclonal antibody NEO-201, derived from a cancer vaccine, which targets human malignancies and immune suppressor cells. Expert Rev Vaccines 2024; 23:812-829. [PMID: 39186325 DOI: 10.1080/14760584.2024.2397011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 08/13/2024] [Accepted: 08/22/2024] [Indexed: 08/27/2024]
Abstract
INTRODUCTION Cancer vaccines stimulate the activation of specific humoral and cellular adaptive responses against cancer cells.Antibodies generated post vaccination can be isolated and further selected to develop highly specific and potent monoclonal antibodies (mAbs) against tumor-associated antigens. AREAS COVERED This review describes different types of cancer vaccines, the process of the generation of the mAb NEO-201 from the Hollinshead cancer vaccine platform, the characterization of the antigen recognized by NEO-201, the ability of NEO-201 to bind and mediate the killing of cancer cells and immunosuppressive cells (gMDSCs and Tregs) through ADCC and CDC, NEO-201 preclinical and clinical toxicity and efficacy. EXPERT OPINION To overcome the problem of poor clinical efficacy of cancer vaccines, due to the activity of immunosuppressive cells, cancer vaccines could be combined with other immunotherapeutics able to deplete immunosuppressive cells. Results from clinical trials, employing NEO-201 alone or in combination with pembrolizumab, showed that durable stabilization of disease after treatment was due to the ability of NEO-201 to target and reduce the percentage of circulating Tregs and gMDSCs.These findings provide compelling support to combine NEO-201 with cancer vaccines to reintegrate their ability to elicit a robust and durable immune adaptive response against cancer.
Collapse
|
3
|
Lih TM, Cho KC, Schnaubelt M, Hu Y, Zhang H. Integrated glycoproteomic characterization of clear cell renal cell carcinoma. Cell Rep 2023; 42:112409. [PMID: 37074911 DOI: 10.1016/j.celrep.2023.112409] [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: 11/16/2022] [Revised: 03/03/2023] [Accepted: 04/04/2023] [Indexed: 04/20/2023] Open
Abstract
Clear cell renal cell carcinoma (ccRCC), a common form of RCC, is responsible for the high mortality rate of kidney cancer. Dysregulations of glycoproteins have been shown to associate with ccRCC. However, the molecular mechanism has not been well characterized. Here, a comprehensive glycoproteomic analysis is conducted using 103 tumors and 80 paired normal adjacent tissues. Altered glycosylation enzymes and corresponding protein glycosylation are observed, while two of the major ccRCC mutations, BAP1 and PBRM1, show distinct glycosylation profiles. Additionally, inter-tumor heterogeneity and cross-correlation between glycosylation and phosphorylation are observed. The relation of glycoproteomic features to genomic, transcriptomic, proteomic, and phosphoproteomic changes shows the role of glycosylation in ccRCC development with potential for therapeutic interventions. This study reports a large-scale tandem mass tag (TMT)-based quantitative glycoproteomic analysis of ccRCC that can serve as a valuable resource for the community.
Collapse
Affiliation(s)
- T Mamie Lih
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| | - Kyung-Cho Cho
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Michael Schnaubelt
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Yingwei Hu
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Hui Zhang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
4
|
RAGE Inhibitors for Targeted Therapy of Cancer: A Comprehensive Review. Int J Mol Sci 2022; 24:ijms24010266. [PMID: 36613714 PMCID: PMC9820344 DOI: 10.3390/ijms24010266] [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: 09/17/2022] [Revised: 10/28/2022] [Accepted: 12/16/2022] [Indexed: 12/28/2022] Open
Abstract
The receptor for advanced glycation end products (RAGE) is a member of the immunoglobulin family that is overexpressed in several cancers. RAGE is highly expressed in the lung, and its expression increases proportionally at the site of inflammation. This receptor can bind a variety of ligands, including advanced glycation end products, high mobility group box 1, S100 proteins, adhesion molecules, complement components, advanced lipoxidation end products, lipopolysaccharides, and other molecules that mediate cellular responses related to acute and chronic inflammation. RAGE serves as an important node for the initiation and stimulation of cell stress and growth signaling mechanisms that promote carcinogenesis, tumor propagation, and metastatic potential. In this review, we discuss different aspects of RAGE and its prominent ligands implicated in cancer pathogenesis and describe current findings that provide insights into the significant role played by RAGE in cancer. Cancer development can be hindered by inhibiting the interaction of RAGE with its ligands, and this could provide an effective strategy for cancer treatment.
Collapse
|
5
|
Brennan K, Iversen KF, Blanco-Fernández A, Lund T, Plesner T, Mc Gee MM. Extracellular Vesicles Isolated from Plasma of Multiple Myeloma Patients Treated with Daratumumab Express CD38, PD-L1, and the Complement Inhibitory Proteins CD55 and CD59. Cells 2022; 11:3365. [PMID: 36359760 PMCID: PMC9658084 DOI: 10.3390/cells11213365] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/14/2022] [Accepted: 10/20/2022] [Indexed: 09/26/2023] Open
Abstract
Daratumumab (DARA) has improved the outcome of treatment of multiple myeloma (MM). DARA acts via complement-dependent and -independent mechanisms. Resistance to DARA may result from upregulation of the complement inhibitory proteins CD55 and CD59, downregulation of the DARA target CD38 on myeloma cells or altered expression of the checkpoint inhibitor ligand programmed death ligand-1 (PD-L1) or other mechanisms. In this study, EVs were isolated from peripheral blood (PB) and bone marrow (BM) from multiple myeloma (MM) patients treated with DARA and PB of healthy controls. EV size and number and the expression of CD38, CD55, CD59 and PD-L1 as well as the EV markers CD9, CD63, CD81, CD147 were determined by flow cytometry. Results reveal that all patient EV samples express CD38, PD-L1, CD55 and CD59. The level of CD55 and CD59 are elevated on MM PB EVs compared with healthy controls, and the level of PD-L1 on MM PB EVs is higher in patients responding to treatment with DARA. CD147, a marker of various aspects of malignant behaviour of cancer cells and a potential target for therapy, was significantly elevated on MM EVs compared with healthy controls. Furthermore, mass spectrometry data suggests that MM PB EVs bind DARA. This study reveals a MM PB and BM EV protein signature that may have diagnostic and prognostic value.
Collapse
Affiliation(s)
- Kieran Brennan
- School of Biomolecular & Biomedical Science, University College Dublin (UCD), Dublin 4, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin (UCD), Dublin 4, Ireland
| | - Katrine F. Iversen
- Institute of Regional Health Science, University of Southern Denmark, 7100 Vejle, Denmark
- Department of Internal Medicine, Section of Hematology, Lillebaelt Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
| | - Alfonso Blanco-Fernández
- Flow Cytometry Core Technology, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin (UCD), Belfield, Dublin 4, Ireland
| | - Thomas Lund
- Department of Hematology, Odense University Hospital, 5000 Odense, Denmark
| | - Torben Plesner
- Institute of Regional Health Science, University of Southern Denmark, 7100 Vejle, Denmark
- Department of Internal Medicine, Section of Hematology, Lillebaelt Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
| | - Margaret M. Mc Gee
- School of Biomolecular & Biomedical Science, University College Dublin (UCD), Dublin 4, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin (UCD), Dublin 4, Ireland
| |
Collapse
|
6
|
Platt JL, Vercellotti GM. He reshaped the forefront of xenotransplantation: Agustin Pasqual Dalmasso (1933-2021). Xenotransplantation 2022. [PMID: 35903857 DOI: 10.1111/xen.12770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Agustin Pasqual Dalmasso died in December 2021. He was 88 years old and an Honorary Member of the International Xenotransplantation Association. Gus made seminal contributions to understanding and overcoming the barrier complement system poses to xenotransplantation. Those endeavoring to advance xenotransplantation to clinical application and those seeking the most topics in which to devote their life's work could do no better than examining how Gus approached the subjects of his life's work.
Collapse
Affiliation(s)
- Jeffrey L Platt
- Departments of Surgery and Microbiology & Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Gregory M Vercellotti
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| |
Collapse
|
7
|
Tsang KY, Fantini M, Mavroukakis SA, Zaki A, Annunziata CM, Arlen PM. Development and Characterization of an Anti-Cancer Monoclonal Antibody for Treatment of Human Carcinomas. Cancers (Basel) 2022; 14:cancers14133037. [PMID: 35804808 PMCID: PMC9264992 DOI: 10.3390/cancers14133037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/14/2022] [Accepted: 06/17/2022] [Indexed: 11/16/2022] Open
Abstract
NEO-201 is an IgG1 humanized monoclonal antibody (mAb) that binds to tumor-associated variants of carcinoembryonic antigen-related cell adhesion molecule (CEACAM)-5 and CEACAM-6. NEO-201 reacts to colon, ovarian, pancreatic, non-small cell lung, head and neck, cervical, uterine and breast cancers, but is not reactive against most normal tissues. NEO-201 can kill tumor cells via antibody-dependent cell-mediated cytotoxicity (ADCC) and complement dependent cytotoxicity (CDC) to directly kill tumor cells expressing its target. We explored indirect mechanisms of its action that may enhance immune tumor killing. NEO-201 can block the interaction between CEACAM-5 expressed on tumor cells and CEACAM-1 expressed on natural killer (NK) cells to reverse CEACAM-1-dependent inhibition of NK cytotoxicity. Previous studies have demonstrated safety/tolerability in non-human primates, and in a first in human phase 1 clinical trial at the National Cancer Institute (NCI). In addition, preclinical studies have demonstrated that NEO-201 can bind to human regulatory T (Treg) cells. The specificity of NEO-201 in recognizing suppressive Treg cells provides the basis for combination cancer immunotherapy with checkpoint inhibitors targeting the PD-1/PD-L1 pathway.
Collapse
Affiliation(s)
- Kwong yok Tsang
- Precision Biologics, Inc., Bethesda, MD 20814, USA; (M.F.); (S.A.M.); (A.Z.); (P.M.A.)
- Correspondence: ; Tel.: +1-301-500-8646
| | - Massimo Fantini
- Precision Biologics, Inc., Bethesda, MD 20814, USA; (M.F.); (S.A.M.); (A.Z.); (P.M.A.)
| | - Sharon A. Mavroukakis
- Precision Biologics, Inc., Bethesda, MD 20814, USA; (M.F.); (S.A.M.); (A.Z.); (P.M.A.)
| | - Anjum Zaki
- Precision Biologics, Inc., Bethesda, MD 20814, USA; (M.F.); (S.A.M.); (A.Z.); (P.M.A.)
| | - Christina M. Annunziata
- Women’s Malignancy Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Philip M. Arlen
- Precision Biologics, Inc., Bethesda, MD 20814, USA; (M.F.); (S.A.M.); (A.Z.); (P.M.A.)
| |
Collapse
|
8
|
Checkpoints and Immunity in Cancers: Role of GNG12. Pharmacol Res 2022; 180:106242. [DOI: 10.1016/j.phrs.2022.106242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/25/2022] [Accepted: 04/28/2022] [Indexed: 12/24/2022]
|
9
|
Senent Y, Ajona D, González-Martín A, Pio R, Tavira B. The Complement System in Ovarian Cancer: An Underexplored Old Path. Cancers (Basel) 2021; 13:3806. [PMID: 34359708 PMCID: PMC8345190 DOI: 10.3390/cancers13153806] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/25/2021] [Accepted: 07/26/2021] [Indexed: 12/15/2022] Open
Abstract
Ovarian cancer is one of the most lethal gynecological cancers. Current therapeutic strategies allow temporary control of the disease, but most patients develop resistance to treatment. Moreover, although successful in a range of solid tumors, immunotherapy has yielded only modest results in ovarian cancer. Emerging evidence underscores the relevance of the components of innate and adaptive immunity in ovarian cancer progression and response to treatment. Particularly, over the last decade, the complement system, a pillar of innate immunity, has emerged as a major regulator of the tumor microenvironment in cancer immunity. Tumor-associated complement activation may support chronic inflammation, promote an immunosuppressive microenvironment, induce angiogenesis, and activate cancer-related signaling pathways. Recent insights suggest an important role of complement effectors, such as C1q or anaphylatoxins C3a and C5a, and their receptors C3aR and C5aR1 in ovarian cancer progression. Nevertheless, the implication of these factors in different clinical contexts is still poorly understood. Detailed knowledge of the interplay between ovarian cancer cells and complement is required to develop new immunotherapy combinations and biomarkers. In this context, we discuss the possibility of targeting complement to overcome some of the hurdles encountered in the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Yaiza Senent
- Translational Oncology Group, Program in Solid Tumors, Cima University of Navarra, 31008 Pamplona, Spain; (Y.S.); (A.G.-M.); (R.P.); (B.T.)
