1
|
Guigue PA, Brezinov Y, Yasmeen A, Mbarik M, Salvador S, Lau S, Gotlieb WH, Brodeur MN. Ascites and Serum Interleukin-10 Levels as a Prognostic Tool for Ovarian Cancer Outcomes. Cancers (Basel) 2024; 16:2840. [PMID: 39199610 PMCID: PMC11352926 DOI: 10.3390/cancers16162840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/09/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024] Open
Abstract
Interleukin-10 (IL-10) has been shown to be present at high levels in the ascites of ovarian cancer (OC) patients; however, little is known about its prognostic value. We sought to correlate IL-10 levels in ascites and sera of OC patients with clinicopathologic characteristics and oncologic outcomes. IL-10 levels and clinical data from biobanked ascites and serum samples of OC patients were evaluated. Receiver operating characteristic curves were used to quantify marker performance and identify IL-10-high and IL-10-low groups. Correlations between IL-10 levels and clinicopathologic data were performed. Survival outcomes were calculated, while the factors affecting them were also investigated. A total of 106 patients had ascites samples, of which 44 serum samples were also available. Mean ascites IL-10 levels were significantly higher in patients with serous histology compared to endometrioid histology (p = 0.024). Fold-change in ascites IL-10 during treatment positively correlated with clinical response, as determined by a change in serum cancer antigen (CA)-125 levels (p = 0.0126). Median progression-free survival (PFS) and overall survival (OS) were shorter in patients with high compared with low ascites IL-10 levels (PFS: 18 versus 60 months; p = 0.007, OS: 42 versus 85 months; p = 0.029). A significant positive correlation was seen between ascites and sera IL-10 levels (p = 0.019). In multivariable analyses, a high ascites IL-10 level was associated with a significantly worse prognosis (PFS hazard ratio (HR) = 1.93; p = 0.02). Patients with high ascites levels of IL-10 have worse outcomes, which are likely reflective of the immunosuppressive effect of IL-10. This highlights its potential role as an immunomodulator in the tumor microenvironment, leading to OC immune evasion.
Collapse
Affiliation(s)
- Paul Adrien Guigue
- Lady Davis Institute for Medical Research at the Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
| | - Yoav Brezinov
- Department of Experimental Surgery, McGill University, Montreal, QC H3T 1E2, Canada
| | - Amber Yasmeen
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC H3T 1E2, Canada
| | - Maroua Mbarik
- Lady Davis Institute for Medical Research at the Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
| | - Shannon Salvador
- Lady Davis Institute for Medical Research at the Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC H3T 1E2, Canada
| | - Susie Lau
- Lady Davis Institute for Medical Research at the Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC H3T 1E2, Canada
| | - Walter Henri Gotlieb
- Lady Davis Institute for Medical Research at the Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
- Department of Experimental Surgery, McGill University, Montreal, QC H3T 1E2, Canada
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC H3T 1E2, Canada
| | - Melica Nourmoussavi Brodeur
- Lady Davis Institute for Medical Research at the Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC H3T 1E2, Canada
| |
Collapse
|
2
|
Elemam NM, Mekky RY, Rashid G, Braoudaki M, Youness RA. Pharmacogenomic and epigenomic approaches to untangle the enigma of IL-10 blockade in oncology. Expert Rev Mol Med 2024; 26:e1. [PMID: 38186186 PMCID: PMC10941350 DOI: 10.1017/erm.2023.26] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/29/2023] [Accepted: 11/10/2023] [Indexed: 01/09/2024]
Abstract
The host immune system status remains an unresolved mystery among several malignancies. An immune-compromised state or smart immune-surveillance tactics orchestrated by cancer cells are the primary cause of cancer invasion and metastasis. Taking a closer look at the tumour-immune microenvironment, a complex network and crosstalk between infiltrating immune cells and cancer cells mediated by cytokines, chemokines, exosomal mediators and shed ligands are present. Cytokines such as interleukins can influence all components of the tumour microenvironment (TME), consequently promoting or suppressing tumour invasion based on their secreting source. Interleukin-10 (IL-10) is an interlocked cytokine that has been associated with several types of malignancies and proved to have paradoxical effects. IL-10 has multiple functions on cellular and non-cellular components within the TME. In this review, the authors shed the light on the regulatory role of IL-10 in the TME of several malignant contexts. Moreover, detailed epigenomic and pharmacogenomic approaches for the regulation of IL-10 were presented and discussed.
