1
|
Aggarwal RR, Vuky J, VanderWeele D, Rettig M, Heath EI, Quigley D, Huang J, Chumber A, Cheung A, Foye A, Leung S, Abbey J, Dorr A, Nasoff M, Hunter J, Wang S, Flavell RR, Fong L, Liu B, Small EJ. Phase I, First-in-Human Study of FOR46 (FG-3246), an Immune-Modulating Antibody-Drug Conjugate Targeting CD46, in Patients With Metastatic Castration-Resistant Prostate Cancer. J Clin Oncol 2025; 43:1824-1834. [PMID: 40138611 PMCID: PMC12084135 DOI: 10.1200/jco-24-01989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 01/12/2025] [Accepted: 02/13/2025] [Indexed: 03/29/2025] Open
Abstract
PURPOSE FOR46, a fully human antibody conjugated to monomethyl auristatin E, targets a tumor-selective epitope of CD46, which is overexpressed in metastatic castration-resistant prostate cancer (mCRPC). FOR46 demonstrates potent nonclinical activity in enzalutamide-resistant CRPC models. PATIENTS AND METHODS This was a phase I, first-in-human, dose escalation/expansion study in patients with progressive mCRPC after treatment with ≥one androgen signaling inhibitors (ClinicalTrials.gov identifier: NCT03575819). The starting dose of FOR46 was 0.1 mg/kg given intravenously every 3 weeks. The primary objective was to determine the maximally tolerated dose (MTD). Whole-blood mass cytometry (cytometry by time of flight) was used to characterize peripheral immune response and CD46 expression in CRPC tissue that underwent central pathology review. RESULTS Fifty-six patients were enrolled. Dose-limiting toxicities included neutropenia (n = 4), febrile neutropenia (n = 1), and fatigue (n = 1). The MTD was 2.7 mg/kg using adjusted body weight. The most common grade ≥3 adverse events across all dose levels were neutropenia (59%), leukopenia (27%), lymphopenia (7%), anemia (7%), and fatigue (5%). One grade 3 febrile neutropenia event was observed. There were no treatment-related deaths. In the efficacy evaluable subset (patients with adenocarcinoma treated with a starting dose ≥1.2 mg/kg, n = 40), the median radiographic progression-free survival was 8.7 months (range, 0.1-33.9). Fourteen of 39 evaluable patients (36%) achieved a PSA50 response. The confirmed objective response rate was 20% (5 of 25 RECIST-evaluable patients). The median duration of response was 7.5 months. Responders had a significantly higher on-treatment frequency of circulating effector CD8+ T cells. CONCLUSION FOR46 demonstrated encouraging preliminary clinical activity with a manageable safety profile. Targeting CD46 elicited an immune priming effect that was associated with clinical outcomes.
Collapse
Affiliation(s)
- Rahul R Aggarwal
- San Francisco Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA
| | | | - David VanderWeele
- Northwestern University Robert H. Lurie Comprehensive Cancer Center, Chicago, IL
| | - Matthew Rettig
- University of California Los Angeles VA Medical Center, Los Angeles, CA
| | | | - David Quigley
- San Francisco Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA
| | | | - Arun Chumber
- San Francisco Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA
| | - Alexander Cheung
- San Francisco Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA
| | - Adam Foye
- San Francisco Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA
| | - Stanley Leung
- San Francisco Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA
| | | | | | | | | | | | - Robert R Flavell
- San Francisco Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA
| | - Lawrence Fong
- San Francisco Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA
- Fred Hutchinson Comprehensive Cancer Center, Seattle, WA
| | - Bin Liu
- San Francisco Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA
| | - Eric J Small
- San Francisco Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA
| |
Collapse
|
2
|
Garlanda C, Dambra M, Magrini E. Interplay between the complement system and other immune pathways in the tumor microenvironment. Semin Immunol 2025; 78:101951. [PMID: 40209638 DOI: 10.1016/j.smim.2025.101951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/01/2025] [Accepted: 04/01/2025] [Indexed: 04/12/2025]
Abstract
Tumor growth and spread are sustained by the tumor microenvironment. Inflammatory cells and pathways have a fundamental role in the tumor microenvironment, driving or conditioning the functional activation of other leukocyte subsets and favoring evasion of anti-tumor immunity. One of the inflammatory pathways contributing to cancer-related inflammation is the complement system. Complement has long been considered an immune mechanism associated with immunosurveillance. More recently it emerged as a tumor promoting pathway, due to direct effects on cancer cells or indirect effects via immunosuppression driven by myeloid cells. The role of complement in cancer is complex and ambiguous, and depends on the tumor type and stage, as well as other factors including oncogenic drivers, leukocyte infiltration, interactions with other tumor microenvironment components or tumor cells. Other factors of complexity include the source of complement molecules, its canonical or non-canonical extracellular functions, its potential intracellular activation, and the interaction with other systems, such as the coagulation or the microbiome. Preclinical studies generally demonstrate the involvement of complement activation in smouldering inflammation in cancer and promotion of an immunosuppressive environment. These studies paved the way for clinical trials aimed at enhancing the potential of immunotherapy, in particular by targeting complement-dependent myeloid-sustained immunosuppression. However, the complex role of complement in cancer and the multiplicity of complement players may represent stumbling blocks and account for failures of clinical trials, and suggest that further studies are required to identify patient subsets who may benefit from specific complement molecule targeting in combination with conventional therapies or immunotherapy. Here, we will discuss the anti- or pro-tumor role of complement activation in cancer, focusing on the interactions of complement with immune cells within the tumor microenvironment, in particular the myeloid compartment. Furthermore, we will examine the potential of complement targeting in cancer treatment, particularly in the context of macrophage reprogramming.
Collapse
Affiliation(s)
- Cecilia Garlanda
- Department of Biomedical Sciences, Humanitas University, Milan, Pieve Emanuele 20072, Italy; IRCCS, Humanitas Research Hospital, Milan, Rozzano 20089, Italy.
| | - Monica Dambra
- IRCCS, Humanitas Research Hospital, Milan, Rozzano 20089, Italy
| | - Elena Magrini
- IRCCS, Humanitas Research Hospital, Milan, Rozzano 20089, Italy
| |
Collapse
|
3
|
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
|
4
|
Panebianco M, Ciccarese C, Strusi A, Beccia V, Carbone C, Agostini A, Piro G, Tortora G, Iacovelli R. The Role of the Complement in Clear Cell Renal Carcinoma (ccRCC)-What Future Prospects Are There for Its Use in Clinical Practice? Cancers (Basel) 2024; 16:490. [PMID: 38339243 PMCID: PMC10854780 DOI: 10.3390/cancers16030490] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/12/2024] [Accepted: 01/18/2024] [Indexed: 02/12/2024] Open
Abstract
In recent years, the first-line available therapeutic options for metastatic renal cell carcinoma (mRCC) have radically changed with the introduction into clinical practice of new immune checkpoint inhibitor (ICI)-based combinations. Many efforts are focusing on identifying novel prognostic and predictive markers in this setting. The complement system (CS) plays a central role in promoting the growth and progression of mRCC. In particular, mRCC has been defined as an "aggressive complement tumor", which encompasses a group of malignancies with poor prognosie and highly expressed complement components. Several preclinical and retrospective studies have demonstrated the negative prognostic role of the complement in mRCC; however, there is little evidence on its possible role as a predictor of the response to ICIs. The purpose of this review is to explore more deeply the physio-pathological role of the complement in the development of RCC and its possible future use in clinical practice as a prognostic and predictive factor.
Collapse
Affiliation(s)
- Martina Panebianco
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (M.P.); (C.C.); (C.C.); (A.A.); (G.P.); (G.T.)
| | - Chiara Ciccarese
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (M.P.); (C.C.); (C.C.); (A.A.); (G.P.); (G.T.)
| | - Alessandro Strusi
- Medical Oncology, Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy; (A.S.); (V.B.)
| | - Viria Beccia
- Medical Oncology, Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy; (A.S.); (V.B.)
| | - Carmine Carbone
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (M.P.); (C.C.); (C.C.); (A.A.); (G.P.); (G.T.)
| | - Antonio Agostini
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (M.P.); (C.C.); (C.C.); (A.A.); (G.P.); (G.T.)
| | - Geny Piro
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (M.P.); (C.C.); (C.C.); (A.A.); (G.P.); (G.T.)
| | - Giampaolo Tortora
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (M.P.); (C.C.); (C.C.); (A.A.); (G.P.); (G.T.)
- Medical Oncology, Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy; (A.S.); (V.B.)
| | - Roberto Iacovelli
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (M.P.); (C.C.); (C.C.); (A.A.); (G.P.); (G.T.)
- Medical Oncology, Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy; (A.S.); (V.B.)
| |
Collapse
|
5
|
Meri S, Magrini E, Mantovani A, Garlanda C. The Yin Yang of Complement and Cancer. Cancer Immunol Res 2023; 11:1578-1588. [PMID: 37902610 DOI: 10.1158/2326-6066.cir-23-0399] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/07/2023] [Accepted: 09/12/2023] [Indexed: 10/31/2023]
Abstract
Cancer-related inflammation is a crucial component of the tumor microenvironment (TME). Complement activation occurs in cancer and supports the development of an inflammatory microenvironment. Complement has traditionally been considered a mechanism of immune resistance against cancer, and its activation is known to contribute to the cytolytic effects of antibody-based immunotherapeutic treatments. However, several studies have recently revealed that complement activation may exert protumoral functions by sustaining cancer-related inflammation and immunosuppression through different molecular mechanisms, targeting both the TME and cancer cells. These new discoveries have revealed that complement manipulation can be considered a new strategy for cancer therapies. Here we summarize our current understanding of the mechanisms by which the different elements of the complement system exert antitumor or protumor functions, both in preclinical studies and in human tumorigenesis. Complement components can serve as disease biomarkers for cancer stratification and prognosis and be exploited for tumor treatment.
Collapse
Affiliation(s)
- Seppo Meri
- Department of Bacteriology and Immunology and Translational Immunology Research Program, University and University Hospital of Helsinki, Helsinki, Finland
| | | | - Alberto Mantovani
- IRCCS-Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Cecilia Garlanda
- IRCCS-Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| |
Collapse
|
6
|
Collier-Bain HD, Brown FF, Causer AJ, Emery A, Oliver R, Moore S, Murray J, Turner JE, Campbell JP. Harnessing the immunomodulatory effects of exercise to enhance the efficacy of monoclonal antibody therapies against B-cell haematological cancers: a narrative review. Front Oncol 2023; 13:1244090. [PMID: 37681023 PMCID: PMC10482436 DOI: 10.3389/fonc.2023.1244090] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/09/2023] [Indexed: 09/09/2023] Open
Abstract
Therapeutic monoclonal antibodies (mAbs) are standard care for many B-cell haematological cancers. The modes of action for these mAbs include: induction of cancer cell lysis by activating Fcγ-receptors on innate immune cells; opsonising target cells for antibody-dependent cellular cytotoxicity or phagocytosis, and/or triggering the classical complement pathway; the simultaneous binding of cancer cells with T-cells to create an immune synapse and activate perforin-mediated T-cell cytotoxicity against cancer cells; blockade of immune checkpoints to facilitate T-cell cytotoxicity against immunogenic cancer cell clones; and direct delivery of cytotoxic agents via internalisation of mAbs by target cells. While treatment regimens comprising mAb therapy can lead to durable anti-cancer responses, disease relapse is common due to failure of mAb therapy to eradicate minimal residual disease. Factors that limit mAb efficacy include: suboptimal effector cell frequencies, overt immune exhaustion and/or immune anergy, and survival of diffusely spread tumour cells in different stromal niches. In this review, we discuss how immunomodulatory changes arising from exposure to structured bouts of acute exercise might improve mAb treatment efficacy by augmenting (i) antibody-dependent cellular cytotoxicity, (ii) antibody-dependent cellular phagocytosis, (iii) complement-dependent cytotoxicity, (iv) T-cell cytotoxicity, and (v) direct delivery of cytotoxic agents.