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IdISNA), 31008 Pamplona, Spain
| | - Daniel Ajona
- Translational Oncology Group, Program in Solid Tumors, Cima University of Navarra, 31008 Pamplona, Spain; (Y.S.); (A.G.-M.); (R.P.); (B.T.)
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IdISNA), 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Antonio González-Martín
- Translational Oncology Group, Program in Solid Tumors, Cima University of Navarra, 31008 Pamplona, Spain; (Y.S.); (A.G.-M.); (R.P.); (B.T.)
- Department of Oncology, Clinica Universidad de Navarra, 28027 Madrid, Spain
| | - Ruben Pio
- Translational Oncology Group, Program in Solid Tumors, Cima University of Navarra, 31008 Pamplona, Spain; (Y.S.); (A.G.-M.); (R.P.); (B.T.)
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IdISNA), 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Beatriz Tavira
- Translational Oncology Group, Program in Solid Tumors, Cima University of Navarra, 31008 Pamplona, Spain; (Y.S.); (A.G.-M.); (R.P.); (B.T.)
- Navarra Institute for Health Research (IdISNA), 31008 Pamplona, Spain
- Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, 31008 Pamplona, Spain
| |
Collapse
|
10
|
Schmitz R, Fitch ZW, Schroder PM, Choi AY, Jackson AM, Knechtle SJ, Kwun J. B cells in transplant tolerance and rejection: friends or foes? Transpl Int 2021; 33:30-40. [PMID: 31705678 DOI: 10.1111/tri.13549] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/21/2019] [Accepted: 11/04/2019] [Indexed: 12/14/2022]
Abstract
Our understanding of the role of B cells in organ transplantation remains incomplete and continues to grow. The majority of research has focused on the detrimental role of antibodies that drive the development of pathogenesis of the transplanted organ. However, it has been shown that not all donor-specific antibodies are harmful and in some circumstances can even promote tolerance through the mechanism of accommodation. Furthermore, B cells can have effects on transplanted organs through their interaction with T cells, namely antigen presentation, cytokine production, and costimulation. More recently, the role and importance of Bregs was introduced to the field of transplantation. Due to this functional and ontogenetic heterogeneity, targeting B cells in transplantation may bring undesired immunologic side effects including increased rejection. Therefore, the selective control of B cells that contribute to the humoral response against donor antigens will continue to be an important and challenging area of research and potentially lead to improved long-term transplant outcomes.
Collapse
Affiliation(s)
- Robin Schmitz
- Department of Surgery, Duke Transplant Center, Duke University Medical Center, Durham, NC, USA
| | - Zachary W Fitch
- Department of Surgery, Duke Transplant Center, Duke University Medical Center, Durham, NC, USA
| | - Paul M Schroder
- Department of Surgery, Duke Transplant Center, Duke University Medical Center, Durham, NC, USA
| | - Ashley Y Choi
- Department of Surgery, Duke Transplant Center, Duke University Medical Center, Durham, NC, USA
| | - Annette M Jackson
- Department of Surgery, Duke Transplant Center, Duke University Medical Center, Durham, NC, USA
| | - Stuart J Knechtle
- Department of Surgery, Duke Transplant Center, Duke University Medical Center, Durham, NC, USA
| | - Jean Kwun
- Department of Surgery, Duke Transplant Center, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
11
|
Shah A, Rauth S, Aithal A, Kaur S, Ganguly K, Orzechowski C, Varshney GC, Jain M, Batra SK. The Current Landscape of Antibody-based Therapies in Solid Malignancies. Am J Cancer Res 2021; 11:1493-1512. [PMID: 33391547 PMCID: PMC7738893 DOI: 10.7150/thno.52614] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 10/21/2020] [Indexed: 02/06/2023] Open
Abstract
Over the past three decades, monoclonal antibodies (mAbs) have revolutionized the landscape of cancer therapy. Still, this benefit remains restricted to a small proportion of patients due to moderate response rates and resistance emergence. The field has started to embrace better mAb-based formats with advancements in molecular and protein engineering technologies. The development of a therapeutic mAb with long-lasting clinical impact demands a prodigious understanding of target antigen, effective mechanism of action, gene engineering technologies, complex interplay between tumor and host immune system, and biomarkers for prediction of clinical response. This review discusses the various approaches used by mAbs for tumor targeting and mechanisms of therapeutic resistance that is not only caused by the heterogeneity of tumor antigen, but also the resistance imposed by tumor microenvironment (TME), including inefficient delivery to the tumor, alteration of effector functions in the TME, and Fc-gamma receptor expression diversity and polymorphism. Further, this article provides a perspective on potential strategies to overcome these barriers and how diagnostic and prognostic biomarkers are being used in predicting response to mAb-based therapies. Overall, understanding these interdependent parameters can improve the current mAb-based formulations and develop novel mAb-based therapeutics for achieving durable clinical outcomes in a large subset of patients.
Collapse
|
12
|
Lin WD, Fan TC, Hung JT, Yeo HL, Wang SH, Kuo CW, Khoo KH, Pai LM, Yu J, Yu AL. Sialylation of CD55 by ST3GAL1 Facilitates Immune Evasion in Cancer. Cancer Immunol Res 2020; 9:113-122. [PMID: 33177111 DOI: 10.1158/2326-6066.cir-20-0203] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 08/13/2020] [Accepted: 11/03/2020] [Indexed: 11/16/2022]
Abstract
Altered glycosylations, which are associated with expression and activities of glycosyltransferases, can dramatically affect the function of glycoproteins and modify the behavior of tumor cells. ST3GAL1 is a sialyltransferase that adds sialic acid to core 1 glycans, thereby terminating glycan chain extension. In breast carcinomas, overexpression of ST3GAL1 promotes tumorigenesis and correlates with increased tumor grade. In pursuing the role of ST3GAL1 in breast cancer using ST3GAL1-siRNA to knockdown ST3GAL1, we identified CD55 to be one of the potential target proteins of ST3GAL1. CD55 is an important complement regulatory protein, preventing cells from complement-mediated cytotoxicity. CD55 had one N-linked glycosylation site in addition to a Ser/Thr-rich domain, which was expected to be heavily O-glycosylated. Detailed analyses of N- and O-linked oligosaccharides of CD55 released from scramble or ST3GAL1 siRNA-treated breast cancer cells by tandem mass spectrometry revealed that the N-glycan profile was not affected by ST3GAL1 silencing. The O-glycan profile of CD55 demonstrated a shift in abundance to nonsialylated core 1 and monosialylated core 2 at the expense of the disialylated core 2 structure after ST3GAL1 silencing. We also demonstrated that O-linked desialylation of CD55 by ST3GAL1 silencing resulted in increased C3 deposition and complement-mediated lysis of breast cancer cells and enhanced sensitivity to antibody-dependent cell-mediated cytotoxicity. These data demonstrated that ST3GAL1-mediated O-linked sialylation of CD55 acts like an immune checkpoint molecule for cancer cells to evade immune attack and that inhibition of ST3GAL1 is a potential strategy to block CD55-mediated immune evasion.
Collapse
Affiliation(s)
- Wen-Der Lin
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Tan-Chi Fan
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Jung-Tung Hung
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Hui-Ling Yeo
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Sheng-Hung Wang
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chu-Wei Kuo
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Kay-Hooi Khoo
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Li-Mei Pai
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - John Yu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Alice L Yu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan. .,Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Department of Pediatrics, University of California, San Diego, La Jolla, California
| |
Collapse
|
13
|
Bordron A, Bagacean C, Tempescul A, Berthou C, Bettacchioli E, Hillion S, Renaudineau Y. Complement System: a Neglected Pathway in Immunotherapy. Clin Rev Allergy Immunol 2020; 58:155-171. [PMID: 31144209 DOI: 10.1007/s12016-019-08741-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Approved for the treatment of autoimmune diseases, hematological malignancies, and solid cancers, several monoclonal antibodies (mAb) make use of complement in their mechanism of action. Such an assessment is based on comprehensive investigations that used mouse models, in vitro studies, and analyses from patients at initiation (basal level to highlight deficiencies) and after treatment initiation (mAb impact on complement), which have further provided key insights into the importance of the complement activation and/or complement deficiencies in mAb activity. Accordingly, new approaches can now be developed with the final objective of increasing the clinical efficacy of mAb. These improvements include (i) the concurrent administration of fresh frozen plasma during mAb therapy; (ii) mAb modifications such as immunoglobulin G subclass switching, Fc mutation, or IgG hexamerization to improve the fixation and activation of C1q; (iii) optimization of the target recognition to induce a higher complement-dependent cytotoxicity (CDC) and/or complement-dependant cellular cytotoxicity (CDCC); and (iv) the control of soluble and cellular complement inhibitors.
Collapse
Affiliation(s)
- Anne Bordron
- Inserm UMR1227, B lymphocytes and autoimmunity, University of Brest, Brest, France
| | - Cristina Bagacean
- Inserm UMR1227, B lymphocytes and autoimmunity, University of Brest, Brest, France.,Service d'Hématologie, CHU de Brest, Brest, France
| | - Adrian Tempescul
- Inserm UMR1227, B lymphocytes and autoimmunity, University of Brest, Brest, France.,Service d'Hématologie, CHU de Brest, Brest, France
| | - Christian Berthou
- Inserm UMR1227, B lymphocytes and autoimmunity, University of Brest, Brest, France.,Service d'Hématologie, CHU de Brest, Brest, France
| | | | - Sophie Hillion
- Inserm UMR1227, B lymphocytes and autoimmunity, University of Brest, Brest, France.,Laboratory of Immunology and Immunotherapy, CHU de Brest, Brest, France
| | - Yves Renaudineau
- Inserm UMR1227, B lymphocytes and autoimmunity, University of Brest, Brest, France. .,Laboratory of Immunology and Immunotherapy, CHU de Brest, Brest, France.
| |
Collapse
|
14
|
Ustyanovska Avtenyuk N, Visser N, Bremer E, Wiersma VR. The Neutrophil: The Underdog That Packs a Punch in the Fight against Cancer. Int J Mol Sci 2020; 21:E7820. [PMID: 33105656 PMCID: PMC7659937 DOI: 10.3390/ijms21217820] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/19/2020] [Accepted: 10/19/2020] [Indexed: 02/07/2023] Open
Abstract
The advent of immunotherapy has had a major impact on the outcome and overall survival in many types of cancer. Current immunotherapeutic strategies typically aim to (re)activate anticancer T cell immunity, although the targeting of macrophage-mediated anticancer innate immunity has also emerged in recent years. Neutrophils, although comprising ≈ 60% of all white blood cells in the circulation, are still largely overlooked in this respect. Nevertheless, neutrophils have evident anticancer activity and can induce phagocytosis, trogocytosis, as well as the direct cytotoxic elimination of cancer cells. Furthermore, therapeutic tumor-targeting monoclonal antibodies trigger anticancer immune responses through all innate Fc-receptor expressing cells, including neutrophils. Indeed, the depletion of neutrophils strongly reduced the efficacy of monoclonal antibody treatment and increased tumor progression in various preclinical studies. In addition, the infusion of neutrophils in murine cancer models reduced tumor progression. However, evidence on the anticancer effects of neutrophils is fragmentary and mostly obtained in in vitro assays or murine models with reports on anticancer neutrophil activity in humans lagging behind. In this review, we aim to give an overview of the available knowledge of anticancer activity by neutrophils. Furthermore, we will describe strategies being explored for the therapeutic activation of anticancer neutrophil activity.