Collapse
Affiliation(s)
- Noha M. Elemam
- Research Instiute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Clinical Sciences Department, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Radwa Y. Mekky
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA University), Cairo 12622, Egypt
| | - Gowhar Rashid
- Amity Medical School, Amity University, Gurugram (Manesar) 122413, Haryana, India
| | - Maria Braoudaki
- Department of Clinical, Pharmaceutical and Biological Sciences, School of Life and Medical Sciences, University of Hertfordshire, Hatfield AL10 9AB, UK
| | - Rana A. Youness
- Biology and Biochemistry Department, Faculty of Biotechnology, German International University, Cairo 11835, Egypt
| |
Collapse
|
3
|
Węgierek-Ciura K, Mierzejewska J, Szczygieł A, Rossowska J, Wróblewska A, Świtalska M, Goszczyński TM, Szermer-Olearnik B, Pajtasz-Piasecka E. Inhibition of MC38 colon cancer growth by multicomponent chemoimmunotherapy with anti-IL-10R antibodies, HES-MTX nanoconjugate, depends on application of IL-12, IL-15 or IL-18 secreting dendritic cell vaccines. Front Immunol 2023; 14:1212606. [PMID: 37545526 PMCID: PMC10399586 DOI: 10.3389/fimmu.2023.1212606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 07/04/2023] [Indexed: 08/08/2023] Open
Abstract
Background The tumor microenvironment (TME) provides a conducive environment for the growth and survival of tumors. Negative factors present in TME, such as IL-10, may limit the effectiveness of cellular vaccines based on dendritic cells, therefore, it is important to control its effect. The influence of IL-10 on immune cells can be abolished e.g., by using antibodies against the receptor for this cytokine - anti-IL-10R. Furthermore, the anticancer activity of cellular vaccines can be enhanced by modifying them to produce proinflammatory cytokines, such as IL-12, IL-15 or IL-18. Additionally, an immunomodulatory dose of methotrexate and hydroxyethyl starch (HES-MTX) nanoconjugate may stimulate effector immune cells and eliminate regulatory T cells, which should enhance the antitumor action of immunotherapy based on DC vaccines. The main aim of our study was to determine whether the HES-MTX administered before immunotherapy with anti-IL-10R antibodies would change the effect of vaccines based on dendritic cells overproducing IL-12, IL-15, or IL-18. Methods The activity of modified DCs was checked in two therapeutic protocols - immunotherapy with the addition of anti-IL10R antibodies and chemoimmunotherapy with HES-MTX and anti-IL10R antibodies. The inhibition of tumor growth and the effectiveness of the therapy in inducing a specific antitumor response were determined by analyzing lymphoid and myeloid cell populations in tumor nodules, and the activity of restimulated splenocytes. Results and conclusions Using the HES-MTX nanoconjugate before immunotherapy based on multiple administrations of anti-IL-10R antibodies and cellular vaccines capable of overproducing proinflammatory cytokines IL-12, IL-15 or IL-18 created optimal conditions for the effective action of these vaccines in murine colon carcinoma MC38 model. The applied chemoimmunotherapy caused the highest inhibition of tumor growth in the group receiving DC/IL-15/IL-15Rα/TAg + DC/IL-18/TAg at the level of 72.4%. The use of cellular vaccines resulted in cytotoxic activity increase in both immuno- or chemoimmunotherapy. However, the greatest potential was observed both in tumor tissue and splenocytes obtained from mice receiving two- or three-component vaccines in the course of combined application. Thus, the designed treatment schedule may be promising in anticancer therapy.