Collapse
Affiliation(s)
| | - Frankie F. Brown
- Department for Health, University of Bath, Bath, United Kingdom
- School of Applied Sciences, Edinburgh Napier University, Edinburgh, United Kingdom
| | - Adam J. Causer
- Department for Health, University of Bath, Bath, United Kingdom
| | - Annabelle Emery
- Department for Health, University of Bath, Bath, United Kingdom
| | - Rebecca Oliver
- Department for Health, University of Bath, Bath, United Kingdom
- Department of Haematology, Royal United Hospitals Bath NHS Foundation Trust, Bath, United Kingdom
| | - Sally Moore
- Department of Haematology, Royal United Hospitals Bath NHS Foundation Trust, Bath, United Kingdom
| | - James Murray
- Department of Haematology, Royal United Hospitals Bath NHS Foundation Trust, Bath, United Kingdom
| | - James E. Turner
- Department for Health, University of Bath, Bath, United Kingdom
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
| | | |
Collapse
|
7
|
Liu J, Fu N, Yang Z, Li A, Wu H, Jin Y, Song Q, Ji S, Xu H, Zhang Z, Zhang X. The genetic and epigenetic regulation of CD55 and its pathway analysis in colon cancer. Front Immunol 2023; 13:947136. [PMID: 36741376 PMCID: PMC9889927 DOI: 10.3389/fimmu.2022.947136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 12/28/2022] [Indexed: 01/20/2023] Open
Abstract
Background CD55 plays an important role in the development of colon cancer. This study aims to evaluate the expression of CD55 in colon cancer and discover how it is regulated by transcriptional factors and miRNA. Methods The expression of CD55 was explored by TIMER2.0, UALCAN, and Human Protein Atlas (HPA) databases. TRANSFAC and Contra v3 were used to predict the potential binding sites of transcription factors in the CD55 promoter. TargetScan and starBase v2.0 were used to predict the potential binding ability of miRNAs to the 3' untranslated region (3'UTR) of CD55. SurvivalMeth was used to explore the differentially methylated sites in the CD55 promoter. Western blotting was used to detect the expression of TFCP2 and CD55. Dual-luciferase reporter assay and chromatin immunoprecipitation (ChIP) assay were performed to determine the targeting relationship of TFCP2, NF-κB, or miR-27a-3p with CD55. CD55-related genes were explored by constructing a protein-protein interaction (PPI) network and performing pathway analysis by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Results CD55 was highly expressed in colon cancer tissues. The mRNA and protein expression levels of TFCP2 were reduced by si-TFCP2. NF-κB mRNA was obviously reduced by NF-κB inhibitor and increased by NF-κB activator. CD55 protein was also inhibited by miR-27a-3p. Dual-luciferase reporter assays showed that after knocking down TFCP2 or inhibiting NF-κB, the promoter activity of CD55 was decreased by 21% and 70%, respectively; after activating NF-κB, the promoter activity of CD55 increased by 2.3 times. As TFCP2 or NF-κB binding site was mutated, the transcriptional activity of CD55 was significantly decreased. ChIP assay showed that TFCP2 and NF-κB combined to the promoter of CD55. The luciferase activity of CD55 3'UTR decreased after being co-transfected with miR-27a-3p mimics and increased by miR-27a-3p antagomir. As the miR-27a-3p binding site was mutated, we did not find any significant effect of miR-27a-3p on reporter activity. PPI network assay revealed a set of CD55-related genes, which included CFP, CFB, C4A, and C4B. GO and KEGG analyses revealed that the target genes occur more frequently in immune-related pathways. Conclusion Our results indicated that CD55 is regulated by TFCP2, NF-κB, miR-27a-3p, and several immune-related genes, which in turn affects colon cancer.
Collapse
Affiliation(s)
- Jiawei Liu
- Affiliated Tangshan Gongren Hospital, North China University of Science and Technology, Tangshan, China
- College of Life Science, North China University of Science and Technology, Tangshan, China
| | - Ning Fu
- College of Life Science, North China University of Science and Technology, Tangshan, China
| | - Zhenbang Yang
- School of Public Health, North China University of Science and Technology, Tangshan, China
| | - Ang Li
- School of Public Health, North China University of Science and Technology, Tangshan, China
| | - Hongjiao Wu
- School of Public Health, North China University of Science and Technology, Tangshan, China
| | - Ye Jin
- College of Life Science, North China University of Science and Technology, Tangshan, China
| | - Qinqin Song
- Affiliated Tangshan Gongren Hospital, North China University of Science and Technology, Tangshan, China
| | - Shanshan Ji
- Affiliated Tangshan Gongren Hospital, North China University of Science and Technology, Tangshan, China
| | - Hongxue Xu
- School of Public Health, North China University of Science and Technology, Tangshan, China
| | - Zhi Zhang
- Affiliated Tangshan Gongren Hospital, North China University of Science and Technology, Tangshan, China
| | - Xuemei Zhang
- College of Life Science, North China University of Science and Technology, Tangshan, China
- School of Public Health, North China University of Science and Technology, Tangshan, China
| |
Collapse
|
8
|
Xu H, Dun B, Liu B, Mysona D, She J, Ma R. A novel monoclonal antibody associated with glucoside kills gastric adenocarcinoma AGS cells based on glycosylation target. J Cell Mol Med 2022; 26:4781-4791. [PMID: 35946053 PMCID: PMC9465190 DOI: 10.1111/jcmm.17504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 07/07/2022] [Accepted: 07/13/2022] [Indexed: 11/29/2022] Open
Abstract
Glycosylation results in the production of glycans which are required for certain proteins to function. These glycans are also present on cell surfaces where they help maintain cell membrane integrity and are a key component of immune recognition. As such, cancer has been shown to alter glycosylation to promote tumour proliferation, invasion, angiogenesis, and immune envasion. Currently, there are few therapeutic monoclonal antibodies (mAb) which target glycosylation alterations in cancer. Here, we report a novel mAb associated with a glucoside, mAb 201E4, which is able induce cancer cell death and apoptosis based on a specific glycosylation target. This mAb evokes cancer cell death in vitro via caspase, fas, and mitochondrial associated apoptotic pathways. The efficacy of this mAb was further confirmed in vivo as treatment of mice with mAb 201E4 resulted in potent tumour shrinkage. Finally, the antibody was proven to be specific to glycosylation alterations in cancer and have no binding to normal tissues. This data indicates that mAb 201E4 successfully targets glycosylation alterations in neoplasms to induce cancer cell death, which may provide a new strategy for therapy in cancer.
Collapse
Affiliation(s)
- Heng Xu
- Jiangsu Provincial Institute of Materia MedicaNanjing Tech UniversityNanjingChina
- Jinfiniti Precision MedicineAugustaGeorgiaUSA
| | - Boying Dun
- Jinfiniti Precision MedicineAugustaGeorgiaUSA
| | - Beiyi Liu
- Institute of Animal ScienceJiangsu Academy of Agricultural ScienceNanjingJiangsuChina
| | | | | | - Rong Ma
- Research Center For Clinical OncologyJiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer HospitalNanjingChina
| |
Collapse
|
9
|
Gakis G, Perner S, Stenzl A, Renninger M. The role of single-nucleotide polymorphisms of the 8q24 chromosome region in patients with concomitant bladder and prostate cancer. Scand J Urol 2022; 56:126-130. [PMID: 35274594 DOI: 10.1080/21681805.2022.2049362] [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: 10/18/2022]
Abstract
OBJECTIVE To assess whether single-nucleotide polymorphisms (SNPs) of the 8q24 chromosome region are associated with recurrence-free survival (RFS) after radical cystoprostatectomy (RC) in patients with concomitant bladder (BC) and prostate cancer (PC). MATERIALS AND METHODS A cohort of thirty-six patients treated with RC and pelvic lymph node dissection and histologically exhibited invasive BC and incidental PC. Using Sanger sequencing, a total of seven SNPs in the androgen-responsive element of the promoter region of the following genes were assessed in tumor-free lymph nodes and correlated with oncological outcomes: PSCA (rs2294008, rs2978974, rs1045531, rs3736001), MYC (rs6983267), FXBO32 (rs7830622), and MIR151A (rs14974929). The median follow-up was 26 months (range: 4-68). RESULTS In a dominant model, patients exhibiting rs2978974 as a minor allelic variant of the PSCA gene had worse RFS (32 vs. 75%, p = 0.015). No associations were found for the other SNPs. CONCLUSIONS These data suggest that the rs2978974 of the PSCA gene correlates with inferior BC-specific RFS after RC and should be further evaluated in larger studies.
Collapse
Affiliation(s)
- Georgios Gakis
- Department of Urology and Pediatric Urology, University Hospital of Würzburg, Julius-Maximillians University, Würzburg, Germany
| | - Sven Perner
- Institute of Pathology, University of Luebeck, University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany.,Department of Pathology, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Arnulf Stenzl
- Department of Urology, University Hospital, Eberhard-Karls University, Tübingen, Germany
| | - Markus Renninger
- Department of Urology, University Hospital, Eberhard-Karls University, Tübingen, Germany
| |
Collapse
|
10
|
Complement activation in cancer: Effects on tumor-associated myeloid cells and immunosuppression. Semin Immunol 2022; 60:101642. [PMID: 35842274 DOI: 10.1016/j.smim.2022.101642] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/28/2022] [Accepted: 07/05/2022] [Indexed: 01/15/2023]
Abstract
Cancer-related inflammation plays a central role in the establishment of tumor-promoting mechanisms. Tumor-associated myeloid cells, which engage in complex interactions with cancer cells, as well as stromal and tumor immune infiltrating cells, promote cancer cell proliferation and survival, angiogenesis, and the generation of an immunosuppressive microenvironment. The complement system is one of the inflammatory mechanisms activated in the tumor microenvironment. Beside exerting anti-tumor mechanisms such as complement-dependent cytotoxicity and phagocytosis induced by therapeutic monoclonal antibodies, the complement system may promote immunosuppression and tumor growth and invasiveness, in particular, through the anaphylatoxins which target both leukocytes and cancer cells. In this review, we will discuss complement-mediated mechanisms acting on leukocytes, in particular on cells of the myelomonocytic cell lineage (macrophages, neutrophils, myeloid derived suppressor cells), which promote myeloid cell recruitment and functional skewing, leading to immunosuppression and resistance to tumor-specific immunity. Pre-clinical studies, which have elucidated the role of complement in activating pro-tumor mechanisms in myeloid cells, showing the relevance of these mechanisms in human, and therapeutic approaches based on complement targeting support the hypothesis that complement directly and indirectly interferes with many of the effector pathways associated with the cancer-immunity cycle, suggesting the relevance of complement targeting to improve responses to immunotherapeutic approaches.