Collapse
Affiliation(s)
| | | | - Edwin Bremer
- Department of Hematology, Cancer Research Center Groningen, University Medical Center Groningen (UMCG), University of Groningen, Hanzeplein 1/DA13, 9713 GZ Groningen, The Netherlands; (N.U.A.); (N.V.)
| | - Valerie R. Wiersma
- Department of Hematology, Cancer Research Center Groningen, University Medical Center Groningen (UMCG), University of Groningen, Hanzeplein 1/DA13, 9713 GZ Groningen, The Netherlands; (N.U.A.); (N.V.)
| |
Collapse
|
15
|
Kumar D, Romero Y, Schuck KN, Smalley H, Subedi B, Fleming SD. Drivers and regulators of humoral innate immune responses to infection and cancer. Mol Immunol 2020; 121:99-110. [PMID: 32199212 PMCID: PMC7207242 DOI: 10.1016/j.molimm.2020.03.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 03/06/2020] [Accepted: 03/09/2020] [Indexed: 12/21/2022]
Abstract
The complement cascade consists of cell bound and serum proteins acting together to protect the host from pathogens, remove cancerous cells and effectively links innate and adaptive immune responses. Despite its usefulness in microbial neutralization and clearance of cancerous cells, excessive complement activation causes an immune imbalance and tissue damage in the host. Hence, a series of complement regulatory proteins present at a higher concentration in blood plasma and on cell surfaces tightly regulate the cascade. The complement cascade can be initiated by B-1 B cell production of natural antibodies. Natural antibodies arise spontaneously without any known exogenous antigenic or microbial stimulus and protect against invading pathogens, clear apoptotic cells, provide tissue homeostasis, and modulate adaptive immune functions. Natural IgM antibodies recognize microbial and cancer antigens and serve as an activator of complement mediated lysis. This review will discuss advances in complement activation and regulation in bacterial and viral infections, and cancer. We will also explore the crosstalk of natural antibodies with bacterial populations and cancer.
Collapse
MESH Headings
- Antigens, Bacterial/immunology
- Antigens, Bacterial/metabolism
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Antigens, Viral/immunology
- Antigens, Viral/metabolism
- Apoptosis/immunology
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- Bacterial Infections/immunology
- Complement Activation
- Complement System Proteins/immunology
- Complement System Proteins/metabolism
- Humans
- Immunity, Humoral
- Immunity, Innate
- Immunoglobulin M/immunology
- Immunoglobulin M/metabolism
- Neoplasms/immunology
- Receptors, Complement/immunology
- Receptors, Complement/metabolism
- Tumor Escape
- Virus Diseases/immunology
Collapse
Affiliation(s)
- Deepak Kumar
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Yeni Romero
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, USA
| | - Kaitlynn N Schuck
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Haley Smalley
- Division of Biology, Kansas State University, Manhattan, KS, USA
| | - Bibek Subedi
- Division of Biology, Kansas State University, Manhattan, KS, USA
| | - Sherry D Fleming
- Division of Biology, Kansas State University, Manhattan, KS, USA.
| |
Collapse
|
16
|
Sinha A, Singh V, Tandon R, Mohan Srivastava L. Dichotomy of complement system: Tumorigenesis or destruction. Immunol Lett 2020; 223:89-96. [PMID: 32333965 DOI: 10.1016/j.imlet.2020.04.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 04/06/2020] [Accepted: 04/18/2020] [Indexed: 01/12/2023]
Abstract
Complement system proteins, their regulators and endpoint effector complex significantly promote tumor growth by upregulation of oncogenic growth factors, activation of mitogenic signalling pathways and breakage of normal cell cycle. Contrastingly, complement cascades, initiated by anti-tumor therapeutic antibodies, also play a pivotal role in therapy response. This contradictory role of complement system possibly be a very crucial factor for the outcomes of antibody mediated immunotherapies. Herein, we reviewed the twin role of the complement system in cancer and also the genetic variations in complement system genes. Future studies should be focused on the biomarker discovery for the personalised cancer immunotherapies.
Collapse
Affiliation(s)
- Ashima Sinha
- Department of BiochemIstry, Sir Ganga Ram Hospital, New Delhi-110060, India; SAGE Publications India Pvt Ltd., New Delhi-110044, India
| | - Virendra Singh
- Laboratory of Precision Medicine Research, School of Biotechnology, Jawaharlal Nehru University, New Delhi-110067, India.
| | - Ravi Tandon
- Laboratory of AIDS research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi-110067, India
| | - Lalit Mohan Srivastava
- Department of Biochemistry and Lab Medicine, Sir Ganga Ram Kolmet Hospital, New Delhi-110005, India.
| |
Collapse
|
17
|
Yeh SJ, Chang CA, Li CW, Wang LHC, Chen BS. Comparing progression molecular mechanisms between lung adenocarcinoma and lung squamous cell carcinoma based on genetic and epigenetic networks: big data mining and genome-wide systems identification. Oncotarget 2019; 10:3760-3806. [PMID: 31217907 PMCID: PMC6557199 DOI: 10.18632/oncotarget.26940] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 04/29/2019] [Indexed: 02/07/2023] Open
Abstract
Non-small-cell lung cancer (NSCLC) is the predominant type of lung cancer in the world. Lung adenocarcinoma (LADC) and lung squamous cell carcinoma (LSCC) are subtypes of NSCLC. We usually regard them as different disease due to their unique molecular characteristics, distinct cells of origin and dissimilar clinical response. However, the differences of genetic and epigenetic progression mechanism between LADC and LSCC are complicated to analyze. Therefore, we applied systems biology approaches and big databases mining to construct genetic and epigenetic networks (GENs) with next-generation sequencing data of LADC and LSCC. In order to obtain the real GENs, system identification and system order detection are conducted on gene regulatory networks (GRNs) and protein-protein interaction networks (PPINs) for each stage of LADC and LSCC. The core GENs were extracted via principal network projection (PNP). Based on the ranking of projection values, we got the core pathways in respect of KEGG pathway. Compared with the core pathways, we found significant differences between microenvironments, dysregulations of miRNAs, epigenetic modifications on certain signaling transduction proteins and target genes in each stage of LADC and LSCC. Finally, we proposed six genetic and epigenetic multiple-molecule drugs to target essential biomarkers in each progression stage of LADC and LSCC, respectively.
Collapse
Affiliation(s)
- Shan-Ju Yeh
- Laboratory of Automatic Control, Signaling Processing, and Systems Biology, Department of Electrical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Chien-An Chang
- Laboratory of Automatic Control, Signaling Processing, and Systems Biology, Department of Electrical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Cheng-Wei Li
- Laboratory of Automatic Control, Signaling Processing, and Systems Biology, Department of Electrical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Lily Hui-Ching Wang
- Department of Medical Science, Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Bor-Sen Chen
- Laboratory of Automatic Control, Signaling Processing, and Systems Biology, Department of Electrical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan.,Department of Electrical Engineering, Yuan Ze University, Chungli 32003, Taiwan
| |
Collapse
|
18
|
Geller A, Yan J. The Role of Membrane Bound Complement Regulatory Proteins in Tumor Development and Cancer Immunotherapy. Front Immunol 2019; 10:1074. [PMID: 31164885 PMCID: PMC6536589 DOI: 10.3389/fimmu.2019.01074] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 04/26/2019] [Indexed: 12/17/2022] Open
Abstract
It has long been understood that the control and surveillance of tumors within the body involves an intricate dance between the adaptive and innate immune systems. At the center of the interplay between the adaptive and innate immune response sits the complement system—an evolutionarily ancient response that aids in the destruction of microorganisms and damaged cells, including cancer cells. Membrane-bound complement regulatory proteins (mCRPs), such as CD46, CD55, and CD59, are expressed throughout the body in order to prevent over-activation of the complement system. These mCRPs act as a double-edged sword however, as they can also over-regulate the complement system to the extent that it is no longer effective at eliminating cancerous cells. Recent studies are now indicating that mCRPs may function as a biomarker of a malignant transformation in numerous cancer types, and further, are being shown to interfere with anti-tumor treatments. This highlights the critical roles that therapeutic blockade of mCRPs can play in cancer treatment. Furthermore, with the complement system having the ability to both directly and indirectly control adaptive T-cell responses, the use of a combinatorial approach of complement-related therapy along with other T-cell activating therapies becomes a logical approach to treatment. This review will highlight the biomarker-related role that mCRP expression may have in the classification of tumor phenotype and predicted response to different anti-cancer treatments in the context of an emerging understanding that complement activation within the Tumor Microenvironment (TME) is actually harmful for tumor control. We will discuss what is known about complement activation and mCRPs relating to cancer and immunotherapy, and will examine the potential for combinatorial approaches of anti-mCRP therapy with other anti-tumor therapies, especially checkpoint inhibitors such as anti PD-1 and PD-L1 monoclonal antibodies (mAbs). Overall, mCRPs play an essential role in the immune response to tumors, and understanding their role in the immune response, particularly in modulating currently used cancer therapeutics may lead to better clinical outcomes in patients with diverse cancer types.
Collapse
Affiliation(s)
- Anne Geller
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY, United States
| | - Jun Yan
- Immuno-Oncology Program, Department of Medicine, The James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY, United States
| |
Collapse
|
19
|
Vlaicu SI, Tatomir A, Rus V, Rus H. Role of C5b-9 and RGC-32 in Cancer. Front Immunol 2019; 10:1054. [PMID: 31156630 PMCID: PMC6530392 DOI: 10.3389/fimmu.2019.01054] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 04/24/2019] [Indexed: 01/13/2023] Open
Abstract
The complement system represents an effective arsenal of innate immunity as well as an interface between innate and adaptive immunity. Activation of the complement system culminates with the assembly of the C5b-9 terminal complement complex on cell membranes, inducing target cell lysis. Translation of this sequence of events into a malignant setting has traditionally afforded C5b-9 a strict antitumoral role, in synergy with antibody-dependent tumor cytolysis. However, in recent decades, a plethora of evidence has revised this view, highlighting the tumor-promoting properties of C5b-9. Sublytic C5b-9 induces cell cycle progression by activating signal transduction pathways (e.g., Gi protein/ phosphatidylinositol 3-kinase (PI3K)/Akt kinase and Ras/Raf1/ERK1) and modulating the activation of cancer-related transcription factors, while shielding malignant cells from apoptosis. C5b-9 also induces Response Gene to Complement (RGC)-32, a gene that contributes to cell cycle regulation by activating the Akt and CDC2 kinases. RGC-32 is expressed by tumor cells and plays a dual role in cancer, functioning as either a tumor promoter by endorsing malignancy initiation, progression, invasion, metastasis, and angiogenesis, or as a tumor suppressor. In this review, we present recent data describing the versatile, multifaceted roles of C5b-9 and its effector, RGC-32, in cancer.
Collapse
Affiliation(s)
- Sonia I Vlaicu
- Department of Internal Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Neurology, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Alexandru Tatomir
- Department of Neurology, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Violeta Rus
- Division of Rheumatology and Immunology, Department of Medicine, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Horea Rus
- Department of Neurology, School of Medicine, University of Maryland, Baltimore, MD, United States
| |
Collapse
|
20
|
Fishelson Z, Kirschfink M. Complement C5b-9 and Cancer: Mechanisms of Cell Damage, Cancer Counteractions, and Approaches for Intervention. Front Immunol 2019; 10:752. [PMID: 31024572 PMCID: PMC6467965 DOI: 10.3389/fimmu.2019.00752] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 03/20/2019] [Indexed: 01/14/2023] Open
Abstract
The interactions of cancer cells with components of the complement system are highly complex, leading to an outcome that is either favorable or detrimental to cancer cells. Currently, we perceive only the "tip of the iceberg" of these interactions. In this review, we focus on the complement terminal C5b-9 complex, known also as the complement membrane attack complex (MAC) and discuss the complexity of its interaction with cancer cells, starting with a discussion of its proposed mode of action in mediating cell death, and continuing with a portrayal of the strategies of evasion exhibited by cancer cells, and closing with a proposal of treatment approaches targeted at evasion strategies. Upon intense complement activation and membrane insertion of sufficient C5b-9 complexes, the afflicted cells undergo regulated necrotic cell death with characteristic damage to intracellular organelles, including mitochondria, and perforation of the plasma membrane. Several pro-lytic factors have been proposed, including elevated intracellular calcium ion concentrations and activated JNK, Bid, RIPK1, RIPK3, and MLKL; however, further research is required to fully characterize the effective cell death signals activated by the C5b-9 complexes. Cancer cells over-express a multitude of protective measures which either block complement activation, thus reducing the number of membrane-inserted C5b-9 complexes, or facilitate the elimination of C5b-9 from the cell surface. Concomitantly, cancer cells activate several protective pathways that counteract the death signals. Blockage of complement activation is mediated by the complement membrane regulatory proteins CD46, CD55, and CD59 and by soluble complement regulators, by proteases that cleave complement proteins and by protein kinases, like CK2, which phosphorylate complement proteins. C5b-9 elimination and inhibition of cell death signals are mediated by caveolin and dynamin, by Hsp70 and Hsp90, by the mitochondrial stress protein mortalin, and by the protein kinases PKC and ERK. It is conceivable that various cancers and cancers at different stages of development will utilize distinct patterns of these and other MAC resistance strategies. In order to enhance the impact of antibody-based therapy on cancer, novel precise reagents that block the most effective protective strategies will have to be designed and applied as adjuvants to the therapeutic antibodies.