Collapse
|
4
|
de Mey W, Locy H, De Ridder K, De Schrijver P, Autaers D, Lakdimi A, Esprit A, Franceschini L, Thielemans K, Verdonck M, Breckpot K. An mRNA mix redirects dendritic cells towards an antiviral program, inducing anticancer cytotoxic stem cell and central memory CD8 + T cells. Front Immunol 2023; 14:1111523. [PMID: 36860873 PMCID: PMC9969480 DOI: 10.3389/fimmu.2023.1111523] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 01/26/2023] [Indexed: 02/16/2023] Open
Abstract
Dendritic cell (DC)-maturation stimuli determine the potency of these antigen-presenting cells and, therefore, the quality of the T-cell response. Here we describe that the maturation of DCs via TriMix mRNA, encoding CD40 ligand, a constitutively active variant of toll-like receptor 4 and the co-stimulatory molecule CD70, enables an antibacterial transcriptional program. Besides, we further show that the DCs are redirected into an antiviral transcriptional program when CD70 mRNA in TriMix is replaced with mRNA encoding interferon-gamma and a decoy interleukin-10 receptor alpha, forming a four-component mixture referred to as TetraMix mRNA. The resulting TetraMixDCs show a high potential to induce tumor antigen-specific T cells within bulk CD8+ T cells. Tumor-specific antigens (TSAs) are emerging and attractive targets for cancer immunotherapy. As T-cell receptors recognizing TSAs are predominantly present on naive CD8+ T cells (TN), we further addressed the activation of tumor antigen-specific T cells when CD8+ TN cells are stimulated by TriMixDCs or TetraMixDCs. In both conditions, the stimulation resulted in a shift from CD8+ TN cells into tumor antigen-specific stem cell-like memory, effector memory and central memory T cells with cytotoxic capacity. These findings suggest that TetraMix mRNA, and the antiviral maturation program it induces in DCs, triggers an antitumor immune reaction in cancer patients.
Collapse
|
5
|
Szczygieł A, Węgierek-Ciura K, Wróblewska A, Mierzejewska J, Rossowska J, Szermer-Olearnik B, Świtalska M, Anger-Góra N, Goszczyński TM, Pajtasz-Piasecka E. Combined therapy with methotrexate nanoconjugate and dendritic cells with downregulated IL-10R expression modulates the tumor microenvironment and enhances the systemic anti-tumor immune response in MC38 murine colon carcinoma. Front Immunol 2023; 14:1155377. [PMID: 37033926 PMCID: PMC10078943 DOI: 10.3389/fimmu.2023.1155377] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/13/2023] [Indexed: 04/11/2023] Open
Abstract
Background Understanding the negative impact of the tumor microenvironment on the creation of an effective immune response has contributed to the development of new therapeutic anti-cancer strategies. One such solution is combined therapy consisting of chemotherapeutic administration followed by dendritic cell (DC)-based vaccines. The use of cytostatic leads to the elimination of cancer cells, but can also modulate the tumor milieu. Moreover, great efforts are being made to increase the therapeutic outcome of immunotherapy, e.g. by enhancing the ability of DCs to generate an efficient immune response, even in the presence of immunosuppressive cytokines such as IL-10. The study aimed to determine the effectiveness of combined therapy with chemotherapeutic with immunomodulatory potential - HES-MTX nanoconjugate (composed of methotrexate (MTX) and hydroxyethyl starch (HES)) and DCs with downregulated expression of IL-10 receptor stimulated with tumor antigens (DC/shIL-10R/TAg) applied in MC38 murine colon carcinoma model. Methods With the use of lentiviral vectors the DCs with decreased expression of IL-10R were obtained and characterized. During in vivo studies MC38-tumor bearing mice received MTX or HES-MTX nanoconjugate as a sole treatment or combined with DC-based immunotherapy containing unmodified DCs or DCs transduced with shRNA against IL-10R (or control shRNA sequence). Tumor volume was monitored during the experiment. One week after the last injection of DC-based vaccines, tumor nodules and spleens were dissected for ex vivo analysis. The changes in the local and systemic anti-tumor immune response were estimated with the use of flow cytometry and ELISA methods. Results and conclusions In vitro studies showed that the downregulation of IL-10R expression in DCs enhances their ability to activate the specific anti-tumor immune response. The use of HES-MTX nanoconjugate and DC/shIL-10R/TAg in the therapy of MC38-tumor bearing mice resulted in the greatest tumor growth inhibition. At the local anti-tumor immune response level a decrease in the infiltration of cells with suppressor activity and an increase in the influx of effector cells into MC38 tumor tissue was observed. These changes were crucial to enhance the effective specific immune response at the systemic level, which was revealed in the greatest cytotoxic activity of spleen cells against MC38 cells.