Collapse
|
11
|
Kneiber D, Kowalski EH, Amber KT. The Immunogenetics of Autoimmune Blistering Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1367:173-212. [DOI: 10.1007/978-3-030-92616-8_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
|
12
|
Yu F, Yu C, Li F, Zuo Y, Wang Y, Yao L, Wu C, Wang C, Ye L. Wnt/β-catenin signaling in cancers and targeted therapies. Signal Transduct Target Ther 2021; 6:307. [PMID: 34456337 PMCID: PMC8403677 DOI: 10.1038/s41392-021-00701-5] [Citation(s) in RCA: 389] [Impact Index Per Article: 97.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 06/19/2021] [Accepted: 06/28/2021] [Indexed: 02/07/2023] Open
Abstract
Wnt/β-catenin signaling has been broadly implicated in human cancers and experimental cancer models of animals. Aberrant activation of Wnt/β-catenin signaling is tightly linked with the increment of prevalence, advancement of malignant progression, development of poor prognostics, and even ascendence of the cancer-associated mortality. Early experimental investigations have proposed the theoretical potential that efficient repression of this signaling might provide promising therapeutic choices in managing various types of cancers. Up to date, many therapies targeting Wnt/β-catenin signaling in cancers have been developed, which is assumed to endow clinicians with new opportunities of developing more satisfactory and precise remedies for cancer patients with aberrant Wnt/β-catenin signaling. However, current facts indicate that the clinical translations of Wnt/β-catenin signaling-dependent targeted therapies have faced un-neglectable crises and challenges. Therefore, in this study, we systematically reviewed the most updated knowledge of Wnt/β-catenin signaling in cancers and relatively targeted therapies to generate a clearer and more accurate awareness of both the developmental stage and underlying limitations of Wnt/β-catenin-targeted therapies in cancers. Insights of this study will help readers better understand the roles of Wnt/β-catenin signaling in cancers and provide insights to acknowledge the current opportunities and challenges of targeting this signaling in cancers.
Collapse
Affiliation(s)
- Fanyuan Yu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Endodontics, West China Stomatology Hospital, Sichuan University, Chengdu, China
| | - Changhao Yu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Endodontics, West China Stomatology Hospital, Sichuan University, Chengdu, China
| | - Feifei Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yanqin Zuo
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Endodontics, West China Stomatology Hospital, Sichuan University, Chengdu, China
| | - Yitian Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lin Yao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Endodontics, West China Stomatology Hospital, Sichuan University, Chengdu, China
| | - Chenzhou Wu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chenglin Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Endodontics, West China Stomatology Hospital, Sichuan University, Chengdu, China
| | - Ling Ye
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
- Department of Endodontics, West China Stomatology Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
13
|
Potent Preclinical Efficacy of DuoHexaBody-CD37 in B-Cell Malignancies. Hemasphere 2020; 5:e504. [PMID: 33324950 PMCID: PMC7732266 DOI: 10.1097/hs9.0000000000000504] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/22/2020] [Indexed: 11/27/2022] Open
|
14
|
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: 3.0] [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
|
15
|
Liu F, Luo L, Liu Z, Wu S, Zhang W, Li Q, Peng Y, Wei Y, Li B. A genetic variant in the promoter of CD46 is associated with the risk and prognosis of hepatocellular carcinoma. Mol Carcinog 2020; 59:1243-1255. [PMID: 32869896 DOI: 10.1002/mc.23252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/26/2020] [Accepted: 08/12/2020] [Indexed: 02/05/2023]
Abstract
CD46 (also known as membrane cofactor protein), which is a member of the membrane-bound complement regulatory protein family, has been reported to cause cancer cells to escape complement-dependent cytotoxicity. However, the association between CD46 polymorphisms and the risk of hepatocellular carcinoma (HCC) has not been investigated. This two-stage association study was conducted to assess the relationship between the tagging single nucleotide polymorphisms (tagSNPs) of CD46 and HCC risk and prognosis. A series of functional analyses were performed to study the underlying mechanisms. Among the eight tagSNPs, rs2796267 (P = .003) and rs2796268 (P = .011) were found to modify HCC risk in the discovery set. Only rs2796267 (P < .0001) was confirmed to be associated with HCC susceptibility in the validation set. Compared with the wild-type AA genotype, the GG genotype significantly increased the HCC risk (adjusted odds ratio [OR] = 2.03; 95% confidence interval [CI], 1.34-3.08; P = .001). Moreover, subgroups analysis suggested a positive correlation among male and younger patients, especially among drinkers, smokers, and hepatitis B surface antigen-positive individuals. In functional analyses, we found that the rs2796267 G allele in the promoter region of CD46 could increase the expression of CD46 by affecting the binding affinity of STAT5a. Furthermore, Cox regression analysis revealed that the rs2796267 AG/GG genotype was significantly associated with worse prognosis of resected patients with HCC (hazard ratio = 2.27; 95% CI, 1.27-4.05; P = .006). These results suggest that the CD46 rs2796267 polymorphism may contribute to susceptibility and prognosis of HCC by altering promoter activity.
Collapse
Affiliation(s)
- Fei Liu
- Department of Liver Surgery, Liver Transplantation Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Limei Luo
- Department of Clinical Immunological Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhongjian Liu
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Sisi Wu
- Division of Core Facilities, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Zhang
- Department of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Qin Li
- Department of Liver Surgery, Liver Transplantation Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yufu Peng
- Department of Liver Surgery, Liver Transplantation Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yonggang Wei
- Department of Liver Surgery, Liver Transplantation Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bo Li
- Department of Liver Surgery, Liver Transplantation Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
16
|
Liu Y, Xu P, Liu H, Fang C, Guo H, Chen X, Tan M, Zhang Y, Min W. Silencing IDO2 in dendritic cells: A novel strategy to strengthen cancer immunotherapy in a murine lung cancer model. Int J Oncol 2020; 57:587-597. [PMID: 32468023 DOI: 10.3892/ijo.2020.5073] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 05/06/2020] [Indexed: 11/05/2022] Open
Abstract
While dendritic cell (DC)‑based immunotherapy has achieved satisfactory results in animal models, its effects were not satisfactory as initially expected in clinical applications, despite the safety and varying degrees of effectiveness in various types of cancer. Improving the efficacy of the DC‑based vaccine is essential for cancer immunotherapy. The present study aimed to investigate methods with which to amplify and enhance the antitumor immune response of a DC‑based tumor vaccine by silencing the expression of indoleamine 2,3‑dioxygenase 2 (IDO2), a tryptophan rate‑limiting metabolic enzyme in DCs. In vitro experiments revealed that the silencing of IDO2 in DCs did not affect the differentiation of DCs, whereas it increased their expression of costimulatory molecules following stimulation with tumor necrosis factor (TNF)‑α and tumor lysate from Lewis lung cancer (LLC) cells. In a mixed co‑culture system, the IDO2‑silenced DCs promoted the proliferation of T‑cells and reduced the induction of regulatory T‑cells (Tregs). Further in vivo experiments revealed that the silencing of IDO2 in DCs markedly suppressed the growth of tumor cells. Moreover, treatment with the IDO2‑silenced DC‑based cancer vaccine enhanced cytotoxic T lymphocyte activity, whereas it decreased T‑cell apoptosis and the percentage of CD4+CD25+Foxp3+ Tregs. On the whole, the present study provides evidence that the silencing of the tryptophan rate‑limiting metabolic enzyme, IDO2, has the potential to enhance the efficacy of DC‑based cancer immunotherapy.
Collapse
Affiliation(s)
- Yanling Liu
- Medical Laboratory, Jiangxi University of Technology, Nanchang, Jiangxi 330098, P.R. China
| | - Ping Xu
- Medical Laboratory, Jiangxi University of Technology, Nanchang, Jiangxi 330098, P.R. China
| | - Huan Liu
- Medical Laboratory, Jiangxi University of Technology, Nanchang, Jiangxi 330098, P.R. China
| | - Chunjuan Fang
- Medical Laboratory, Jiangxi University of Technology, Nanchang, Jiangxi 330098, P.R. China
| | - Haihe Guo
- Medical Laboratory, Jiangxi University of Technology, Nanchang, Jiangxi 330098, P.R. China
| | - Xiaoyan Chen
- Medical Laboratory, Jiangxi University of Technology, Nanchang, Jiangxi 330098, P.R. China
| | - Manman Tan
- Institute of Immunotherapy, Nanchang University and Jiangxi Academy of Medical Science, Nanchang, Jiangxi 330098, P.R. China
| | - Yujuan Zhang
- Institute of Immunotherapy, Nanchang University and Jiangxi Academy of Medical Science, Nanchang, Jiangxi 330098, P.R. China
| | - Weiping Min
- Institute of Immunotherapy, Nanchang University and Jiangxi Academy of Medical Science, Nanchang, Jiangxi 330098, P.R. China
| |
Collapse
|
17
|
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: 11] [Impact Index Per Article: 2.2] [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
|
18
|
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: 0.8] [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
|
19
|
Olcina MM, Balanis NG, Kim RK, Aksoy BA, Kodysh J, Thompson MJ, Hammerbacher J, Graeber TG, Giaccia AJ. Mutations in an Innate Immunity Pathway Are Associated with Poor Overall Survival Outcomes and Hypoxic Signaling in Cancer. Cell Rep 2019; 25:3721-3732.e6. [PMID: 30590044 PMCID: PMC6405289 DOI: 10.1016/j.celrep.2018.11.093] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 10/01/2018] [Accepted: 11/27/2018] [Indexed: 12/18/2022] Open
Abstract
Complement-mediated cytotoxicity may act as a selective pressure for tumor overexpression of complement regulators. We hypothesize that the same selective pressure could lead to complement alterations at the genetic level. We find that, when analyzed as a pathway, mutations in complement genes occur at a relatively high frequency and are associated with changes in overall survival across a number of cancer types. Analysis of pathways expressed in patients with complement mutations that are associated with poor overall survival reveals crosstalk between complement and hypoxia in colorectal cancer. The importance of this crosstalk is highlighted by two key findings: hypoxic signaling is increased in tumors harboring complement mutations, and hypoxic tumor cells are resistant to complement-mediated cytotoxicity due, in part, to hypoxia-induced expression of complement regulator CD55. The range of strategies employed by tumors to dysregulate the complement system testifies to the importance of this pathway in tumor progression.
Collapse
Affiliation(s)
- Monica M Olcina
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA.
| | - Nikolas G Balanis
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ryan K Kim
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - B Arman Aksoy
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Julia Kodysh
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Michael J Thompson
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jeff Hammerbacher
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Thomas G Graeber
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA
| | - Amato J Giaccia
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
20
|
Seguin-Devaux C, Plesseria JM, Verschueren C, Masquelier C, Iserentant G, Fullana M, Józsi M, Cohen JHM, Dervillez X. FHR4-based immunoconjugates direct complement-dependent cytotoxicity and phagocytosis towards HER2-positive cancer cells. Mol Oncol 2019; 13:2531-2553. [PMID: 31365168 PMCID: PMC6887587 DOI: 10.1002/1878-0261.12554] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 06/13/2019] [Accepted: 07/30/2019] [Indexed: 12/22/2022] Open
Abstract
Directing selective complement activation towards tumour cells is an attractive strategy to promote their elimination. In the present work, we have generated heteromultimeric immunoconjugates that selectively activate the complement alternative pathway (AP) on tumour cells. We used the C4b‐binding protein C‐terminal‐α‐/β‐chain scaffold for multimerisation to generate heteromultimeric immunoconjugates displaying (a) a multivalent‐positive regulator of the AP, the human factor H‐related protein 4 (FHR4) with; (b) a multivalent targeting function directed against erbB2 (HER2); and (c) a monovalent enhanced GFP tracking function. Two distinct VHH targeting two different epitopes against HER2 and competing either with trastuzumab or with pertuzumab‐recognising epitopes [VHH(T) or VHH(P)], respectively, were used as HER2 anchoring moieties. Optimised high‐FHR4 valence heteromultimeric immunoconjugates [FHR4/VHH(T) or FHR4/VHH(P)] were selected by sequential cell cloning and a selective multistep His‐Trap purification. Optimised FHR4‐heteromultimeric immunoconjugates successfully overcame FH‐mediated complement inhibition threshold, causing increased C3b deposition on SK‐OV‐3, BT474 and SK‐BR3 tumour cells, and increased formation of lytic membrane attack complex densities and complement‐dependent cytotoxicity (CDC). CDC varies according to the pattern expression and densities of membrane‐anchored complement regulatory proteins on tumour cell surfaces. In addition, opsonised BT474 tumour cells were efficiently phagocytosed by macrophages through complement‐dependent cell‐mediated cytotoxicity. We showed that the degree of FHR4‐multivalency within the multimeric immunoconjugates was the key element to efficiently compete and deregulate FH and FH‐mediated convertase decay locally on tumour cell surface. FHR4 can thus represent a novel therapeutic molecule, when expressed as a multimeric entity and associated with an anchoring system, to locally shift the complement steady‐state towards activation on tumour cell surface.