Collapse
Affiliation(s)
- Zvi Fishelson
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | |
Collapse
|
21
|
Okrój M, Potempa J. Complement Activation as a Helping Hand for Inflammophilic Pathogens and Cancer. Front Immunol 2019; 9:3125. [PMID: 30687327 PMCID: PMC6335266 DOI: 10.3389/fimmu.2018.03125] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 12/18/2018] [Indexed: 01/01/2023] Open
Abstract
The complement system, an evolutionarily ancient component of innate immunity, is capable of protecting hosts from invading pathogens, either directly, by lysis of target cells, or indirectly, by mobilization of host immune mechanisms. However, this potentially cytotoxic cascade must be tightly regulated, since improperly controlled complement can damage healthy cells and tissues. The practical importance of this axis is highlighted when impairment of complement regulators or bacterial mechanisms of complement evasion result in pathogenic conditions. Recognition of complement as a "double-edged sword" is widely acknowledged, but another, currently underappreciated aspect of complement function has emerged as an important player in homeostatic balance-the dual outcome of complement-mediated inflammation. In most cases, the proinflammatory properties of complement are beneficial to the host. However, certain pathogens have developed the ability to utilize local inflammation as a source of nutrients and as a way to establish a niche for further colonization. Such a strategy can be illustrated in the example of periodontitis. Interestingly, certain tumors also seem to benefit from complement activation products, which promote a proangiogenic and immunosuppressive microenvironment.
Collapse
Affiliation(s)
- Marcin Okrój
- Department of Medical Biotechnology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland
| | - Jan Potempa
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, United States.,Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| |
Collapse
|
22
|
Bordron A, Bagacean C, Mohr A, Tempescul A, Bendaoud B, Deshayes S, Dalbies F, Buors C, Saad H, Berthou C, Pers JO, Renaudineau Y. Resistance to complement activation, cell membrane hypersialylation and relapses in chronic lymphocytic leukemia patients treated with rituximab and chemotherapy. Oncotarget 2018; 9:31590-31605. [PMID: 30167081 PMCID: PMC6114972 DOI: 10.18632/oncotarget.25657] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 06/04/2018] [Indexed: 01/27/2023] Open
Abstract
The anti-CD20-specific monoclonal antibody rituximab (RTX), in combination with chemotherapy, is commonly used for primary treatment in chronic lymphocytic leukemia (CLL). However, relapses remain important and activation of the complement pathway is one of the mechanisms by which RTX generates the destruction of B cells directly by complement-dependent cytotoxicity (CDC), or indirectly by antibody-dependent cellular phagocytosis. In this study, the RTX capacity to induce CDC was established in 69 untreated CLL patients, this cohort including 34 patients tested before the initiation of RTX-chemotherapy. In vitro CDC-resistance to RTX predicts lower response rates to RTX-chemotherapy and shorter treatment free survival. Furthermore, the predictive value of CDC-resistance was independent from the clinical, cytogenetic and FcγR3A V158F polymorphism status. In contrast, CLL cell resistance to CDC predominates in IGHV unmutated patients and was related to an important α2-6 sialyl transferase activity, which in turn increases cell surface α2-6 hypersialylation. Suspected factors associated with resistance to CDC (CD20, CD55, CD59, factor H, GM1, and sphingomyelin) were not differentially expressed or recruited between the two CLL groups. Altogether, results provide evidence that testing RTX capacity to induce CDC in vitro represents an independent predictive factor of therapeutic effects of RTX, and that α2-6 hypersialylation in CLL cells controls RTX response through the control of the complement pathway. At a time when CLL therapy is moving towards chemo-free treatments, further experiments are required to determine whether performing an initial in vitro assay to appreciate CLL CDC resistance might be useful to select patients.
Collapse
Affiliation(s)
- Anne Bordron
- U1227 B Lymphocytes and Autoimmunity, Université de Brest, INSERM, IBSAM, Labex IGO, Networks IC-CGO and REpiCGO from 'Canceropole Grand Ouest, Brest, France
| | - Cristina Bagacean
- U1227 B Lymphocytes and Autoimmunity, Université de Brest, INSERM, IBSAM, Labex IGO, Networks IC-CGO and REpiCGO from 'Canceropole Grand Ouest, Brest, France.,Laboratory of Immunology and Immunotherapy, CHRU Brest, Hôpital Morvan, Brest, France
| | - Audrey Mohr
- U1227 B Lymphocytes and Autoimmunity, Université de Brest, INSERM, IBSAM, Labex IGO, Networks IC-CGO and REpiCGO from 'Canceropole Grand Ouest, Brest, France
| | - Adrian Tempescul
- U1227 B Lymphocytes and Autoimmunity, Université de Brest, INSERM, IBSAM, Labex IGO, Networks IC-CGO and REpiCGO from 'Canceropole Grand Ouest, Brest, France.,Department of Haematology, CHRU Brest, Hôpital Morvan, Brest, France
| | - Boutahar Bendaoud
- U1227 B Lymphocytes and Autoimmunity, Université de Brest, INSERM, IBSAM, Labex IGO, Networks IC-CGO and REpiCGO from 'Canceropole Grand Ouest, Brest, France.,Laboratory of Immunology and Immunotherapy, CHRU Brest, Hôpital Morvan, Brest, France
| | - Stéphanie Deshayes
- U1227 B Lymphocytes and Autoimmunity, Université de Brest, INSERM, IBSAM, Labex IGO, Networks IC-CGO and REpiCGO from 'Canceropole Grand Ouest, Brest, France
| | - Florence Dalbies
- Department of Haematology, CHRU Brest, Hôpital Morvan, Brest, France
| | - Caroline Buors
- Laboratory of Haematology, CHRU Brest, Hôpital Morvan, Brest, France
| | - Hussam Saad
- Department of Haematology, CHRU Brest, Hôpital Morvan, Brest, France
| | - Christian Berthou
- U1227 B Lymphocytes and Autoimmunity, Université de Brest, INSERM, IBSAM, Labex IGO, Networks IC-CGO and REpiCGO from 'Canceropole Grand Ouest, Brest, France.,Department of Haematology, CHRU Brest, Hôpital Morvan, Brest, France
| | - Jacques-Olivier Pers
- U1227 B Lymphocytes and Autoimmunity, Université de Brest, INSERM, IBSAM, Labex IGO, Networks IC-CGO and REpiCGO from 'Canceropole Grand Ouest, Brest, France
| | - Yves Renaudineau
- U1227 B Lymphocytes and Autoimmunity, Université de Brest, INSERM, IBSAM, Labex IGO, Networks IC-CGO and REpiCGO from 'Canceropole Grand Ouest, Brest, France.,Laboratory of Immunology and Immunotherapy, CHRU Brest, Hôpital Morvan, Brest, France
| |
Collapse
|
23
|
Lu M, Zhao X, Xing H, Xun Z, Yang T, Cai C, Wang D, Ding P. Liposome-chaperoned cell-free synthesis for the design of proteoliposomes: Implications for therapeutic delivery. Acta Biomater 2018; 76:1-20. [PMID: 29625253 DOI: 10.1016/j.actbio.2018.03.043] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 03/20/2018] [Accepted: 03/27/2018] [Indexed: 12/12/2022]
Abstract
Cell-free (CF) protein synthesis has emerged as a powerful technique platform for efficient protein production in vitro. Liposomes have been widely studied as therapeutic carriers due to their biocompatibility, biodegradability, low toxicity, flexible surface manipulation, easy preparation, and higher cargo encapsulation capability. However, rapid immune clearance, insufficient targeting capacity, and poor cytoplasmic delivery efficiency substantially restrict their clinical application. The incorporation of functional membrane proteins (MPs) or peptides allows the transfer of biological properties to liposomes and imparts them with improved circulation, increased targeting, and efficient intracellular delivery. Liposome-chaperoned CF synthesis enables production of proteoliposomes in one-step reaction, which not only substantially simplifies the production procedure but also keeps protein functionality intact. Building off these observations, proteoliposomes with integrated MPs represent an excellent candidate for therapeutic delivery. In this review, we describe recent advances in CF synthesis with emphasis on detailing key factors for improving CF expression efficiency. Furthermore, we provide insights into strategies for rational design of proteoliposomal nanodelivery systems via CF synthesis. STATEMENT OF SIGNIFICANCE Liposome-chaperoned CF synthesis has emerged as a powerful approach for the design of recombinant proteoliposomes in one-step reaction. The incorporation of bioactive MPs or peptides into liposomes via CF synthesis can facilitate the development of proteoliposomal nanodelivery systems with improved circulation, increased targeting, and enhanced cellular delivery capacity. Moreover, by adapting lessons learned from natural delivery vehicles, novel bio-inspired proteoliposomes with enhanced delivery properties could be produced in CF systems. In this review, we first give an overview of CF synthesis with focus on enhancing protein expression in liposome-chaperoned CF systems. Furthermore, we intend to provide insight into harnessing CF-synthesized proteoliposomes for efficient therapeutic delivery.
Collapse
|
24
|
Kochanek DM, Ghouse SM, Karbowniczek MM, Markiewski MM. Complementing Cancer Metastasis. Front Immunol 2018; 9:1629. [PMID: 30061895 PMCID: PMC6054933 DOI: 10.3389/fimmu.2018.01629] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 07/02/2018] [Indexed: 12/21/2022] Open
Abstract
Complement is an effector of innate immunity and a bridge connecting innate immunity and subsequent adaptive immune responses. It is essential for protection against infections and for orchestrating inflammatory responses. Recent studies have also demonstrated contribution of the complement system to several homeostatic processes that are traditionally not considered to be involved in immunity. Thus, complement regulates homeostasis and immunity. However, dysregulation of this system contributes to several pathologies including inflammatory and autoimmune diseases. Unexpectedly, studies of the last decade have also revealed that complement promotes cancer progression. Since the initial discovery of tumor promoting role of complement, numerous preclinical and clinical studies demonstrated contribution of several complement components to regulation of tumor growth through their direct interactions with the corresponding receptors on tumor cells or through suppression of antitumor immunity. Most of this work, however, focused on a role of complement in regulating growth of primary tumors. Only recently, a few studies showed that complement promotes cancer metastasis through its contribution to epithelial-to-mesenchymal transition and the premetastatic niche. This latter work has shown that complement activation and generation of complement effectors including C5a occur in organs that are target for metastasis prior to arrival of the very first tumor cells. C5a through its interactions with C5a receptor 1 inhibits antitumor immunity by activating and recruiting immunosuppressive cells from the bone marrow to the premetastatic niche and by regulating function and self-renewal of pulmonary tissue-resident alveolar macrophages. These new advancements provide additional evidence for multifaceted functions of complement in cancer.
Collapse
Affiliation(s)
- Dawn M Kochanek
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, United States
| | - Shanawaz M Ghouse
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, United States
| | - Magdalena M Karbowniczek
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, United States
| | - Maciej M Markiewski
- Department of Immunotherapeutics and Biotechnology, School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, United States
| |
Collapse
|
25
|
Bareke H, Akbuga J. Complement system's role in cancer and its therapeutic potential in ovarian cancer. Scand J Immunol 2018; 88:e12672. [PMID: 29734524 DOI: 10.1111/sji.12672] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 04/30/2018] [Indexed: 12/31/2022]
Abstract
Cancer immunotherapy is a strong candidate for the long-awaited new edition to standard cancer therapies. For an effective immunotherapy, it is imperative to delineate the players of antitumour immune response. As an important innate immune system effector mechanism, complement is highly likely to play a substantial role in cancer immunity. Studies suggest that there may be two different "states of complement" that show opposing effects on cancer cells; a complement profile that has antitumour effects with low expression of membrane-bound complement regulator proteins (mCRPs), lytic membrane attack complex (MAC) concentration and moderate C5a concentration, and a complement profile that has protumour effects with high expression of mCRPs, sublytic MAC and high concentrations of C5a. One of the cancers that urgently require innovative therapeutic approaches is ovarian cancer, and complement has a potential to be a good target for this purpose. A combinatorial approach where the complement cascade is fine-tuned by inhibiting some of its activities while promoting the others can prove to be a fruitful approach. Herein, we will briefly discuss the cancer-immune system interaction and then present a discussion of complement system's role in tumour immunity and its therapeutic potential for ovarian cancer immunotherapy.