Collapse
|
6
|
Rallis KS, Corrigan AE, Dadah H, Stanislovas J, Zamani P, Makker S, Szabados B, Sideris M. IL-10 in cancer: an essential thermostatic regulator between homeostatic immunity and inflammation - a comprehensive review. Future Oncol 2022; 18:3349-3365. [PMID: 36172856 DOI: 10.2217/fon-2022-0063] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Cytokines are soluble proteins that mediate intercellular signaling regulating immune and inflammatory responses. Cytokine modulation represents a promising cancer immunotherapy approach for immune-mediated tumor regression. However, redundancy in cytokine signaling and cytokines' pleiotropy, narrow therapeutic window, systemic toxicity, short half-life and limited efficacy represent outstanding challenges for cytokine-based cancer immunotherapies. Recently, there has been interest in the paradoxical role of IL-10 in cancer, its controversial prognostic utility and novel strategies to enhance its therapeutic profile. Here, the authors review the literature surrounding the role of IL-10 within the tumor microenvironment, its prognostic correlates to cancer patient outcomes and its pro- and antitumor effects, and they assess the legitimacy of potential therapeutic strategies harnessing IL-10 by outlining the notable preclinical and clinical evidence to date.
Collapse
Affiliation(s)
- Kathrine S Rallis
- Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, E1 2AD, UK.,Barts Cancer Institute, Queen Mary University of London, London, EC1M 5PZ, UK
| | - Amber E Corrigan
- GKT School of Medicine, King's College London, London, SE1 9RT, UK
| | - Hashim Dadah
- GKT School of Medicine, King's College London, London, SE1 9RT, UK
| | - Justas Stanislovas
- Barts Cancer Institute, Queen Mary University of London, London, EC1M 5PZ, UK
| | - Parisa Zamani
- GKT School of Medicine, King's College London, London, SE1 9RT, UK
| | - Shania Makker
- Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, E1 2AD, UK
| | - Bernadett Szabados
- Barts Cancer Institute, Queen Mary University of London, London, EC1M 5PZ, UK
| | - Michail Sideris
- Women's Health Research Unit, Queen Mary University of London, London, E1 2AB, UK
| |
Collapse
|
7
|
Ricciardi A, Hassan SA, Kamenyeva O, Bennuru S, Andersen J, Nutman TB. A filarial parasite-encoded human IL-10 receptor antagonist reveals a novel strategy to modulate host responses. PNAS NEXUS 2022; 1:pgac184. [PMID: 36246151 PMCID: PMC9552326 DOI: 10.1093/pnasnexus/pgac184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 08/31/2022] [Indexed: 02/05/2023]
Abstract
Interleukin (IL)-10 is the primary cytokine driving the modulation of the host response in filarial infections. We performed binding assays with Brugia malayi antigen extracts and human IL-10R1. Bm5539 was the top-binding hit. We identified a short sequence, termed truncated Bm5339, that has structural similarities to the human IL-10 functional dimer. Sequence comparisons revealed that other filarial parasites possess Bm5539 orthologues. Using recombinant Bm5539 in a modified Luciferase Immunoprecipitation System assay, we confirmed that both the truncated and full-length forms of the protein can bind to human IL-10R1. Truncated Bm5539 could inhibit human IL-10-driven phosphorylation of STAT3, thereby demonstrating that Bm5539 acts as an IL-10 antagonist, most likely through competitive binding to the receptor. We provide a structural basis for these observations using computational modeling and simulations. This parasite-encoded cytokine receptor antagonist provides an additional lens through which parasite-induced modulation of the host immune response can be examined.