Collapse
Affiliation(s)
- Carole Seguin-Devaux
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Jean-Marc Plesseria
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Charlène Verschueren
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Cécile Masquelier
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Gilles Iserentant
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Marie Fullana
- Société d'Accélération des Transferts de Technologies du Nord, Direction Territoriale Reims, Reims, France
| | - Mihály Józsi
- Complement Research Group, Department of Immunology, ELTE Eötvös Loránd University, Budapest, Hungary
| | | | - Xavier Dervillez
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg.,Life Sciences Research Unit (LSRU), Signal Transduction Laboratory, University of Luxembourg, Belvaux, Luxembourg
| |
Collapse
|
21
|
DeCordova S, Abdelgany A, Murugaiah V, Pathan AA, Nayak A, Walker T, Shastri A, Alrokayan SH, Khan HA, Singh SK, De Pennington N, Sim RB, Kishore U. Secretion of functionally active complement factor H related protein 5 (FHR5) by primary tumour cells derived from Glioblastoma Multiforme patients. Immunobiology 2019; 224:625-631. [PMID: 31519376 DOI: 10.1016/j.imbio.2019.07.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/11/2019] [Accepted: 07/30/2019] [Indexed: 02/08/2023]
Abstract
The complement system is an important humoral immune surveillance mechanism against tumours. However, many malignant tumours are resistant to complement mediated lysis. Here, we report secretion of complement factor H related protein 5 (FHR5) by primary tumour cells derived from Glioblastoma multiforme (GBM) patients. We investigated whether the secreted FHR5 exhibited functional activity similar to factor H, including inhibition of complement mediated lysis, acting as a co-factor for factor I mediated cleavage of C3b, and decay acceleration of C3 convertase. Immunoblotting analysis of primary GBM cells (B30, B31 and B33) supernatant showed the active secretion of FHR5, but not of Factor H. ELISA revealed that the secretion of soluble GBM-FHR5 by cultured GBM cells increased in a time-dependent manner. Primary GBM-FHR5 inhibited complement mediated lysis, possessed co-factor activity for factor I mediated cleavage and displayed decay acceleration of C3 convertase. In summary, we detected the secretion of FHR5 by primary GBM cells B30, B31 and B33. The results demonstrated that GBM-FHR5 shares biological function with FH as a mechanism primary GBM cells potentially use to resist complement mediated lysis.
Collapse
Affiliation(s)
- Syreeta DeCordova
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, UB8 3PH, UK
| | - Amr Abdelgany
- Nuffield Department of Clinical Neuroscience, John Radcliffe Hospital, University of Oxford, Headington, OX3 9DS, UK; Department of Oncology, University of Oxford, Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Valarmathy Murugaiah
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, UB8 3PH, UK
| | - Ansar A Pathan
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, UB8 3PH, UK
| | - Annapurna Nayak
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, UB8 3PH, UK
| | - Tom Walker
- Nuffield Department of Clinical Neuroscience, John Radcliffe Hospital, University of Oxford, Headington, OX3 9DS, UK
| | - Abhishek Shastri
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, UB8 3PH, UK; Westminster Community Mental Health Team, Central and North-West London NHS Foundation Trust, London SW1V 1DX, UK
| | - Salman H Alrokayan
- Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Haseeb A Khan
- Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Shiv K Singh
- Department of Gastroenterology and Gastrointestinal Oncology, University Medical Centre, Goettingen, Germany
| | - Nick De Pennington
- Nuffield Department of Clinical Neuroscience, John Radcliffe Hospital, University of Oxford, Headington, OX3 9DS, UK
| | - Robert B Sim
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
| | - Uday Kishore
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, UB8 3PH, UK.
| |
Collapse
|
22
|
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: 6.5] [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
|
23
|
Brady JV, Troyer RM, Ramsey SA, Leeper H, Yang L, Maier CS, Goodall CP, Ruby CE, Albarqi HAM, Taratula O, Bracha S. A Preliminary Proteomic Investigation of Circulating Exosomes and Discovery of Biomarkers Associated with the Progression of Osteosarcoma in a Clinical Model of Spontaneous Disease. Transl Oncol 2018; 11:1137-1146. [PMID: 30053712 PMCID: PMC6077151 DOI: 10.1016/j.tranon.2018.07.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 07/02/2018] [Accepted: 07/06/2018] [Indexed: 12/21/2022] Open
Abstract
Circulating cancer exosomes are microvesicles which originate from malignant cells and other organs influenced by the disease and can be found in blood. The exosomal proteomic cargo can often be traced to the cells from which they originated, reflecting the physiological status of these cells. The similarities between cancer exosomes and the tumor cells they originate from exhibit the potential of these vesicles as an invaluable target for liquid biopsies. Exosomes were isolated from the serum of eight osteosarcoma-bearing dogs, five healthy dogs, and five dogs with traumatic fractures. We also characterized exosomes which were collected longitudinally from patients with osteosarcoma prior and 2 weeks after amputation, and eventually upon detection of lung metastasis. Exosomal proteins fraction were analyzed by label-free mass spectrometry proteomics and were validated with immunoblots of selected proteins. Ten exosomal proteins were found that collectively discriminate serum of osteosarcoma patients from serum healthy or fractured dogs with an accuracy of 85%. Additionally, serum from different disease stages could be distinguished with an accuracy of 77% based on exosomal proteomic composition. The most discriminating protein changes for both sample group comparisons were related to complement regulation, suggesting an immune evasion mechanism in early stages of osteosarcoma as well as in advanced disease.
Collapse
Affiliation(s)
- Jacqueline V Brady
- Carlson College of Veterinary Medicine, Department of Clinical Sciences, Oregon State University, Corvallis, OR, USA
| | - Ryan M Troyer
- Carlson College of Veterinary Medicine, Department of Clinical Sciences, Oregon State University, Corvallis, OR, USA
| | - Stephen A Ramsey
- Carlson College of Veterinary Medicine, Department of Biomedical Sciences, Oregon State University, Corvallis, OR, USA
| | - Haley Leeper
- Carlson College of Veterinary Medicine, Department of Clinical Sciences, Oregon State University, Corvallis, OR, USA
| | - Liping Yang
- College of Science, Department of Chemistry, Oregon State University, Corvallis, OR, USA
| | - Claudia S Maier
- College of Science, Department of Chemistry, Oregon State University, Corvallis, OR, USA
| | - Cheri P Goodall
- Carlson College of Veterinary Medicine, Department of Clinical Sciences, Oregon State University, Corvallis, OR, USA
| | - Carl E Ruby
- Carlson College of Veterinary Medicine, Department of Clinical Sciences, Oregon State University, Corvallis, OR, USA
| | | | - Oleh Taratula
- College of Pharmacy, Oregon State University, Corvallis, OR, USA
| | - Shay Bracha
- Carlson College of Veterinary Medicine, Department of Clinical Sciences, Oregon State University, Corvallis, OR, USA.
| |
Collapse
|
24
|
Zhang L, Sorensen MD, Kristensen BW, Reifenberger G, McIntyre TM, Lin F. D-2-Hydroxyglutarate Is an Intercellular Mediator in IDH-Mutant Gliomas Inhibiting Complement and T Cells. Clin Cancer Res 2018; 24:5381-5391. [PMID: 30006485 DOI: 10.1158/1078-0432.ccr-17-3855] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 05/16/2018] [Accepted: 07/09/2018] [Indexed: 11/16/2022]
Abstract
Purpose: Somatic mutations in the isocitrate dehydrogenase (IDH)-1 and -2 genes are remarkably penetrant in diffuse gliomas. These highly effective gain-of-function mutations enable mutant IDH to efficiently metabolize isocitrate to D-2-hydroxyglutarate (D 2-HG) that accumulates to high concentrations within the tumor microenvironment. D 2-HG is an intracellular effector that promotes tumor growth through widespread epigenetic changes in IDH-mutant tumor cells, but its potential role as an intercellular immune regulator remains understudied.Experimental Design: Complement activation and CD4+, CD8+, or FOXP3+ T-cell infiltration into primary tumor tissue were determined by immunohistochemistry using sections from 72 gliomas of World Health Organization (WHO) grade III and IV with or without IDH mutations. Ex vivo experiments with D 2-HG identified immune inhibitory mechanisms.Results: IDH mutation associated with significantly reduced complement activation and decreased numbers of tumor-infiltrating CD4+ and CD8+ T cells with comparable FOXP3+/CD4+ ratios. D 2-HG potently inhibited activation of complement by the classical and alternative pathways, attenuated complement-mediated glioma cell damage, decreased cellular C3b(iC3b) opsonization, and impaired complement-mediated phagocytosis. Although D 2-HG did not affect dendritic cell differentiation or function, it significantly inhibited activated T-cell migration, proliferation, and cytokine secretion.Conclusions: D 2-HG suppresses the host immune system, potentially promoting immune escape of IDH-mutant tumors. Clin Cancer Res; 24(21); 5381-91. ©2018 AACR.
Collapse
Affiliation(s)
- Lingjun Zhang
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Mia D Sorensen
- Department of Pathology, Odense University Hospital, Odense C, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense C, Denmark
| | - Bjarne W Kristensen
- Department of Pathology, Odense University Hospital, Odense C, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense C, Denmark
| | - Guido Reifenberger
- Department of Neuropathology, Heinrich Heine University, Düsseldorf, Germany
| | - Thomas M McIntyre
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Feng Lin
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.
| |
Collapse
|
25
|
Wang Y, Liao J, Yang YJ, Wang Z, Qin F, Zhu SM, Zheng H, Wang YP. Effect of membrane-bound complement regulatory proteins on tumor cell sensitivity to complement-dependent cytolysis triggered by heterologous expression of the α-gal xenoantigen. Oncol Lett 2018; 15:9061-9068. [PMID: 29805637 PMCID: PMC5958734 DOI: 10.3892/ol.2018.8478] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 02/28/2018] [Indexed: 02/05/2023] Open
Abstract
Engineering malignant cells to express a heterologous α-gal antigen can induce heterograft hyperacute rejection, resulting in complement-dependent cytolysis (CDC) of tumor cells, which has been considered as a novel strategy for antitumor therapy. A549 cells engineered to express Galα1-3Galβ1-4GlcNAc-R (α-gal) epitope exhibited strong resistance to CDC treated by normal human serum (NHS) in a previous study. We hypothesized that the expression of membrane-bound complement regulatory proteins (mCRPs) decay accelerating factor (CD55) and protectin (CD59) influenced the efficacy of the α-gal/NHS-mediated antitumor effect to tumor cells in vitro. The present study confirmed that A549 cells expressed high levels of CD55 and CD59, whereas Lovo cells expressed relatively low levels of these proteins. A549 and Lovo cells transfected with plasmids containing or lacking the α-gal epitope were evaluated for their susceptibility to CDC by NHS and detected using a trypan blue exclusion assay. α-gal-expressing Lovo (Lovo-GT) cells were almost completely killed by α-gal-mediated CDC following incubation with 50% NHS, whereas no cytolysis was observed in α-gal expressing A549 (A549-GT) cells. Abrogating CD55 and CD59 function from A549-GT cells by various concentrations of phosphatidylinositol-specific phospholipase C (PI-PLC) or blocking antibodies increased the susceptibility of cells to CDC, and the survival rate decreased significantly comparing to the controls (P<0.05). The findings of the present study indicated that using the α-gal/NHS system to eliminate tumor cells via inducing the complement cascade reaction might represent a feasible approach for the treatment of cancer. However, high levels of mCRP expression may limit the efficacy of this approach. Therefore, an improved efficacy of cancer cell killing may be achieved by combining strategies of heterologous α-gal expression and mCRP downregulation.