Collapse
Affiliation(s)
- H Bareke
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Marmara University, Istanbul, Turkey.,Faculty of Pharmacy, Girne American University, Kyrenia, North Cyprus, Turkey
| | - J Akbuga
- Faculty of Pharmacy, Girne American University, Kyrenia, North Cyprus, Turkey
| |
Collapse
|
26
|
Abstract
CD59 has been identified as a glycosylphosphatidylinositol-anchored membrane protein that acts as an inhibitor of the formation of the membrane attack complex to regulate complement activation. Recent studies have shown that CD59 is highly expressed in several cancer cell lines and tumor tissues. CD59 also regulates the function, infiltration and phenotypes of a variety of immune cells in the tumor microenvironment. Herein, we summarized recent advances related to the functions and mechanisms of CD59 in the tumor microenvironment. Therapeutic strategies that seek to modulate the functions of CD59 in the tumor microenvironment could be a promising direction for tumor immunotherapy.
Collapse
Affiliation(s)
- Ronghua Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, PR China
| | - Qiaofei Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, PR China
| | - Quan Liao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, PR China
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, PR China
| |
Collapse
|
27
|
Rozenberg P, Ziporen L, Gancz D, Saar-Ray M, Fishelson Z. Cooperation between Hsp90 and mortalin/GRP75 in resistance to cell death induced by complement C5b-9. Cell Death Dis 2018; 9:150. [PMID: 29396434 PMCID: PMC5833442 DOI: 10.1038/s41419-017-0240-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 12/14/2017] [Accepted: 12/18/2017] [Indexed: 12/14/2022]
Abstract
Cancer cells are commonly more resistant to cell death activated by the membranolytic protein complex C5b-9. Several surface-expressed and intracellular proteins that protect cells from complement-dependent cytotoxicity (CDC) have been identified. In this study, we investigated the function of heat shock protein 90 (Hsp90), an essential and ubiquitously expressed chaperone, overexpressed in cancer cells, in C5b-9-induced cell death. As shown, inhibition of Hsp90 with geldanamycin or radicicol is enhancing sensitivity of K562 erythroleukemia cells to CDC. Similarly, Hsp90 inhibition confers in Ramos B cell lymphoma cells elevated sensitivity to treatment with rituximab and complement. C5b-9 deposition is elevated on geldanamycin-treated cells. Purified Hsp90 binds directly to C9 and inhibits zinc-induced C9 polymerization, indicating that Hsp90 may act directly on the C5b-9 complex. Mortalin, also known as stress protein 70 or GRP75, is a mitochondrial chaperone that confers resistance to CDC. The postulated cooperation between Hsp90 and mortalin in protection from CDC was tested. Geldanamycin failed to sensitize toward CDC cells with knocked down mortalin. Direct binding of Hsp90 to mortalin was shown by co-immunoprecipitation in cell extracts after triggering with complement as well as by using purified recombinant proteins. These results provide an insight into the protective mechanisms utilized by cancer cells to evade CDC. They suggest that Hsp90 protects cells from CDC by inhibiting, together with mortalin, C5b-9 assembly and/or stability at the plasma membrane.
Collapse
Affiliation(s)
- Perri Rozenberg
- Department of Cell and Developmental Biology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Lea Ziporen
- Department of Cell and Developmental Biology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Dana Gancz
- Department of Cell and Developmental Biology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Moran Saar-Ray
- Department of Cell and Developmental Biology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Zvi Fishelson
- Department of Cell and Developmental Biology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel.
| |
Collapse
|
28
|
Fantini M, David JM, Saric O, Dubeykovskiy A, Cui Y, Mavroukakis SA, Bristol A, Annunziata CM, Tsang KY, Arlen PM. Preclinical Characterization of a Novel Monoclonal Antibody NEO-201 for the Treatment of Human Carcinomas. Front Immunol 2018; 8:1899. [PMID: 29354121 PMCID: PMC5758533 DOI: 10.3389/fimmu.2017.01899] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 12/12/2017] [Indexed: 12/27/2022] Open
Abstract
NEO-201 is a novel humanized IgG1 monoclonal antibody that was derived from an immunogenic preparation of tumor-associated antigens from pooled allogeneic colon tumor tissue extracts. It was found to react against a variety of cultured human carcinoma cell lines and was highly reactive against the majority of tumor tissues from many different carcinomas, including colon, pancreatic, stomach, lung, and breast cancers. NEO-201 also exhibited tumor specificity, as the majority of normal tissues were not recognized by this antibody. Functional assays revealed that treatment with NEO-201 is capable of mediating both antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) against tumor cells. Furthermore, the growth of human pancreatic xenograft tumors in vivo was largely attenuated by treatment with NEO-201 both alone and in combination with human peripheral blood mononuclear cells as an effector cell source for ADCC. In vivo biodistribution studies in human tumor xenograft-bearing mice revealed that NEO-201 preferentially accumulates in the tumor but not organ tissue. Finally, a single-dose toxicity study in non-human primates demonstrated safety and tolerability of NEO-201, as a transient decrease in circulating neutrophils was the only related adverse effect observed. These findings indicate that NEO-201 warrants clinical testing as both a novel diagnostic and therapeutic agent for the treatment of a broad variety of carcinomas.
Collapse
Affiliation(s)
| | | | - Olga Saric
- Precision Biologics, Inc., Rockville, MD, United States
| | | | - Yongzhi Cui
- Precision Biologics, Inc., Rockville, MD, United States
| | | | | | - Christina M Annunziata
- Women's Malignancy Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Kwong Y Tsang
- Precision Biologics, Inc., Rockville, MD, United States
| | | |
Collapse
|
29
|
Ajona D, Ortiz-Espinosa S, Pio R. Complement anaphylatoxins C3a and C5a: Emerging roles in cancer progression and treatment. Semin Cell Dev Biol 2017; 85:153-163. [PMID: 29155219 DOI: 10.1016/j.semcdb.2017.11.023] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 11/07/2017] [Accepted: 11/15/2017] [Indexed: 02/06/2023]
Abstract
Recent insights into the role of complement anaphylatoxins C3a and C5a in cancer provide new opportunities for the development of innovative biomarkers and therapeutic strategies. These two complement activation products can maintain chronic inflammation, promote an immunosuppressive microenvironment, induce angiogenesis, and increase the motility and metastatic potential of cancer cells. Still, the diverse heterogeneity of responses mediated by these peptides poses a challenge both to our understanding of the role played by these molecules in cancer progression and to the development of effective treatments. This review attempts to summarize the evidence surrounding the involvement of anaphylatoxins in the biological contexts associated with tumor progression. We also describe the recent developments that support the inhibition of anaphylatoxins, or their cognate receptors C3aR and C5aR1, as a treatment option for maximizing the clinical efficacy of current immunotherapies that target the PD-1/PD-L1 immune checkpoint.
Collapse
Affiliation(s)
- Daniel Ajona
- University of Navarra, Center for Applied Medical Research (CIMA), Program in Solid Tumors and Biomarkers, Pamplona, Spain; Navarra's Health Research Institute (IdiSNA), Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Spain; University of Navarra, School of Sciences, Department of Biochemistry and Genetics, Pamplona, Spain
| | - Sergio Ortiz-Espinosa
- University of Navarra, Center for Applied Medical Research (CIMA), Program in Solid Tumors and Biomarkers, Pamplona, Spain; University of Navarra, School of Sciences, Department of Biochemistry and Genetics, Pamplona, Spain
| | - Ruben Pio
- University of Navarra, Center for Applied Medical Research (CIMA), Program in Solid Tumors and Biomarkers, Pamplona, Spain; Navarra's Health Research Institute (IdiSNA), Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Spain; University of Navarra, School of Sciences, Department of Biochemistry and Genetics, Pamplona, Spain.
| |
Collapse
|
30
|
Englund E, Canesin G, Papadakos KS, Vishnu N, Persson E, Reitsma B, Anand A, Jacobsson L, Helczynski L, Mulder H, Bjartell A, Blom AM. Cartilage oligomeric matrix protein promotes prostate cancer progression by enhancing invasion and disrupting intracellular calcium homeostasis. Oncotarget 2017; 8:98298-98311. [PMID: 29228690 PMCID: PMC5716730 DOI: 10.18632/oncotarget.21176] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 08/27/2017] [Indexed: 12/30/2022] Open
Abstract
Cartilage oligomeric matrix protein (COMP) was recently implicated in the progression of breast cancer. Immunostaining of 342 prostate cancer specimens in tissue microarrays showed that COMP expression is not breast cancer-specific but also occurs in prostate cancer. The expression of COMP in prostate cancer cells correlated with a more aggressive disease with faster recurrence. Subcutaneous xenografts in immunodeficient mice showed that the prostate cancer cell line DU145 overexpressing COMP formed larger tumors in vivo as compared to mock-transfected cells. Purified COMP bound to and enhanced the invasion of DU145 cells in vitro in an integrin-dependent manner. In addition, intracellular COMP expression interfered with cellular metabolism by causing a decreased level of oxidative phosphorylation with a concurrent upregulation of lactate production (Warburg effect). Further, expression of COMP protected cells from induction of apoptosis via several pathways. The effect of COMP on metabolism and apoptosis induction was dependent on the ability of COMP to disrupt intracellular Ca2+ signalling by preventing Ca2+ release from the endoplasmic reticulum. In conclusion, COMP is a potent driver of the progression of prostate cancer, acting in an anti-apoptotic fashion by interfering with the Ca2+ homeostasis of cancer cells.
Collapse
Affiliation(s)
- Emelie Englund
- Department of Translational Medicine, Division of Medical Protein Chemistry, Lund University, Malmö, Sweden
| | - Giacomo Canesin
- Department of Translational Medicine, Division of Urological Cancers, Lund University, Malmö, Sweden
| | - Konstantinos S Papadakos
- Department of Translational Medicine, Division of Medical Protein Chemistry, Lund University, Malmö, Sweden
| | - Neelanjan Vishnu
- Department of Clinical Sciences Malmö, Unit of Molecular Metabolism, Lund University, Malmö, Sweden
| | - Emma Persson
- Department of Translational Medicine, Division of Medical Protein Chemistry, Lund University, Malmö, Sweden
| | - Bart Reitsma
- Department of Translational Medicine, Division of Medical Protein Chemistry, Lund University, Malmö, Sweden
| | - Aseem Anand
- Department of Translational Medicine, Division of Urological Cancers, Lund University, Malmö, Sweden
| | - Laila Jacobsson
- Department of Clinical Sciences Malmö, Unit of Molecular Metabolism, Lund University, Malmö, Sweden
| | - Leszek Helczynski
- Department of Translational Medicine, Division of Urological Cancers, Lund University, Malmö, Sweden
| | - Hindrik Mulder
- Department of Clinical Sciences Malmö, Unit of Molecular Metabolism, Lund University, Malmö, Sweden
| | - Anders Bjartell
- Department of Translational Medicine, Division of Urological Cancers, Lund University, Malmö, Sweden
| | - Anna M Blom
- Department of Translational Medicine, Division of Medical Protein Chemistry, Lund University, Malmö, Sweden
| |
Collapse
|
31
|
Prognostic Significance of Preoperative and Postoperative Complement C3 Depletion in Gastric Cancer: A Three-Year Survival Investigation. BIOMED RESEARCH INTERNATIONAL 2017; 2017:2161840. [PMID: 29062836 PMCID: PMC5618749 DOI: 10.1155/2017/2161840] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 08/07/2017] [Indexed: 12/14/2022]
Abstract
OBJECTIVES The role of complement system in predicting prognosis of gastric cancer (GC) remains obscured. This study aims to explore the incidence of complement C3 depletion and associated outcomes in GC patients. METHODS between August 2013 and December 2013, 106 patients with gastric adenocarcinoma were prospectively analyzed. Plasma levels of complement C3 and C4 were detected at baseline, one day before surgery, and postoperative day 3, respectively. Patients with low C3 levels (<0.75 mg/mL) were considered as having complement depletion (CD), while others with normal C3 levels were included as control. The 3-year overall survival (OS), disease-free survival (DFS), and other outcomes were compared between both groups, with the CD incidence explored meanwhile. RESULTS The CD incidence was 28.3% before surgery but increased to 37.7% after surgery. Preoperative CD was related to prolonged hospital stay (22.7 versus 19.2 day, P = 0.032) and increased postoperative complications (33.3% versus 14.5%, P = 0.030) and hospital costs (P = 0.013). Besides, postoperative C3 depletion was significantly associated with decreased 3-year OS (P = 0.022) and DFS (P = 0.003). Moreover, postoperative C3 depletion and advanced tumor stage were independent predictive factors of poor prognosis. CONCLUSIONS Complement C3 depletion occurring in gastric cancer was associated with poor short-term and long-term outcomes.