Collapse
Affiliation(s)
- Alessandra Ricciardi
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA
| | - Sergio A Hassan
- Bioinformatics and Computational Biosciences Branch, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA
| | - Olena Kamenyeva
- Research Technology Branch, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA
| | - Sasisekhar Bennuru
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA
| | - John Andersen
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, 12735 Twinbrook Parkway, Rockville, MD 20852, USA
| | | |
Collapse
|
8
|
Dwivedi V, Gautam S, Headley CA, Piergallini T, Torrelles JB, Turner J. IL-10 Receptor Blockade Delivered Simultaneously with Bacillus Calmette-Guérin Vaccination Sustains Long-Term Protection against Mycobacterium tuberculosis Infection in Mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1406-1416. [PMID: 35181640 PMCID: PMC11075079 DOI: 10.4049/jimmunol.2100900] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 01/12/2022] [Indexed: 11/19/2022]
Abstract
Mycobacterium bovis bacillus Calmette-Guérin (BCG) immunization still remains the best vaccination strategy available to control the development of active tuberculosis. Protection afforded by BCG vaccination gradually wanes over time and although booster strategies have promise, they remain under development. An alternative approach is to improve BCG efficacy through host-directed therapy. Building upon prior knowledge that blockade of IL-10R1 during early Mycobacterium tuberculosis infection improves and extends control of M. tuberculosis infection in mice, we employed a combined anti-IL-10R1/BCG vaccine strategy. An s.c. single vaccination of BCG/anti-IL10-R1 increased the numbers of CD4+ and CD8+ central memory T cells and reduced Th1 and Th17 cytokine levels in the lung for up to 7 wk postvaccination. Subsequent M. tuberculosis challenge in mice showed both an early (4 wk) and sustained long-term (47 wk) control of infection, which was associated with increased survival. In contrast, protection of BCG/saline-vaccinated mice waned 8 wk after M. tuberculosis infection. Our findings demonstrate that a single and simultaneous vaccination with BCG/anti-IL10-R1 sustains long-term protection, identifying a promising approach to enhance and extend the current BCG-mediated protection against TB.
Collapse
Affiliation(s)
- Varun Dwivedi
- Disease Intervention and Prevention Program, Texas Biomedical Research Institute, San Antonio, TX
| | - Shalini Gautam
- Host Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, TX; and
| | - Colwyn A Headley
- Host Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, TX; and
| | - Tucker Piergallini
- Host Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, TX; and
| | - Jordi B Torrelles
- Population Health Program, Texas Biomedical Research Institute, San Antonio, TX
| | - Joanne Turner
- Host Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, TX; and
| |
Collapse
|
9
|
Piatakova A, Polakova I, Smahelova J, Johari SD, Nunvar J, Smahel M. Distinct Responsiveness of Tumor-Associated Macrophages to Immunotherapy of Tumors with Different Mechanisms of Major Histocompatibility Complex Class I Downregulation. Cancers (Basel) 2021; 13:cancers13123057. [PMID: 34205330 PMCID: PMC8235485 DOI: 10.3390/cancers13123057] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/02/2021] [Accepted: 06/16/2021] [Indexed: 01/16/2023] Open
Abstract
Simple Summary Tumor-associated macrophages (TAMs) are one of the major cell subpopulations in the tumor microenvironment (TME) where they can either be pro-tumorigenic or contribute to an anti-tumor immunity. The TME and TAM phenotype were analyzed after combined immuno-therapy (IT) in tumor models characterized by distinct expression of major histocompatibility class I complex (MHC-I) molecules, i.e., tumors induced with TC-1 (MHC-I-proficient), TC-1/A9 (reversibly downregulated), and TC-1/dB2m (irreversibly downregulated) cells. We found out that combined IT highly activated immune reactions in the TME of TC-1 and TC-1/A9 tumors, but the TME of TC-1/dB2m tumors remained almost