Collapse
Affiliation(s)
- Yu Wang
- Laboratory of Molecular Diagnosis of Cancer, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Juan Liao
- Laboratory of Molecular Diagnosis of Cancer, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Ya-Jun Yang
- Laboratory of Molecular Diagnosis of Cancer, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Zhu Wang
- Laboratory of Molecular Diagnosis of Cancer, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Feng Qin
- Basic Medical Faculty, Dali Medical College, Dali, Yunnan 671003, P.R. China
| | - Sheng-Ming Zhu
- Department of Oncology, Affiliated Taihe Hospital, Yunyang Medical College, Shiyan, Hubei 442000, P.R. China
| | - Hong Zheng
- Laboratory of Molecular Diagnosis of Cancer, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yan-Ping Wang
- Laboratory of Molecular Diagnosis of Cancer, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
26
|
Olcina MM, Kim RK, Melemenidis S, Graves EE, Giaccia AJ. The tumour microenvironment links complement system dysregulation and hypoxic signalling. Br J Radiol 2018; 92:20180069. [PMID: 29544344 PMCID: PMC6435069 DOI: 10.1259/bjr.20180069] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The complement system is an innate immune pathway typically thought of as part of the first line of defence against “non-self” species. In the context of cancer, complement has been described to have an active role in facilitating cancer-associated processes such as increased proliferation, angiogenesis and migration. Several cellular members of the tumour microenvironment express and/or produce complement proteins locally, including tumour cells. Dysregulation of the complement system has been reported in numerous tumours and increased expression of complement activation fragments in cancer patient specimens correlates with poor patient prognosis. Importantly, genetic or pharmacological targeting of complement has been shown to reduce tumour growth in several cancer preclinical models, suggesting that complement could be an attractive therapeutic target. Hypoxia (low oxygen) is frequently found in solid tumours and has a profound biological impact on cellular and non-cellular components of the tumour microenvironment. In this review, we focus on hypoxia since this is a prevailing feature of the tumour microenvironment that, like increased complement, is typically associated with poor prognosis. Furthermore, interesting links between hypoxia and complement have been recently proposed but never collectively reviewed. Here, we explore how hypoxia alters regulation of complement proteins in different cellular components of the tumour microenvironment, as well as the downstream biological consequences of this regulation.
Collapse
Affiliation(s)
- Monica M Olcina
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Ryan K Kim
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | | | - Edward E Graves
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Amato J Giaccia
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| |
Collapse
|
27
|
|
28
|
Lee CH, Romain G, Yan W, Watanabe M, Charab W, Todorova B, Lee J, Triplett K, Donkor M, Lungu OI, Lux A, Marshall N, Lindorfer MA, Goff ORL, Balbino B, Kang TH, Tanno H, Delidakis G, Alford C, Taylor RP, Nimmerjahn F, Varadarajan N, Bruhns P, Zhang YJ, Georgiou G. IgG Fc domains that bind C1q but not effector Fcγ receptors delineate the importance of complement-mediated effector functions. Nat Immunol 2017; 18:889-898. [PMID: 28604720 DOI: 10.1038/ni.3770] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 05/16/2017] [Indexed: 12/17/2022]
Abstract
Engineered crystallizable fragment (Fc) regions of antibody domains, which assume a unique and unprecedented asymmetric structure within the homodimeric Fc polypeptide, enable completely selective binding to the complement component C1q and activation of complement via the classical pathway without any concomitant engagement of the Fcγ receptor (FcγR). We used the engineered Fc domains to demonstrate in vitro and in mouse models that for therapeutic antibodies, complement-dependent cell-mediated cytotoxicity (CDCC) and complement-dependent cell-mediated phagocytosis (CDCP) by immunological effector molecules mediated the clearance of target cells with kinetics and efficacy comparable to those of the FcγR-dependent effector functions that are much better studied, while they circumvented certain adverse reactions associated with FcγR engagement. Collectively, our data highlight the importance of CDCC and CDCP in monoclonal-antibody function and provide an experimental approach for delineating the effect of complement-dependent effector-cell engagement in various therapeutic settings.
Collapse
Affiliation(s)
- Chang-Han Lee
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Gabrielle Romain
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, Texas, USA
| | - Wupeng Yan
- Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, USA
| | - Makiko Watanabe
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Wissam Charab
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Biliana Todorova
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, Paris, France.,INSERM, U760, Paris, France
| | - Jiwon Lee
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Kendra Triplett
- Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, USA
| | - Moses Donkor
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Oana I Lungu
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Anja Lux
- Institute of Genetics, Department of Biology, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Nicholas Marshall
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Margaret A Lindorfer
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Odile Richard-Le Goff
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, Paris, France.,INSERM, U760, Paris, France
| | - Bianca Balbino
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, Paris, France.,INSERM, U760, Paris, France.,Université Pierre et Marie Curie, Paris, France
| | - Tae Hyun Kang
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Hidetaka Tanno
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - George Delidakis
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Corrine Alford
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Ronald P Taylor
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Falk Nimmerjahn
- Institute of Genetics, Department of Biology, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Navin Varadarajan
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, Texas, USA
| | - Pierre Bruhns
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, Paris, France.,INSERM, U760, Paris, France
| | - Yan Jessie Zhang
- Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, USA.,Institute for Cell and Molecular Biology, University of Texas at Austin, Austin, Texas, USA
| | - George Georgiou
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, USA.,Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, USA.,Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, USA.,Institute for Cell and Molecular Biology, University of Texas at Austin, Austin, Texas, USA.,Center for Systems and Synthetic Biology University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
29
|
Salviano-Silva A, Petzl-Erler ML, Boldt ABW. CD59 polymorphisms are associated with gene expression and different sexual susceptibility to pemphigus foliaceus. Autoimmunity 2017; 50:377-385. [DOI: 10.1080/08916934.2017.1329830] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Amanda Salviano-Silva
- Laboratory of Human Molecular Genetics, Department of Genetics, Universidade Federal do Paraná, Curitiba, Brazil
| | - Maria Luiza Petzl-Erler
- Laboratory of Human Molecular Genetics, Department of Genetics, Universidade Federal do Paraná, Curitiba, Brazil
| | - Angelica Beate Winter Boldt
- Laboratory of Human Molecular Genetics, Department of Genetics, Universidade Federal do Paraná, Curitiba, Brazil
- Laboratory of Molecular Immunopathology, Department of Clinical Pathology, Hospital de Clínicas, Universidade Federal do Paraná, Curitiba, Brazil
| |
Collapse
|
30
|
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
|
31
|
Parente R, Clark SJ, Inforzato A, Day AJ. Complement factor H in host defense and immune evasion. Cell Mol Life Sci 2016; 74:1605-1624. [PMID: 27942748 PMCID: PMC5378756 DOI: 10.1007/s00018-016-2418-4] [Citation(s) in RCA: 136] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 11/08/2016] [Accepted: 11/09/2016] [Indexed: 12/30/2022]
Abstract
Complement is the major humoral component of the innate immune system. It recognizes pathogen- and damage-associated molecular patterns, and initiates the immune response in coordination with innate and adaptive immunity. When activated, the complement system unleashes powerful cytotoxic and inflammatory mechanisms, and thus its tight control is crucial to prevent damage to host tissues and allow restoration of immune homeostasis. Factor H is the major soluble inhibitor of complement, where its binding to self markers (i.e., particular glycan structures) prevents complement activation and amplification on host surfaces. Not surprisingly, mutations and polymorphisms that affect recognition of self by factor H are associated with diseases of complement dysregulation, such as age-related macular degeneration and atypical haemolytic uremic syndrome. In addition, pathogens (i.e., non-self) and cancer cells (i.e., altered-self) can hijack factor H to evade the immune response. Here we review recent (and not so recent) literature on the structure and function of factor H, including the emerging roles of this protein in the pathophysiology of infectious diseases and cancer.
Collapse
Affiliation(s)
- Raffaella Parente
- Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, 20089, Milan, Italy
| | - Simon J Clark
- Division of Evolution and Genomic Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - Antonio Inforzato
- Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, 20089, Milan, Italy. .,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Via Vanvitelli 32, 20129, Milan, Italy.
| | - Anthony J Day
- Wellcome Trust Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, M13 9PT, UK.
| |
Collapse
|
32
|
Meri S. Self-nonself discrimination by the complement system. FEBS Lett 2016; 590:2418-34. [PMID: 27393384 DOI: 10.1002/1873-3468.12284] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 06/29/2016] [Accepted: 07/06/2016] [Indexed: 01/09/2023]
Abstract
The alternative pathway (AP) of complement can recognize nonself structures by only two molecules, C3b and factor H. The AP deposits C3b covalently on nonself structures via an amplification system. The actual discrimination is performed by factor H, which has binding sites for polyanions (sialic acids, glycosaminoglycans, phospholipids). This robust recognition of 'self' protects our own intact viable cells and tissues, while activating structures are recognized by default. Foreign targets are opsonized for phagocytosis or killed. Mutations in factor H predispose to severe diseases. In hemolytic uremic syndrome, they promote complement attack against blood cells and vascular endothelial cells and lead, for example, to kidney and brain damage. Even pathogens can exploit factor H. In fact, the ability to bind factor H discriminates most pathogenic microbes from nonpathogenic ones.
Collapse
Affiliation(s)
- Seppo Meri
- Immunobiology, Research Programs Unit, Department of Bacteriology and Immunology, Haartman Institute, University of Helsinki, Finland.,HUSLAB, Helsinki University Hospital, Finland.,Humanitas University, Milan, Italy
| |
Collapse
|
33
|
Nissinen L, Farshchian M, Riihilä P, Kähäri VM. New perspectives on role of tumor microenvironment in progression of cutaneous squamous cell carcinoma. Cell Tissue Res 2016; 365:691-702. [PMID: 27411692 DOI: 10.1007/s00441-016-2457-z] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 06/16/2016] [Indexed: 12/29/2022]
Abstract
Epidermal keratinocyte-derived cutaneous squamous cell carcinoma (cSCC) is the most common metastatic skin cancer, and its incidence is increasing worldwide. Solar UV radiation is an important risk factor for cSCC and leads to genetic and epigenetic changes both in epidermal keratinocytes and dermal cells. Tumor cells in cutaneous cSCCs typically harbor several driver gene mutations, but epidermal keratinocytes in sun-exposed normal skin also contain mutations in these same genes. Therefore, alterations in the microenvironment of premalignant lesions are evidently required for their progression to invasive and metastatic cSCC. For example, alterations in the composition of basement membrane and dermal extracellular matrix are early events in cSCC progression. The presence of microbial structures and the influx of inflammatory cells promote the secretion of proteases, which in turn regulate the availability of growth factors, cytokines, and chemokines and thus influence the growth and invasion of cSCC. Together, these observations emphasize the role of the tumor microenvironment in the progression of cSCC and identify it as a novel therapeutic target in cSCC and other malignant tumors. Graphical abstract Tumor-stroma interactions in the progression of cutaneous squamous cell carcinoma (cSCC). Epidermal layer is separated by a well-organized basement membrane (BM) from the dermal layer. UV radiation, other environmental insults, and aging target both epidermal keratinocytes and dermal fibroblasts and lead to genetic and epigenetic changes in these cells. In addition, epidermal keratinocytes in normal sun-exposed skin harbor several mutations in the cSCC driver genes. During transition to premalignant actinic keratosis (AK), the differentiation of keratinocytes is disturbed resulting in a neoplastic epithelium with hyperplastic cells. Expression of proteinases, such as matrix metalloproteinases (MMP) by neoplastic cells and activated stromal fibroblasts and macrophages is induced in AK, and collagen XV and XVIII are lost from the dermal BM. Furthermore, inflammatory cells accumulate at the site of the hyperplastic epithelium. During a later stage of cSCC progression, the number of inflammatory cells increases, and the expression of complement components and inhibitors by tumor cells is induced (CFI complement factor I, CFH complement factor H, FHL-1 Factor H-like protein 1). In addition to MMPs, activated fibroblasts also produce growth factors and promote inflammation, growth, and invasion of tumor cells.