Collapse
|
32
|
Chockley PJ, Keshamouni VG. Immunological Consequences of Epithelial-Mesenchymal Transition in Tumor Progression. THE JOURNAL OF IMMUNOLOGY 2017; 197:691-8. [PMID: 27431984 DOI: 10.4049/jimmunol.1600458] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 05/18/2016] [Indexed: 12/26/2022]
Abstract
Microenvironments that tumor cells encounter are different during the stages of cancer progression-primary tumor, metastasis, and at the metastatic site. This suggests potential differences in immune surveillance of primary tumor and metastasis. Epithelial-mesenchymal transition (EMT) is a key reversible process in which cancer cells transition into highly motile and invasive cells for dissemination. Only a tiny proportion successfully metastasize, supporting the notion of metastasis-specific immune surveillance. EMT involves extensive molecular reprogramming of cells conferring many clinically relevant features to cancer cells and affects tumor cell interactions within the tumor microenvironment. We review the impact of tumor immune infiltrates on tumor cell EMT and the consequences of EMT in shaping the immune microenvironment of tumors. The usefulness of EMT as a model to investigate metastasis-specific immune surveillance mechanisms are also explored. Finally, we discuss potential implications of EMT for tumor immunogenicity, as well as current immunotherapies and future strategies.
Collapse
Affiliation(s)
- Peter J Chockley
- Graduate Program in Immunology, University of Michigan Medical Center, Ann Arbor, MI 48109; and
| | - Venkateshwar G Keshamouni
- Graduate Program in Immunology, University of Michigan Medical Center, Ann Arbor, MI 48109; and Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI 48109
| |
Collapse
|
33
|
Granados-Durán P, López-Ávalos MD, Cifuentes M, Pérez-Martín M, Fernández-Arjona MDM, Hughes TR, Johnson K, Morgan BP, Fernández-Llebrez P, Grondona JM. Microbial Neuraminidase Induces a Moderate and Transient Myelin Vacuolation Independent of Complement System Activation. Front Neurol 2017; 8:78. [PMID: 28326060 PMCID: PMC5339270 DOI: 10.3389/fneur.2017.00078] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 02/20/2017] [Indexed: 02/05/2023] Open
Abstract
AIMS Some central nervous system pathogens express neuraminidase (NA) on their surfaces. In the rat brain, a single intracerebroventricular (ICV) injection of NA induces myelin vacuolation in axonal tracts. Here, we explore the nature, the time course, and the role of the complement system in this damage. METHODS The spatiotemporal analysis of myelin vacuolation was performed by optical and electron microscopy. Myelin basic protein-positive area and oligodendrocyte transcription factor (Olig2)-positive cells were quantified in the damaged bundles. Neuronal death in the affected axonal tracts was assessed by Fluoro-Jade B and anti-caspase-3 staining. To evaluate the role of the complement, membrane attack complex (MAC) deposition on damaged bundles was analyzed using anti-C5b9. Rats ICV injected with the anaphylatoxin C5a were studied for myelin damage. In addition, NA-induced vacuolation was studied in rats with different degrees of complement inhibition: normal rats treated with anti-C5-blocking antibody and C6-deficient rats. RESULTS The stria medullaris, the optic chiasm, and the fimbria were the most consistently damaged axonal tracts. Vacuolation peaked 7 days after NA injection and reverted by day 15. Olig2+ cell number in the damaged tracts was unaltered, and neurodegeneration associated with myelin alterations was not detected. MAC was absent on damaged axonal tracts, as revealed by C5b9 immunostaining. Rats ICV injected with the anaphylatoxin C5a displayed no myelin injury. When the complement system was experimentally or constitutively inhibited, NA-induced myelin vacuolation was similar to that observed in normal rats. CONCLUSION Microbial NA induces a moderate and transient myelin vacuolation that is not caused either by neuroinflammation or complement system activation.
Collapse
Affiliation(s)
- Pablo Granados-Durán
- Laboratorio de Fisiología Animal, Facultad de Ciencias, Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga , Málaga , Spain
| | - María Dolores López-Ávalos
- Laboratorio de Fisiología Animal, Facultad de Ciencias, Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga , Málaga , Spain
| | - Manuel Cifuentes
- Laboratorio de Fisiología Animal, Facultad de Ciencias, Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain; Centro de Investigaciones Biomédicas en Red de Bioingeniería, Biomateriales y Nanomedicina, CIBER BBN, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
| | - Margarita Pérez-Martín
- Laboratorio de Fisiología Animal, Facultad de Ciencias, Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga , Málaga , Spain
| | - María Del Mar Fernández-Arjona
- Laboratorio de Fisiología Animal, Facultad de Ciencias, Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga , Málaga , Spain
| | - Timothy R Hughes
- Division of Infection and Immunity, School of Medicine, Cardiff University , Cardiff , UK
| | | | - B Paul Morgan
- Division of Infection and Immunity, School of Medicine, Cardiff University , Cardiff , UK
| | - Pedro Fernández-Llebrez
- Laboratorio de Fisiología Animal, Facultad de Ciencias, Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga , Málaga , Spain
| | - Jesús M Grondona
- Laboratorio de Fisiología Animal, Facultad de Ciencias, Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga , Málaga , Spain
| |
Collapse
|
34
|
Abstract
In addition to being a component of innate immunity and an ancient defense mechanism against invading pathogens, complement activation also participates in the adaptive immune response, inflammation, hemostasis, embryogenesis, and organ repair and development. Activation of the complement system via classical, lectin, or alternative pathways generates anaphylatoxins (C3a and C5a) and membrane attack complex (C5b-9) and opsonizes targeted cells. Complement activation end products and their receptors mediate cell-cell interactions that regulate several biological functions in the extravascular tissue. Signaling of anaphylatoxin receptors or assembly of membrane attack complex promotes cell dedifferentiation, proliferation, and migration in addition to reducing apoptosis. As a result, complement activation in the tumor microenvironment enhances tumor growth and increases metastasis. In this Review, I discuss immune and nonimmune functions of complement proteins and the tumor-promoting effect of complement activation.
Collapse
|
35
|
Granados-Durán P, López-Ávalos MD, Hughes TR, Johnson K, Morgan BP, Tamburini PP, Fernández-Llebrez P, Grondona JM. Complement system activation contributes to the ependymal damage induced by microbial neuraminidase. J Neuroinflammation 2016; 13:115. [PMID: 27209022 PMCID: PMC4875702 DOI: 10.1186/s12974-016-0576-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 05/09/2016] [Indexed: 01/18/2023] Open
Abstract
Background In the rat brain, a single intracerebroventricular injection of neuraminidase from Clostridium perfringens induces ependymal detachment and death. This injury occurs before the infiltration of inflammatory blood cells; some reports implicate the complement system as a cause of these injuries. Here, we set out to test the role of complement. Methods The assembly of the complement membrane attack complex on the ependymal epithelium of rats injected with neuraminidase was analyzed by immunohistochemistry. Complement activation, triggered by neuraminidase, and the participation of different activation pathways were analyzed by Western blot. In vitro studies used primary cultures of ependymal cells and explants of the septal ventricular wall. In these models, ependymal cells were exposed to neuraminidase in the presence or absence of complement, and their viability was assessed by observing beating of cilia or by trypan blue staining. The role of complement in ependymal damage induced by neuraminidase was analyzed in vivo in two rat models of complement blockade: systemic inhibition of C5 by using a function blocking antibody and testing in C6-deficient rats. Results The complement membrane attack complex immunolocalized on the ependymal surface in rats injected intracerebroventricularly with neuraminidase. C3 activation fragments were found in serum and cerebrospinal fluid of rats treated with neuraminidase, suggesting that neuraminidase itself activates complement. In ventricular wall explants and isolated ependymal cells, treatment with neuraminidase alone induced ependymal cell death; however, the addition of complement caused increased cell death and disorganization of the ependymal epithelium. In rats treated with anti-C5 and in C6-deficient rats, intracerebroventricular injection of neuraminidase provoked reduced ependymal alterations compared to non-treated or control rats. Immunohistochemistry confirmed the absence of membrane attack complex on the ependymal surfaces of neuraminidase-exposed rats treated with anti-C5 or deficient in C6. Conclusions These results demonstrate that the complement system contributes to ependymal damage and death caused by neuraminidase. However, neuraminidase alone can induce moderate ependymal damage without the aid of complement.
Collapse
Affiliation(s)
- Pablo Granados-Durán
- Departamento de Biología Celular, Genética y Fisiología, IBIMA, Facultad de Ciencias, Universidad de Málaga, Málaga, 29071, Spain
| | - María Dolores López-Ávalos
- Departamento de Biología Celular, Genética y Fisiología, IBIMA, Facultad de Ciencias, Universidad de Málaga, Málaga, 29071, Spain
| | - Timothy R Hughes
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Krista Johnson
- Alexion Pharmaceuticals Inc., 352 Knotter Drive, Cheshire, CT, 06410, USA
| | - B Paul Morgan
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Paul P Tamburini
- Alexion Pharmaceuticals Inc., 352 Knotter Drive, Cheshire, CT, 06410, USA
| | - Pedro Fernández-Llebrez
- Departamento de Biología Celular, Genética y Fisiología, IBIMA, Facultad de Ciencias, Universidad de Málaga, Málaga, 29071, Spain
| | - Jesús M Grondona
- Departamento de Biología Celular, Genética y Fisiología, IBIMA, Facultad de Ciencias, Universidad de Málaga, Málaga, 29071, Spain.
| |
Collapse
|
36
|
Zhang X, Chen B, He M, Zhang Y, Peng L, Hu B. Boronic acid recognition based-gold nanoparticle-labeling strategy for the assay of sialic acid expression on cancer cell surface by inductively coupled plasma mass spectrometry. Analyst 2016; 141:1286-93. [DOI: 10.1039/c5an02402a] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Sialic acids are special sugars widely expressed at the termini of glycan chains on the cell surface, and their expression level on the cancer cell surface is much higher than on the normal cell surface.