Collapse
Affiliation(s)
- Liisa Nissinen
- The Department of Dermatology, University of Turku and Turku University Hospital, P.O.B 52, FI-20521, Turku, Finland.,MediCity Research Laboratory University of Turku, Turku, Finland
| | - Mehdi Farshchian
- The Department of Dermatology, University of Turku and Turku University Hospital, P.O.B 52, FI-20521, Turku, Finland.,MediCity Research Laboratory University of Turku, Turku, Finland
| | - Pilvi Riihilä
- The Department of Dermatology, University of Turku and Turku University Hospital, P.O.B 52, FI-20521, Turku, Finland.,MediCity Research Laboratory University of Turku, Turku, Finland
| | - Veli-Matti Kähäri
- The Department of Dermatology, University of Turku and Turku University Hospital, P.O.B 52, FI-20521, Turku, Finland. .,MediCity Research Laboratory University of Turku, Turku, Finland.
| |
Collapse
|
34
|
Xi W, Liu L, Wang J, Xia Y, Bai Q, Long Q, Wang Y, Xu J, Guo J. High Level of Anaphylatoxin C5a Predicts Poor Clinical Outcome in Patients with Clear Cell Renal Cell Carcinoma. Sci Rep 2016; 6:29177. [PMID: 27381421 PMCID: PMC4933887 DOI: 10.1038/srep29177] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 06/08/2016] [Indexed: 12/18/2022] Open
Abstract
Anaphylatoxin C5a, a potent pro-inflammatory peptide produced in the process of complement activation, was proved to have a vital role in tumor initiation and progession by previous investigations. However whether it could act as a prognostic marker remains unknown. Here we retrospectively enrolled 272 ccRCC patients undergoing nephrectomy in Zhongshan Hospital, Shanghai between 2005 and 2007. C5a level was assessed by immunohistochemistry and its association with clinicopathologic features and prognosis were evaluated. Our results indicated that high tumoral C5a level was associated with poor overall survival (OS) (hazard ratio = 1.753, 95% CI 1.068–2.878, P = 0.026). In addition, tumoral C5a could significantly stratify patients’ prognosis both in advanced stage (TNM III + IV) and intermediate/high risk group (SSIGN score ≥4) (P < 0.001 and = 0.008, respectively). Furthermore, incorporating tumoral C5a with other parameters could improve the predicting accuracy, compared with TNM and SSIGN system (c-index = 0.789, 0.713 and 0.727, respectively). In conclusion, tumoral C5a is an independent adverse prognostic biomarker for clinical outcome of ccRCC patients after nephectomy.
Collapse
Affiliation(s)
- Wei Xi
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Li Liu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jiajun Wang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yu Xia
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qi Bai
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qilai Long
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yiwei Wang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jiejie Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Jianming Guo
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
35
|
Fujinami N, Yoshikawa T, Sawada Y, Shimomura M, Iwama T, Sugai S, Kitano S, Uemura Y, Nakatsura T. Enhancement of antitumor effect by peptide vaccine therapy in combination with anti-CD4 antibody: Study in a murine model. Biochem Biophys Rep 2016; 5:482-491. [PMID: 28955856 PMCID: PMC5600353 DOI: 10.1016/j.bbrep.2016.02.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 02/17/2016] [Accepted: 02/17/2016] [Indexed: 01/30/2023] Open
Abstract
Purpose The clinical efficacy of cancer peptide vaccine therapy is insufficient. To enhance the anti-tumor effect of peptide vaccine therapy, we combined this therapy with an anti-CD4 mAb (GK1.5), which is known to deplete CD4+ cells, including regulatory T cells (Tregs). Methods To determine the treatment schedule, the number of lymphocyte subsets in the peripheral blood of mice was traced by flow cytometry after administration of anti-CD4 mAb. The ovalbumin (OVA)257–264 peptide vaccine was injected intradermally and anti-CD4 mAb was administered intraperitoneally into C57BL/6 mice at different schedules. We evaluated the enhancement of OVA peptide-specific cytotoxic T lymphocyte (CTL) induction in the combination therapy using the ELISPOT assay, CD107a assay, and cytokine assay. We then examined the in vivo metastasis inhibitory effect by OVA peptide vaccine therapy in combination with anti-CD4 mAb against OVA-expressing thymoma (EG7) in a murine liver metastatic model. Results We showed that peptide-specific CTL induction was enhanced by the peptide vaccine in combination with anti-CD4 mAb and that the optimized treatment schedule had the strongest induction effect of peptide-specific CTLs using an IFN-γ ELISPOT assay. We also confirmed that the CD107a+ cells secreted perforin and granzyme B and the amount of IL-2 and TNF produced by these CTLs increased when the peptide vaccine was combined with anti-CD4 mAb. Furthermore, metastasis was inhibited by peptide vaccines in combination with anti-CD4 mAb compared to peptide vaccine alone in a murine liver metastatic model. Conclusion The use of anti-CD4 mAb in combination with the OVA peptide vaccine therapy increased the number of peptide-specific CTLs and showed a higher therapeutic effect against OVA-expressing tumors. The combination with anti-CD4 mAb may provide a new cancer vaccine strategy. Peptide-specific CTL induction and function were enhanced by depletion of CD4+ cells. Anti-tumor effect by the peptide vaccine was enhanced by the depletion of CD4+ cells. Metastasis was inhibited by vaccine with depletion of CD4+ cells in a murine model. Combination with the depletion of CD4+ cells could be a new cancer vaccine strategy.
Collapse
Key Words
- 7-AAD, 7-amino-actinomycin D
- Anti-CD4 antibody
- CTL, cytotoxic T lymphocyte
- Cancer
- DC, dendritic cell
- ELISPOT assay, enzyme-linked immunospot assay
- FITC, fluorescein isothiocyanate
- FOXP3, forkhead box P3
- GPC3, glypican-3
- HCC, hepatocellular carcinoma
- IFN-γ, interferon-γ
- IL-2, interleukine-2
- Immunotherapy
- MHC, major histocompatibility complex
- Murine liver metastatic model
- OVA, ovalbumin
- PD-1, programmed death-1
- PE, phycoerythrin
- Peptide vaccine
- QOL, quality of life
- TGF-β, transforming growth factor-βl
- TNF, tumor necrosis factor
- Treg, regulatory T cell
- mAb, monoclonal antibody
Collapse
Affiliation(s)
- Norihiro Fujinami
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center National Cancer Center, Kashiwa, Chiba, Japan.,Research Institute for Biomedical Sciences, Tokyo University of Science, Japan
| | - Toshiaki Yoshikawa
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center National Cancer Center, Kashiwa, Chiba, Japan
| | - Yu Sawada
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center National Cancer Center, Kashiwa, Chiba, Japan.,Department of Gastroenterological Surgery, Graduate School of Medicine, Yokohama City University, Yokohama, Kanagawa, Japan
| | - Manami Shimomura
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center National Cancer Center, Kashiwa, Chiba, Japan
| | - Tatsuaki Iwama
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center National Cancer Center, Kashiwa, Chiba, Japan
| | - Shiori Sugai
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center National Cancer Center, Kashiwa, Chiba, Japan.,Research Institute for Biomedical Sciences, Tokyo University of Science, Japan
| | - Shigehisa Kitano
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center National Cancer Center, Kashiwa, Chiba, Japan.,Department of Experimental Therapeutics, National Cancer Center Hospital, Tsukiji, Tokyo, Japan
| | - Yasushi Uemura
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center National Cancer Center, Kashiwa, Chiba, Japan
| | - Tetsuya Nakatsura
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center National Cancer Center, Kashiwa, Chiba, Japan.,Research Institute for Biomedical Sciences, Tokyo University of Science, Japan
| |
Collapse
|
36
|
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: 85] [Impact Index Per Article: 8.5] [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
|
37
|
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: 1.1] [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
|
38
|
Xu H, Tian YN, Dun BY, Liu HT, Dong GK, Wang JH, Lu SS, Chen B, She JX. A novel monoclonal antibody induces cancer cell apoptosis and enhances the activity of chemotherapeutic drugs. Asian Pac J Cancer Prev 2015; 15:4423-8. [PMID: 24969863 DOI: 10.7314/apjcp.2014.15.11.4423] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
A novel monoclonal antibody (mAb), known as AC10364, was identified from an antibody library generated by immunization of mice with human carcinoma cells. The mAb recognized proteins in lysates from multiple carcinoma cell lines. Cell cytotoxicity assays showed that AC10364 significantly inhibited cell growth and induced apoptosis in multiple carcinoma cell lines, including Bel/fu, KATO-III and A2780. Compared with mAb AC10364 or chemotherapeutic drugs alone, the combination of mAb AC10364 with chemotherapeutic drugs demonstrated enhanced growth inhibitory effects on carcinoma cells. These results suggest that mAb AC10364 is a promising candidate for cancer therapy.
Collapse
Affiliation(s)
- Heng Xu
- Institute of Translational Medicine, School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, China E-mail :
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Mamidi S, Höne S, Teufel C, Sellner L, Zenz T, Kirschfink M. Neutralization of membrane complement regulators improves complement-dependent effector functions of therapeutic anticancer antibodies targeting leukemic cells. Oncoimmunology 2015; 4:e979688. [PMID: 25949896 PMCID: PMC4404820 DOI: 10.4161/2162402x.2014.979688] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 10/16/2014] [Accepted: 10/16/2014] [Indexed: 01/12/2023] Open
Abstract
Complement-dependent cytotoxicity (CDC) is one of the effector mechanisms mediated by therapeutic anticancer monoclonal antibodies (mAbs). However, the efficacy of antibodies is limited by the resistance of malignant cells to complement attack, primarily due to the over-expression of one or more membrane complement regulatory proteins (mCRPs) CD46, CD55, and CD59. CD20-positive Burkitt lymphoma Raji cells and primary CLL cells are resistant to rituximab (RTX)-induced CDC whereas ofatumumab (OFA) proved to be more efficient in cell killing. Primary CLL cells but not CD52-positive acute lymphoblastic leukemia (ALL) REH cells were sensitive to alemtuzumab (ALM)-induced CDC. Upon combined inhibition on Raji and CLL cells by mCRPs-specific siRNAs or neutralizing antibodies, CDC induced by RTX and by OFA was augmented. Similarly, CDC of REH cells was enhanced after mCRPs were inhibited upon treatment with ALM. All mAbs induced C3 opsonization, which was significantly augmented upon blocking mCRPs. C3 opsonization led to enhanced cell-mediated cytotoxicity of leukemia cells exposed to PBLs or macrophages. Furthermore, opsonized CLL cells were efficiently phagocytized by macrophages. Our results provide conclusive evidence that inhibition of mCRPs expression sensitizes leukemic cells to complement attack thereby enhancing the therapeutic effect of mAbs targeting leukemic cells.
Collapse
Key Words
- ADCC, antibody-dependent cellular cytotoxicity
- ALM, Alemtuzumab
- CDC, complement-dependent cytotoxicity
- CDCC, complement-dependent cellular cytotoxicity
- MAC, membrane attack complex
- NHS, Normal Human Serum
- OFA, Ofatumumab
- PBLs, peripheral blood leukocytes
- RTX, Rituximab
- TRX, Trastuzumab
- alemtuzumab
- chronic lymphocytic leukemia
- complement regulatory proteins
- complement-dependent cytotoxicity
- mCRP, membrane-bound complement regulatory protein
- ofatumumab
- opsonization
- rituximab
- siRNA, small interfering RNA
Collapse
Affiliation(s)
- Srinivas Mamidi
- Institute for Immunology; University of Heidelberg; Heidelberg, Germany
| | - Simon Höne
- Institute for Immunology; University of Heidelberg; Heidelberg, Germany
| | - Claudia Teufel
- Institute for Immunology; University of Heidelberg; Heidelberg, Germany
| | - Leopold Sellner
- Department of Translational Oncology; National Center for Tumour Diseases (NCT) and German Cancer Research Center (DKFZ); Heidelberg, Germany
- Department of Medicine V; University of Heidelberg; Heidelberg, Germany
| | - Thorsten Zenz
- Department of Translational Oncology; National Center for Tumour Diseases (NCT) and German Cancer Research Center (DKFZ); Heidelberg, Germany
- Department of Medicine V; University of Heidelberg; Heidelberg, Germany
| | | |
Collapse
|
40
|
Sharma S, Ray S, Moiyadi A, Sridhar E, Srivastava S. Quantitative proteomic analysis of meningiomas for the identification of surrogate protein markers. Sci Rep 2014; 4:7140. [PMID: 25413266 PMCID: PMC5382771 DOI: 10.1038/srep07140] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 09/15/2014] [Indexed: 12/11/2022] Open
Abstract
Meningiomas are the most common non-glial tumors of the brain and spine. Pathophysiology and definite histological grading of meningiomas are frequently found to be deceptive due to their unusual morphological features and locations. Here for the first time we report a comprehensive serum proteomic analysis of different grades of meningiomas by using multiple quantitative proteomic and immunoassay-based approaches to obtain mechanistic insights about disease pathogenesis and identify grade specific protein signatures. In silico functional analysis revealed modulation of different vital physiological pathways including complement and coagulation cascades, metabolism of lipids and lipoproteins, immune signaling, cell growth and apoptosis and integrin signaling in meningiomas. ROC curve analysis demonstrated apolipoprotein E and A-I and hemopexin as efficient predictors for meningiomas. Identified proteins like vimentin, alpha-2-macroglobulin, apolipoprotein B and A-I and antithrombin-III, which exhibited a sequential increase in different malignancy grades of meningiomas, could serve as potential predictive markers.
Collapse
Affiliation(s)
- Samridhi Sharma
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Sandipan Ray
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Aliasgar Moiyadi
- Department of Neurosurgery, Advanced Center for Treatment Research and Education in Cancer, Tata Memorial Center, Kharghar, Navi Mumbai 410210, India
| | - Epari Sridhar
- Department of Pathology, Tata Memorial Hospital, Mumbai 400012, India
| | - Sanjeeva Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| |
Collapse
|
41
|
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: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 10/29/2014] [Indexed: 01/08/2023]
|
42
|
Shang Y, Chai N, Gu Y, Ding L, Yang Y, Zhou J, Ren G, Hao X, Fan D, Wu K, Nie Y. Systematic immunohistochemical analysis of the expression of CD46, CD55, and CD59 in colon cancer. Arch Pathol Lab Med 2014; 138:910-9. [PMID: 24978917 DOI: 10.5858/arpa.2013-0064-oa] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT The expression of membrane-bound complement regulatory proteins (mCRPs) that inhibit the complement system in normal tissues is essential for self-protection against an autologous immune reaction. However, the expression patterns of mCRPs, including CD46, CD55, and CD59, are inconsistent in different types of cancer cells. OBJECTIVES To determine whether CD46, CD55, and CD59 are differentially expressed in neoplastic and adjacent normal colon tissues and to assess their clinical significance. DESIGN Immunohistochemistry was performed on tissue microarrays of cancerous and adjacent normal colon tissues. RESULTS The expression levels of CD46, CD55, and CD59 were significantly higher in colon cancer tissues compared with the normal adjacent colon tissues. We found that the expression levels of CD55 and CD59 correlated with the grade of differentiation in colon cancers. In addition, the expression of CD55 and CD59 was greater in stage III and stage IV colon cancers than in stage I and stage II cancers according to staging by the TNM classification. CONCLUSIONS CD46, CD55, and CD59 are up-regulated in colon cancer. Specifically, CD55 and CD59 are of clinical relevance to differentiation and TNM staging of colon cancer. These data suggest that CD46, CD55, and CD59 have the potential to be used for molecular staging diagnoses and for colon cancer therapies.
Collapse
Affiliation(s)
- Yulong Shang
- From the State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases (Drs Shang, Chai, Zhou, Ren, Fan, Wu, and Nie and Mr Gu, Mss Ding and Yang), and the Department of Medical Laboratory, Xijing Hospital (Dr Hao and Ms Ding), Fourth Military Medical University, and the Department of Digestive Diseases, Shaanxi Provincial Corps Hospital of Chinese People's Armed Police Force (Mr Gu), Xi'an, China. Drs Shang, Chai, and Mr Gu contributed equally to this article
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Complement factor I promotes progression of cutaneous squamous cell carcinoma. J Invest Dermatol 2014; 135:579-588. [PMID: 25184960 DOI: 10.1038/jid.2014.376] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 07/11/2014] [Accepted: 07/11/2014] [Indexed: 01/25/2023]
Abstract
The incidence of cutaneous squamous cell carcinoma (cSCC) is rising worldwide. We have examined the role of complement components in the progression of cSCC. Analysis of cSCC cell lines (n=8) and normal human epidermal keratinocytes (n=11) with whole transcriptome profiling (SOLiD), quantitative real-time reverse transcriptase-PCR, and western blotting revealed marked overexpression of complement factor I (CFI) in cSCC cells. Immunohistochemical analysis for CFI in vivo showed stronger tumor cell-specific labeling intensity in invasive sporadic cSCCs (n=83) and recessive dystrophic epidermolysis bullosa-associated cSCCs (n=7) than in cSCC in situ (n=65), premalignant epidermal lesions (actinic keratoses, n=64), benign epidermal papillomas (seborrheic keratoses, n=39), and normal skin (n=9). The expression of CFI was higher in the aggressive Ha-ras-transformed cell line (RT3) than in less tumorigenic HaCaT cell lines (HaCaT, A5, and II-4). The expression of CFI by cSCC cells was upregulated by IFN-γ and IL-1β. Knockdown of CFI expression inhibited proliferation and migration of cSCC cells and resulted in inhibition of basal extracellular signal-regulated kinase (ERK) 1/2 activation. Knockdown of CFI expression potently inhibited growth of human cSCC xenograft tumors in vivo. These results provide evidence for the role of CFI in the progression of cSCC and identify it as a potential therapeutic target in this nonmelanoma skin cancer.
Collapse
|
44
|
Wu Y, Wang Y, Qin F, Wang Z, Wang Y, Yang Y, Zheng H, Wang Y. CD55 limits sensitivity to complement-dependent cytolysis triggered by heterologous expression of α-gal xenoantigen in colon tumor cells. Am J Physiol Gastrointest Liver Physiol 2014; 306:G1056-64. [PMID: 24763553 DOI: 10.1152/ajpgi.00464.2013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Engineering cancer cells to express heterologous antigen α-gal and induce the destruction of tumor cells depending on the complement cascade may be a promising strategy of tumor therapy. However, the feasibility and effect of using α-gal to induce colorectal adenocarcinoma cell line cytolysis is not yet known. In this study, we evaluated α-gal expression's ability to sensitize human colorectal adenocarcinoma cell lines to complement attack in cell lines LoVo, SW620, and Ls-174T. Nearly all α-gal-expressing LoVo and SW620 cells were killed by normal human serum (NHS), but α-gal-expressing Ls-174T cells showed no significant lysis. We analyzed the expression levels of membrane-bound complement regulatory proteins (mCRPs) on the three cell lines, and their protective role in α-gal-mediated activation of the complement. LoVo showed no expression of any of the three proteins. CD59 was strongly expressed by SW620 and Ls-174T. CD46 and CD55 varied between the two cell lines. CD46 on SW620 was only half the intensity of CD46 on Ls-174T. Ls-174T showed a notable expression of CD55, while expression of CD55 on SW620 was not detected. The sensitivity of Ls-174T expressing α-gal to NHS greatly increased following the downregulation of CD46 and CD55 with short hairpin RNA (shRNA). However, there is no increase in cell killing when CD59 expression was diminished. Our findings suggest that the use of α-gal as antigen to induce tumor cell killing may be a potential therapeutic strategy in colon cancer and that CD55 plays a primary role in conferring resistance to lysis.
Collapse
Affiliation(s)
- Yanxia Wu
- Laboratory of Molecular Diagnosis of Cancer, West China Hospital, Sichuan University, Chengdu, China; State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yaogeng Wang
- Laboratory of Molecular Diagnosis of Cancer, West China Hospital, Sichuan University, Chengdu, China; State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Feng Qin
- Basic Medical Faculty, Dali Medical College, Dali, China; and
| | - Zhu Wang
- Laboratory of Molecular Diagnosis of Cancer, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Wang
- Laboratory of Molecular Diagnosis of Cancer, West China Hospital, Sichuan University, Chengdu, China
| | | | - Hong Zheng
- Laboratory of Molecular Diagnosis of Cancer, West China Hospital, Sichuan University, Chengdu, China; State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yanping Wang
- Laboratory of Molecular Diagnosis of Cancer, West China Hospital, Sichuan University, Chengdu, China; State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China;
| |
Collapse
|
45
|
Kesselring R, Thiel A, Pries R, Fichtner-Feigl S, Brunner S, Seidel P, Bruchhage KL, Wollenberg B. The complement receptors CD46, CD55 and CD59 are regulated by the tumour microenvironment of head and neck cancer to facilitate escape of complement attack. Eur J Cancer 2014; 50:2152-61. [PMID: 24915776 DOI: 10.1016/j.ejca.2014.05.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 05/05/2014] [Accepted: 05/09/2014] [Indexed: 12/26/2022]
Abstract
BACKGROUND Membrane-bound complement restriction proteins (mCRPs) CD46, CD55 and CD59 enable tumour cells to evade complement dependent cytotoxicity and antibody-dependent killing mechanisms. But less is known about the role of these mCRPs in head and neck cancer. METHODS In this study we determined the expression of the mCRPs on head and neck squamous cell carcinoma (HNSCC) cell lines, on tumour tissue and TDLNs (tumour-draining lymph nodes) as well as on lymphocytes from HNSCC patients. The influence of the HNSCC microenvironment on the mCRP regulation was analysed using Flow Cytometry, Western blotting and small interfering RNAs (siRNA) transfection studies. RESULTS We examined the effects of the HNSCC tumour milieu on the expression levels of CD46, CD55 and CD59. We investigated the susceptibility of HNSCC cells to CDC (complement-dependent cytotoxicity) while silencing the mCRPs. Our results demonstrate a huge influence of the HNSCC tumour microenvironment on the regulation of mCRP expression and show a reciprocal regulation between the different mCRPs themselves. CONCLUSIONS In summary, our data indicate that HNSCC has evolved different strategies to evade complement attacks and that the tumour microenvironment leads to the enhancement of complement resistance of the surrounding tissue.