Collapse
Affiliation(s)
- Xing Zhang
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education)
- Department of Chemistry
- Wuhan University
- Wuhan 430072
- P R China
| | - Beibei Chen
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education)
- Department of Chemistry
- Wuhan University
- Wuhan 430072
- P R China
| | - Man He
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education)
- Department of Chemistry
- Wuhan University
- Wuhan 430072
- P R China
| | - Yuan Zhang
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education)
- Department of Chemistry
- Wuhan University
- Wuhan 430072
- P R China
| | - Lu Peng
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education)
- Department of Chemistry
- Wuhan University
- Wuhan 430072
- P R China
| | - Bin Hu
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education)
- Department of Chemistry
- Wuhan University
- Wuhan 430072
- P R China
| |
Collapse
|
37
|
Complement factor H polymorphism rs1061170 and the effect of cigarette smoking on the risk of lung cancer. Contemp Oncol (Pozn) 2015; 19:441-5. [PMID: 26843839 PMCID: PMC4731447 DOI: 10.5114/wo.2015.56202] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 10/15/2015] [Indexed: 01/02/2023] Open
Abstract
Aim of the study Complement factor H (CFH) has been known to inhibit the complement pathway and to contribute to tumour growth by suppressing the anti-tumour cell mediated response in cell lines from several malignancies. We examined the association of Try402His single nucleotide polymorphism in CFH gene with lung cancer and the interaction with cigarette smoking. Material and methods This case-control study included 80 primary lung cancer patients and 106 control subjects who were genotyped for Try402His (rs1061170) by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) analysis. Results Variant genotypes (Tyr/His and His/His) were overpresented among patients compared to controls (p = 0.03, OR = 2.510, 95% CI: 1.068–5.899), and the frequency of variant H allele was significantly overexpressed in cases compared to controls (p = 0.021). Tyr/His genotype was identified in 100% of small cell lung cancer (SCLC) patients vs. 34.5% of non-SCLC (NSCLC), while 20.7% of NSCLC patients were homozygous for the variant allele (His/His) (p = 0.001). Binary logistic regression analysis revealed a 2.5 times greater estimated risk for NSCLC than for SCLC among variant allele carriers, and a 7.3-fold increased risk of lung cancer among variant allele smoking carriers vs. 1.3-fold increased risk among wild allele smoking carriers. Moreover, the stage of cancer positively correlated with smoking and pack-years in allele H carriers, and the correlation was stronger among those who were homozygous for it (His/His) than those who were heterozygous (Tyr/His). Conclusions CFH 402H variant is a smoking-related risk factor for lung cancer, particularly the NSCLC.
Collapse
|
38
|
Mamidi S, Höne S, Kirschfink M. The complement system in cancer: Ambivalence between tumour destruction and promotion. Immunobiology 2015; 222:45-54. [PMID: 26686908 DOI: 10.1016/j.imbio.2015.11.008] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 10/08/2015] [Accepted: 11/19/2015] [Indexed: 12/14/2022]
Abstract
Constituting a part of the innate immune system, the complement system consists of over 50 proteins either acting as part of a 3-branch activation cascade, a well-differentiated regulatory system in fluid phase or on each tissue, or as receptors translating the activation signal to multiple cellular effector functions. Complement serves as first line of defence against infections from bacteria, viruses and parasites by orchestrating the immune response through opsonisation, recruitment of immune cells to the site of infection and direct cell lysis. Complement is generally recognised as a protective mechanism against the formation of tumours in humans, but is often limited by various resistance mechanisms interfering with its cytotoxic action, now considered as a great barrier of successful antibody-based immunotherapy. However, recent studies also indicate a pro-tumourigenic potential of complement in certain cancers and under certain conditions. In this review, we present recent findings on the possible dual role of complement in destroying cancer, especially if resistance mechanisms are blocked, but also under certain inflammatory conditions-promoting tumour development.
Collapse
Affiliation(s)
| | - Simon Höne
- Institute for Immunology, University of Heidelberg, Germany
| | | |
Collapse
|
39
|
Englund E, Reitsma B, King BC, Escudero-Esparza A, Owen S, Orimo A, Okroj M, Anagnostaki L, Jiang WG, Jirström K, Blom AM. The human complement inhibitor Sushi Domain-Containing Protein 4 (SUSD4) expression in tumor cells and infiltrating T cells is associated with better prognosis of breast cancer patients. BMC Cancer 2015; 15:737. [PMID: 26480818 PMCID: PMC4615997 DOI: 10.1186/s12885-015-1734-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 10/08/2015] [Indexed: 01/22/2023] Open
Abstract
Background The human Sushi Domain-Containing Protein 4 (SUSD4) was recently shown to function as a novel inhibitor of the complement system, but its role in tumor progression is unknown. Methods Using immunohistochemistry and quantitative PCR, we investigated SUSD4 expression in breast cancer tissue samples from two cohorts. The effect of SUSD4 expression on cell migration and invasion was studied in vitro using two human breast cancer cell lines overexpressing SUSD4. Results Tissue stainings revealed that both tumor cells and tumor-infiltrating cells expressed SUSD4. The highest SUSD4 expression was detected in differentiated tumors with decreased rate of metastasis, and SUSD4 expression was associated with improved survival of the patients. Moreover, forced SUSD4 expression in human breast cancer cells attenuated their migratory and invasive traits in culture. SUSD4 expression also inhibited colony formation of human breast cancer cells cultured on carcinoma-associated fibroblasts. Furthermore, large numbers of SUSD4-expressing T cells in the tumor stroma associated with better overall survival of the breast cancer patients. Conclusion Our findings indicate that SUSD4 expression in both breast cancer cells and T cells infiltrating the tumor-associated stroma is useful to predict better prognosis of breast cancer patients.
Collapse
Affiliation(s)
- Emelie Englund
- Department of Translational Medicine, Division of Medical Protein Chemistry, Lund University, Inga Maria Nilssons gata 53, 20502, Malmö, Sweden.
| | - Bart Reitsma
- Department of Translational Medicine, Division of Medical Protein Chemistry, Lund University, Inga Maria Nilssons gata 53, 20502, Malmö, Sweden.
| | - Ben C King
- Department of Translational Medicine, Division of Medical Protein Chemistry, Lund University, Inga Maria Nilssons gata 53, 20502, Malmö, Sweden.
| | - Astrid Escudero-Esparza
- Department of Translational Medicine, Division of Medical Protein Chemistry, Lund University, Inga Maria Nilssons gata 53, 20502, Malmö, Sweden.
| | - Sioned Owen
- Cardiff's China Medical Research Collaborative (CCMRC), Cardiff University School of Medicine, Cardiff University, Cardiff, UK.
| | - Akira Orimo
- Department of Pathology and Oncology, Juntendo University School of Medicine, Tokyo, Japan.
| | - Marcin Okroj
- Department of Translational Medicine, Division of Medical Protein Chemistry, Lund University, Inga Maria Nilssons gata 53, 20502, Malmö, Sweden. .,Department of Medical Biotechnology, Intercollegiate Faculty of Biotechnology UG‑MUG, Medical University of Gdańsk, 80210, Gdańsk, Poland.
| | - Lola Anagnostaki
- Department of Clinical Pathology, Skåne University Hospital, Malmö, Sweden.
| | - Wen G Jiang
- Cardiff's China Medical Research Collaborative (CCMRC), Cardiff University School of Medicine, Cardiff University, Cardiff, UK.
| | - Karin Jirström
- Department of Clinical Sciences Lund, Division of Oncology and Pathology, Lund University, Lund, Sweden.
| | - Anna M Blom
- Department of Translational Medicine, Division of Medical Protein Chemistry, Lund University, Inga Maria Nilssons gata 53, 20502, Malmö, Sweden.
| |
Collapse
|
40
|
The immunohistochemical analysis of membrane-bound CD55, CD59 and fluid-phase FH and FH-like complement inhibitors in cancers of ovary and corpus uteri origin. Cent Eur J Immunol 2015; 40:349-53. [PMID: 26648780 PMCID: PMC4655386 DOI: 10.5114/ceji.2015.54598] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 05/15/2015] [Indexed: 12/19/2022] Open
Abstract
One of the potential therapeutic methods of cancer treatment is the immunotherapy with monoclonal antibodies. This kind of therapy, although devoid of serious side effects, has often insufficient efficacy. The presence of complement inhibitors on the cancer cells, which are able to inactivate complement-mediated immune response represents one of the main reasons for the inefficiency of such therapy. In our studies we investigated the expression of main membrane–bound and fluid-phase complement regulators: CD55, CD59 and factor H/factor H-like in tumour samples of ovarian and corpus uteri cancer. Tissue samples were collected from 50 patients and stained immunohistochemically, with the use of peroxidase-based immunodetection system. Immunohistochemical analysis revealed that complement inhibitors are present in examined tumors although their presence is heterogenous. The most prevalent is the presence of factor H/H-like, localized mostly in tumor stroma and within vascular structures. Membrane bound complement inhibitors are less prominently expressed by cancer cells. CD55 was detected in low percentage of cells, predominantly within cancer tubules. CD59 immunoreactivity was more prevalent in cancer cells, and was localized particularly at the margin of cancer cell tubules. Our results demonstrate that the most prominent complement inhibitor in cancer of ovary and corpus uteri origin is factor H/factor H-like. Blocking or downregulation of this inhibitor should be taken into consideration with regards to improving the efficiency of immunotherapy with monoclonal antibodies.
Collapse
|
41
|
Chronic Activation of Innate Immunity Correlates With Poor Prognosis in Cancer Patients Treated With Oncolytic Adenovirus. Mol Ther 2015; 24:175-83. [PMID: 26310629 DOI: 10.1038/mt.2015.143] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 07/23/2015] [Indexed: 12/17/2022] Open
Abstract
Despite many clinical trials conducted with oncolytic viruses, the exact tumor-level mechanisms affecting therapeutic efficacy have not been established. Currently there are no biomarkers available that would predict the clinical outcome to any oncolytic virus. To assess the baseline immunological phenotype and find potential prognostic biomarkers, we monitored mRNA expression levels in 31 tumor biopsy or fluid samples from 27 patients treated with oncolytic adenovirus. Additionally, protein expression was studied from 19 biopsies using immunohistochemical staining. We found highly significant changes in several signaling pathways and genes associated with immune responses, such as B-cell receptor signaling (P < 0.001), granulocyte macrophage colony-stimulating factor (GM-CSF) signaling (P < 0.001), and leukocyte extravasation signaling (P < 0.001), in patients surviving a shorter time than their controls. In immunohistochemical analysis, markers CD4 and CD163 were significantly elevated (P = 0.020 and P = 0.016 respectively), in patients with shorter than expected survival. Interestingly, T-cell exhaustion marker TIM-3 was also found to be significantly upregulated (P = 0.006) in patients with poor prognosis. Collectively, these data suggest that activation of several functions of the innate immunity before treatment is associated with inferior survival in patients treated with oncolytic adenovirus. Conversely, lack of chronic innate inflammation at baseline may predict improved treatment outcome, as suggested by good overall prognosis.
Collapse
|
42
|
Kapka-Skrzypczak L, Wolinska E, Szparecki G, Wilczynski GM, Czajka M, Skrzypczak M. CD55, CD59, factor H and factor H-like 1 gene expression analysis in tumors of the ovary and corpus uteri origin. Immunol Lett 2015; 167:67-71. [PMID: 26261870 DOI: 10.1016/j.imlet.2015.06.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 06/24/2015] [Accepted: 06/29/2015] [Indexed: 12/15/2022]
Abstract
The expression level of complement regulators in ovarian and corpus uteri tumors was not fully established so far. In current manuscript we performed gene expression analysis by the real-time PCR approach to investigate both membrane bound - CD55 and CD59 and fluid phase - factor H and factor H-like 1 complement regulators. We found increased CD55 expression in corpus uteri tumors when compared to control tissues, whereas in ovarian cancer CD55 expression was lower than in control sections. Additionally we found CD59 expression to be more prominent in ovarian cancer than in corpus uteri tumor samples. We observed also the strong positive correlation between the level of expression of the whole group of regulators, which was particularly significant between the expression of factor H and factor H- like 1. In conclusion we present novel results which implicates different role of particular complement inhibitors in the regulation of the complement system in two cancer types examined. Strong positive correlation between examined proteins implicates similar pattern of the regulation which should be taken into consideration with regards to the possible immunotherapy applied as adjuvant therapeutic approach in these two indications. The inhibition of complement regulation may serve as a strategy to potentiate the efficacy of such treatment.