Collapse
Affiliation(s)
- Rebecca Kesselring
- Department of Otorhinolaryngology and Plastic Surgery, University of Luebeck, Ratzeburger Allee 160, 23562 Luebeck, Germany; Department of Surgery, University Medical Center Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Annette Thiel
- Department of Otorhinolaryngology and Plastic Surgery, University of Luebeck, Ratzeburger Allee 160, 23562 Luebeck, Germany; Department of Internal Medicine, University Medical Center Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Ralph Pries
- Department of Otorhinolaryngology and Plastic Surgery, University of Luebeck, Ratzeburger Allee 160, 23562 Luebeck, Germany
| | - Stefan Fichtner-Feigl
- Department of Surgery, University Medical Center Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Stefan Brunner
- Department of Surgery, University Medical Center Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Philipp Seidel
- Department of Otorhinolaryngology and Plastic Surgery, University of Luebeck, Ratzeburger Allee 160, 23562 Luebeck, Germany
| | - Karl-Ludwig Bruchhage
- Department of Otorhinolaryngology and Plastic Surgery, University of Luebeck, Ratzeburger Allee 160, 23562 Luebeck, Germany
| | - Barbara Wollenberg
- Department of Otorhinolaryngology and Plastic Surgery, University of Luebeck, Ratzeburger Allee 160, 23562 Luebeck, Germany.
| |
Collapse
|
46
|
Abstract
The complement terminal pathway clears pathogens by generating cytotoxic membrane attack complex (MAC) pores on target cells. For more than 40 years, biochemical and cellular assays have been used to characterize the lytic nature of the MAC and to define its protein composition. Although models for pore formation have been inferred from structures of bacterial cytolysins, it was only recently that we were able to visualize how complement components come together during MAC assembly. This review highlights structural analyses of terminal pathway complexes to explore molecular mechanisms underlying MAC formation.
Collapse
Affiliation(s)
- Doryen Bubeck
- Department of Life Sciences, Sir Ernst Chain Building, South Kensington Campus, Imperial College London , London SW7 2AZ, U.K
| |
Collapse
|
47
|
Cai B, Xie S, Liu F, Simone LC, Caplan S, Qin X, Naslavsky N. Rapid degradation of the complement regulator, CD59, by a novel inhibitor. J Biol Chem 2014; 289:12109-12125. [PMID: 24616098 DOI: 10.1074/jbc.m113.547083] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
There is increased interest in immune-based monoclonal antibody therapies for different malignancies because of their potential specificity and limited toxicity. The activity of some therapeutic monoclonal antibodies is partially dependent on complement-dependent cytolysis (CDC), in which the immune system surveys for invading pathogens, infected cells, and malignant cells and facilitates their destruction. CD59 is a ubiquitously expressed cell-surface glycosylphosphatidylinositol-anchored protein that protects cells from CDC. However, in certain tumors, CD59 expression is enhanced, posing a significant obstacle for treatment, by hindering effective monoclonal antibody-induced CDC. In this study, we used non-small lung carcinoma cells to characterize the mechanism of a novel CD59 inhibitor: the 114-amino acid recombinant form of the 4th domain of intermedilysin (rILYd4), a pore forming toxin secreted by Streptococcus intermedius. We compared the rates of internalization of CD59 in the presence of rILYd4 or anti-CD59 antibodies and determined that rILYd4 induces more rapid CD59 uptake at early time points. Most significantly, upon binding to rILYd4, CD59 is internalized and undergoes massive degradation in lysosomes within minutes. The remaining rILYd4·CD59 complexes recycle to the PM and are shed from the cell. In comparison, upon internalization of CD59 via anti-CD59 antibody binding, the antibody·CD59 complex is recycled via early and recycling endosomes, mostly avoiding degradation. Our study supports a novel role for rILYd4 in promoting internalization and rapid degradation of the complement inhibitor CD59, and highlights the potential for improving CDC-based immunotherapy.
Collapse
Affiliation(s)
- Bishuang Cai
- Department of Biochemistry and Molecular Biology and the Fred and Pamela Buffett Cancer Center, The University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Shuwei Xie
- Department of Biochemistry and Molecular Biology and the Fred and Pamela Buffett Cancer Center, The University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Fengming Liu
- Department of Neuroscience and Center for Neurovirology, Temple University School of Medicine, Philadelphia, Pennsylvania 19140
| | - Laura C Simone
- Department of Biochemistry and Molecular Biology and the Fred and Pamela Buffett Cancer Center, The University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Steve Caplan
- Department of Biochemistry and Molecular Biology and the Fred and Pamela Buffett Cancer Center, The University of Nebraska Medical Center, Omaha, Nebraska 68198.
| | - Xuebin Qin
- Department of Neuroscience and Center for Neurovirology, Temple University School of Medicine, Philadelphia, Pennsylvania 19140.
| | - Naava Naslavsky
- Department of Biochemistry and Molecular Biology and the Fred and Pamela Buffett Cancer Center, The University of Nebraska Medical Center, Omaha, Nebraska 68198.
| |
Collapse
|
48
|
Kaufman KL, Mactier S, Armstrong NJ, Mallawaaratchy D, Byrne SN, Haydu LE, Jakrot V, Thompson JF, Mann GJ, Scolyer RA, Christopherson RI. Surface antigen profiles of leukocytes and melanoma cells in lymph node metastases are associated with survival in AJCC stage III melanoma patients. Clin Exp Metastasis 2014; 31:407-21. [PMID: 24435119 PMCID: PMC3973954 DOI: 10.1007/s10585-014-9636-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 01/09/2014] [Indexed: 12/14/2022]
Abstract
There is an urgent need to identify more accurate prognostic biomarkers in melanoma patients, particularly in those with metastatic disease. This study aimed to identify melanoma and leukocyte surface antigens predictive of survival in a prospective series of AJCC stage IIIb/c melanoma patients (n = 29). Live cell suspensions were prepared from melanoma metastases within lymph nodes (LN). The suspensions were immuno-magnetically separated into CD45+ (leukocyte) and CD45− (non-hematopoietic, enriched melanoma cell) fractions. Surface antigens on CD45− and CD45+ cell populations were profiled using DotScan™ microarrays (Medsaic Pty. Ltd.) and showed differential abundance levels for 52 and 78 antigens respectively. Associations of the surface profiles with clinicopathologic and outcome data (median follow-up 35.4 months post LN resection) were sought using univariate (log-rank test) and multivariate (Wald’s test; modelled with patient’s age, gender and AJCC staging at LN recurrence) survival models. CD9 (p = 0.036), CD39 (p = 0.004) and CD55 (p = 0.005) on CD45+ leukocytes were independently associated with distant metastasis-free survival using multivariate analysis. Leukocytes with high CD39 levels were also significantly associated with increased overall survival (OS) in multivariate analysis (p = 0.016). LNs containing leukocytes expressing CD11b (p = 0.025), CD49d (p = 0.043) and CD79b (p = 0.044) were associated with reduced OS on univariate analysis. For enriched melanoma cells (CD45− cell populations), 11 surface antigens were significantly correlated with the disease-free interval (DFI) between diagnosis of culprit primary melanoma and LN metastasis resection. Nine antigens on CD45+ leukocytes also correlated with DFI. Following validation in independent datasets, surface markers identified here should enable more accurate determination of prognosis in stage III melanoma patients and provide better risk stratification of patients entering clinical trials.
Collapse
Affiliation(s)
- Kimberley L Kaufman
- School of Molecular Bioscience, University of Sydney, Sydney, NSW, 2006, Australia.
| | - Swetlana Mactier
- School of Molecular Bioscience, University of Sydney, Sydney, NSW, 2006, Australia
| | - Nicola J Armstrong
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.,School of Mathematics and Statistics and Prince of Wales Clinical School, University of New South Wales, Kensington, NSW, 2052, Australia
| | | | - Scott N Byrne
- Discipline of Infectious Diseases and Immunology Sydney Medical School, University of Sydney, Sydney, NSW, 2006, Australia.,Discipline of Dermatology, Bosch Institute, Faculty of Medicine, Sydney Medical School, Sydney, NSW, 2006, Australia
| | - Lauren E Haydu
- Melanoma Institute Australia, North Sydney, NSW, 2060, Australia.,Discipline of Surgery, Sydney Medical School, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Valerie Jakrot
- Melanoma Institute Australia, North Sydney, NSW, 2060, Australia
| | - John F Thompson
- Melanoma Institute Australia, North Sydney, NSW, 2060, Australia
| | - Graham J Mann
- Melanoma Institute Australia, North Sydney, NSW, 2060, Australia.,Westmead Institute of Cancer Research, The University of Sydney at Westmead Millennium Institute, Westmead, NSW, 2145, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, North Sydney, NSW, 2060, Australia.,Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, NSW, 2050, Australia.,Discipline of Pathology, Sydney Medical School, The University of Sydney, Sydney, NSW, 2006, Australia
| | | |
Collapse
|
49
|
Yang X, Guo Z, Liu Y, Si T, Yu H, Li B, Tian W. Prostate stem cell antigen and cancer risk, mechanisms and therapeutic implications. Expert Rev Anticancer Ther 2014; 14:31-37. [PMID: 24308679 DOI: 10.1586/14737140.2014.845372] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2023]
Abstract
Prostate stem cell antigen (PSCA) was originally identified as a tumor antigen in prostate cancer. Recent studies indicated that PSCA was correlated with many cancer types. In this review, we will consider the origin of PSCA, discuss the expression of PSCA in normal and cancer tissue, describe PSCA polymorphisms and cancer risk, summarize potential mechanisms for PSCA involvement in cancer; and look into the therapeutic implications of PSCA. PSCA is upregulated in prostate cancer, pancreatic cancer and bladder cancer, as well as a number of others, making it an ideal clinical target for both diagnosis and therapy. Future studies will be required to explore its mechanisms on various cancer types, and to confirm its clinical utility for diagnosis and immunotherapy strategies. The study of PSCA regulation and expression may also provide information on normal prostate development and prostate carcinogenesis.
Collapse
Affiliation(s)
- Xueling Yang
- Department of Interventional Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | | | | | | | | | | | | |
Collapse
|
50
|
Pio R, Corrales L, Lambris JD. The role of complement in tumor growth. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 772:229-62. [PMID: 24272362 DOI: 10.1007/978-1-4614-5915-6_11] [Citation(s) in RCA: 152] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Complement is a central part of the immune system that has developed as a first defense against non-self cells. Neoplastic transformation is accompanied by an increased capacity of the malignant cells to activate complement. In fact, clinical data demonstrate complement activation in cancer patients. On the basis of the use of protective mechanisms by malignant cells, complement activation has traditionally been considered part of the body's immunosurveillance against cancer. Inhibitory mechanisms of complement activation allow cancer cells to escape from complement-mediated elimination and hamper the clinical efficacy of monoclonal antibody-based cancer immunotherapies. To overcome this limitation, many strategies have been developed with the goal of improving complement-mediated effector mechanisms. However, significant work in recent years has identified new and surprising roles for complement activation within the tumor microenvironment. Recent reports suggest that complement elements can promote tumor growth in the context of chronic inflammation. This chapter reviews the data describing the role of complement activation in cancer immunity, which offers insights that may aid the development of more effective therapeutic approaches to control cancer.
Collapse
Affiliation(s)
- Ruben Pio
- Oncology Division (CIMA), and Department of Biochemistry and Genetics (School of Science), University of Navarra, Pamplona, Spain,
| | | | | |
Collapse
|