Collapse
Affiliation(s)
- L Kapka-Skrzypczak
- Department of Medical Biology and Translational Research, University of Information Technology and Management, Faculty of Medicine, Sucharskiego Street 2, 35-225 Rzeszow, Poland; Department of Molecular Biology and Translational Research, Institute of Rural Health, Jaczewskiego Street 2, 20-090 Lublin, Poland
| | - E Wolinska
- Department of Pathology, Medical University of Warsaw, Pawinskiego Street 7, 02-091 Warsaw, Poland.
| | - G Szparecki
- Department of Pathology, Medical University of Warsaw, Pawinskiego Street 7, 02-091 Warsaw, Poland
| | - G M Wilczynski
- Laboratory of Molecular and Systemic Neuromorphology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura Street 3, 02-091 Warsaw, Poland
| | - M Czajka
- Department of Molecular Biology and Translational Research, Institute of Rural Health, Jaczewskiego Street 2, 20-090 Lublin, Poland
| | - M Skrzypczak
- Second Department of Gynecology, Medical University of Lublin, Jaczewskiego Street 8, 20-954 Lublin, Poland
| |
Collapse
|
43
|
Goswami MT, Reka AK, Kurapati H, Kaza V, Chen J, Standiford TJ, Keshamouni VG. Regulation of complement-dependent cytotoxicity by TGF-β-induced epithelial-mesenchymal transition. Oncogene 2015; 35:1888-98. [PMID: 26148233 PMCID: PMC4703565 DOI: 10.1038/onc.2015.258] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 04/08/2015] [Accepted: 04/24/2015] [Indexed: 12/23/2022]
Abstract
The process of Epithelial-mesenchymal transition (EMT), in addition to being an initiating event for tumor metastasis, is implicated in conferring several clinically relevant properties to disseminating cancer cells. These include stem cell like properties, resistance to targeted therapies and ability to evade immune surveillance. Enrichment analysis of gene expression changes during TGF-β induced EMT in lung cancer cells identified complement cascade as one of the significantly enriched pathway. Further analysis of the genes in the complement pathway revealed an increase in the expression of complement inhibitors and a decrease in the expression of proteins essential for complement activity. In this study, we tested whether EMT confers resistance to complement-dependent cytotoxicity (CDC) in lung cancer cells and promotes tumor progression. CD59 is a potent inhibitor of membrane attack complex that mediates complement-dependent cell lysis. We observed a significant increase in the CD59 expression on the surface of cells after TGF-β-induced EMT. Furthermore, CD59 knock down restored susceptibility of cells undergoing EMT to Cetuximab-mediated CDC. TGF-β-induced CD59 expression during EMT is dependent on Smad3 but not Smad2. ChIP analysis confirmed that Smad3 directly binds to the CD59 promoter. Stable knock-down of CD59 in A549 cells inhibited experimental metastasis. These results demonstrate that TGF-β-induced EMT and CD59 expression confers an immune evasive mechanism to disseminating tumor cells facilitating tumor progression. Together, our data demonstrates that CD59 inhibition may serve as an adjuvant to enhance the efficacy of antibody-mediated therapies, as well as to inhibit metastasis in lung cancer.
Collapse
Affiliation(s)
- M T Goswami
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - A K Reka
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - H Kurapati
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - V Kaza
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - J Chen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - T J Standiford
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - V G Keshamouni
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| |
Collapse
|
44
|
Song Q, Zhang Z, Liu Y, Han S, Zhang X. The tag SNP rs10746463 in decay-accelerating factor is associated with the susceptibility to gastric cancer. Mol Immunol 2015; 63:473-8. [PMID: 25457880 DOI: 10.1016/j.molimm.2014.10.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 09/29/2014] [Accepted: 10/03/2014] [Indexed: 01/25/2023]
Abstract
BACKGROUND Complement activation involved in the innate immunity and adaptive immunity and further contributed to the development of tumor growth. This study aimed to investigate the association of genetic variants in complement 3 (C3) and decay-accelerating factor (DAF) genes with the risk of gastric cancer. METHODS This case-control study included 500 gastric cancer patients and 500 cancer-free controls. Based on the Chinese population data from HapMap database, we used Haploview 4.2 program to select candidate tag SNPs. Odds ratios (ORs) and 95% confidence intervals (CIs) were calculated by logistic regression to evaluate the association of each genetic variant with the risk of gastric cancer. RESULTS Among 12 tag SNPs of C3, no correlation was observed between C3 genetic variants and risk of gastric cancer. For tag SNPS of DAF, logistic regression analysis revealed that the carriers with DAF rs10746463 AA genotype had a significantly increased risk for developing gastric cancer (OR = 1.46, 95% CI = 1.01–2.10) when compared with GG genotype, but those carrying with rs10746463 AG genotype didn't (OR = 1.31, 95% CI = 0.98-1.75). When stratified by smoking status, we found that the risk of gastric cancer was associated with rs10746463 GA or AA genotype carriers among smoker with OR (95% CI) of 1.64 (1.06-2.54), but not among non-smoker (OR = 1.37, 95% CI = 0.97-1.94). CONCLUSION DAF rs10746463 polymorphism effects on the risk of developing gastric cancer in Chinese population.
Collapse
Affiliation(s)
- Qinqin Song
- Institute of Molecular Genetics, College of Life Science, Hebei United University, Tangshan, China
| | | | | | | | | |
Collapse
|
45
|
Langford-Smith A, Day AJ, Bishop PN, Clark SJ. Complementing the Sugar Code: Role of GAGs and Sialic Acid in Complement Regulation. Front Immunol 2015; 6:25. [PMID: 25699044 PMCID: PMC4313701 DOI: 10.3389/fimmu.2015.00025] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 01/12/2015] [Indexed: 01/15/2023] Open
Abstract
Sugar molecules play a vital role on both microbial and mammalian cells, where they are involved in cellular communication, govern microbial virulence, and modulate host immunity and inflammatory responses. The complement cascade, as part of a host's innate immune system, is a potent weapon against invading bacteria but has to be tightly regulated to prevent inappropriate attack and damage to host tissues. A number of complement regulators, such as factor H and properdin, interact with sugar molecules, such as glycosaminoglycans (GAGs) and sialic acid, on host and pathogen membranes and direct the appropriate complement response by either promoting the binding of complement activators or inhibitors. The binding of these complement regulators to sugar molecules can vary from location to location, due to their different specificities and because distinct structural and functional subpopulations of sugars are found in different human organs, such as the brain, kidney, and eye. This review will cover recent studies that have provided important new insights into the role of GAGs and sialic acid in complement regulation and how sugar recognition may be compromised in disease.
Collapse
Affiliation(s)
- Alex Langford-Smith
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester , Manchester , UK
| | - Anthony J Day
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester , Manchester , UK
| | - Paul N Bishop
- Centre for Hearing and Vision Research, Institute of Human Development, University of Manchester , Manchester , UK ; Centre for Advanced Discovery and Experimental Therapeutics, University of Manchester and Central Manchester University Hospitals NHS Foundation Trust , Manchester , UK ; Manchester Academic Health Science Centre, University of Manchester and Central Manchester University Hospitals NHS Foundation Trust , Manchester , UK ; Manchester Royal Eye Hospital, Central Manchester University Hospitals NHS Foundation Trust , Manchester , UK
| | - Simon J Clark
- Centre for Hearing and Vision Research, Institute of Human Development, University of Manchester , Manchester , UK ; Centre for Advanced Discovery and Experimental Therapeutics, University of Manchester and Central Manchester University Hospitals NHS Foundation Trust , Manchester , UK
| |
Collapse
|
46
|
Targeted delivery of siRNA using transferrin-coupled lipoplexes specifically sensitizes CD71 high expressing malignant cells to antibody-mediated complement attack. Target Oncol 2014; 10:405-13. [DOI: 10.1007/s11523-014-0345-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 10/29/2014] [Indexed: 01/08/2023]
|
47
|
Meyer S, Leusen JHW, Boross P. Regulation of complement and modulation of its activity in monoclonal antibody therapy of cancer. MAbs 2014; 6:1133-44. [PMID: 25517299 PMCID: PMC4622586 DOI: 10.4161/mabs.29670] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The complement system is a powerful tool of the innate immune system to eradicate pathogens. Both in vitro and in vivo evidence indicates that therapeutic anti-tumor monoclonal antibodies (mAbs) can activate the complement system by the classical pathway. However, the contribution of complement to the efficacy of mAbs is still debated, mainly due to the lack of convincing data in patients. A beneficial role for complement during mAb therapy is supported by the fact that cancer cells often upregulate complement-regulatory proteins (CRPs). Polymorphisms in various CRPs were previously associated with complement-mediated disorders. In this review the role of complement in anti-tumor mAb therapy will be discussed with special emphasis on strategies aiming at modifying complement activity. In the future, clinical efficacy of mAbs with enhanced effector functions together with comprehensive analysis of polymorphisms in CRPs in mAb-treated patients will further clarify the role of complement in mAb therapy.
Collapse
Affiliation(s)
- Saskia Meyer
- a Laboratory for Immunotherapy; Laboratory for Translational Immunology (LTI) ; University Medical Center Utrecht ; Utrecht , The Netherlands
| | | | | |
Collapse
|
48
|
Büll C, den Brok MH, Adema GJ. Sweet escape: sialic acids in tumor immune evasion. Biochim Biophys Acta Rev Cancer 2014; 1846:238-46. [PMID: 25026312 DOI: 10.1016/j.bbcan.2014.07.005] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 07/07/2014] [Accepted: 07/08/2014] [Indexed: 12/24/2022]
Abstract
Sialic acids represent a family of sugar molecules derived from neuraminic acid that frequently terminate glycan chains and contribute to many biological processes. Already five decades ago, aberrantly high expression of sialic acids has been proposed to protect cancer cells from recognition and eradication by the immune system. Today, increased understanding at the molecular level demonstrates the broad immunomodulatory capacity of tumor-derived sialic acids that is, at least in part, mediated through interactions with immunoinhibitory Siglec receptors. Here we will review current studies from a sialic acid sugar perspective showing that tumor-derived sialic acids disable major killing mechanisms of effector immune cells, trigger production of immune suppressive cytokines and dampen activation of antigen-presenting cells and subsequent induction of anti-tumor immune responses. Furthermore, strategies to modulate sialic acid expression in cancer cells to improve cancer immunotherapy will be discussed.
Collapse
Affiliation(s)
- Christian Büll
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Martijn H den Brok
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Gosse J Adema
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, The Netherlands.
| |
Collapse
|
49
|
LIN KAILONG, HE SIYI, HE LUHANG, CHEN JUNYIN, CHENG XIAOMING, ZHANG GUOQIANG, ZHU BO. Complement component 3 is a prognostic factor of non-small cell lung cancer. Mol Med Rep 2014; 10:811-7. [DOI: 10.3892/mmr.2014.2230] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 03/27/2014] [Indexed: 11/05/2022] Open
|
50
|
Yang P, Baciu P, Kerrigan BCP, Etheridge M, Sung E, Toimil BA, Berchuck JE, Jaffe GJ. Retinal pigment epithelial cell death by the alternative complement cascade: role of membrane regulatory proteins, calcium, PKC, and oxidative stress. Invest Ophthalmol Vis Sci 2014; 55:3012-21. [PMID: 24677108 DOI: 10.1167/iovs.13-13554] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
PURPOSE Retinal pigment epithelial (RPE) cell death is an important feature of the advanced forms of AMD. Complement alternative pathway (AP) activation is associated with RPE cell death in AMD. In this study, we developed a new model to initiate AP activation on RPE cells and investigated the cellular mechanisms modulating AP activation-mediated RPE cell death. METHODS An anti-RPE antibody was developed. A spontaneously arising human RPE cell line (ARPE-19) and donor RPE cells were primed with this antibody followed by stimulation with 6% C1q-depleted human serum (C1q-Dep) to activate AP. Complement activation was evaluated by flow cytometry and immunofluorescent staining. Cellular response to complement activation was examined by measurement of intracellular calcium and adenosine triphosphate (ATP) release. Cell viability was assessed by Sytox orange, tetrazolium salt, and lactate dehydrogenase release assays. RESULTS Alternative pathway complement-mediated RPE cell death was associated with membrane attack complex formation and a rapid rise in intracellular calcium followed by release of ATP. Downregulation of membrane complement regulatory proteins and protein kinase C (PKC) inhibition increased cell susceptibility to complement attack. Pretreatment of RPE cells with either hydrogen peroxide or hydroquinone enhanced cell death. Chronic repetitive treatment of RPE cells with low levels of oxidants also enhanced complement-mediated cell death. CONCLUSIONS Activation of complement through the alternative pathway induces sublytic and lytic phases of complement attack on RPE cells, leading to cell death modulated by extracellular calcium, membrane complement regulatory proteins, and intracellular signaling mechanisms. Single-dose oxidant exposure and low-dose repetitive oxidant exposure rendered RPE cells more susceptible to complement-mediated death.
Collapse
Affiliation(s)
- Ping Yang
- Department of Ophthalmology, Duke University Medical Center, Durham, North Carolina, United States
| | | | | | | | | | | | | | | |
Collapse
|