unchanged. Correspondingly, TAMs from TC-1/A9 tumors were able to destroy tumor cells in vitro, while TAMs isolated from TC-1/dB2m tumors showed profoundly decreased cytotoxicity. Hence, various capabilities of TAMs in tumors with distinct expression of MHC-I molecules should be considered when applying IT, particularly IT focused on TAMs. Abstract Tumor-associated macrophages (TAMs) plentifully infiltrate the tumor microenvironment (TME), but their role in anti-tumor immunity is controversial. Depending on the acquired polarization, they can either support tumor growth or participate in the elimination of neoplastic cells. In this study, we analyzed the TME by RNA-seq and flow cytometry and examined TAMs after ex vivo activation. Tumors with normal and either reversibly or irreversibly decreased expression of major histocompatibility complex class I (MHC-I) molecules were induced with TC-1, TC-1/A9, and TC-1/dB2m cells, respectively. We found that combined immunotherapy (IT), composed of DNA immunization and the CpG oligodeoxynucleotide (ODN) ODN1826, evoked immune reactions in the TME of TC-1- and TC-1/A9-induced tumors, while the TME of TC-1/dB2m tumors was mostly immunologically unresponsive. TAMs infiltrated both tumor types with MHC-I downregulation, but only TAMs from TC-1/A9 tumors acquired the M1 phenotype upon IT and were cytotoxic in in vitro assay. The anti-tumor effect of combined IT was markedly enhanced by a blockade of the colony-stimulating factor-1 receptor (CSF-1R), but only against TC-1/A9 tumors. Overall, TAMs from tumors with irreversible MHC-I downregulation were resistant to the stimulation of cytotoxic activity. These data suggest the dissimilarity of TAMs from different tumor types, which should be considered when utilizing TAMs in cancer IT.
Collapse
|
10
|
Gharbavi M, Danafar H, Amani J, Sharafi A. Immuno-informatics analysis and expression of a novel multi-domain antigen as a vaccine candidate against glioblastoma. Int Immunopharmacol 2020; 91:107265. [PMID: 33360829 DOI: 10.1016/j.intimp.2020.107265] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 11/17/2020] [Accepted: 11/27/2020] [Indexed: 12/28/2022]
Abstract
Glioblastoma multiform is the most common of primary malignant brain tumors in adults. Currently, surgical resection of the tumor mass, followed by adjuvant radiotherapy and chemotherapy are standard treatments for glioblastoma multiform but so far are not effective treatments. Thus, the development of a vaccine, as a safe and efficient strategy for prophylactic or therapeutic purposes against glioblastoma multiform is very necessary. The present study aimed to design the multi-domain vaccine for glioblastoma multiform. An in silico approach was used to select the most potent domains of proteins to induce the host's B- and T-cell immune response against glioblastoma multiform. IL-13Rα-2 (amino acid positions 27-144), TNC (amino acid positions 1900-2100), and PTPRZ-1(amino acid positions 731-884) were found to have potent inducible immune responses. So, we considered them for fusing with a linker A(EAAAK)3A to construct the multi-domain recombinant vaccine. The immuno-informatics analysis of the designed recombinant vaccine construct was performed to evaluate its efficacy. Although the designed recombinant vaccine construct did not show allergen property, its antigenicity was estimated at 0.78. The Physico-chemical properties of the recombinant vaccine construct were characterized and revealed the potency of the vaccine candidate. Then its secondary and tertiary structures, mRNA structure, molecular docking, and immune simulation were predicted using bioinformatics tools. Next, the designed recombinant vaccine construct was synthesized, and cloned into the pET28a vector and expressed in E. coli BL21. Besides, the circular dichroism spectroscopy was utilized for the investigation of the secondary structure changes of the recombinant vaccine construct. The results of the verification assessment of the recombinant vaccine construct expression indicated that in silico analysis was relatively accurate, and relatively change occurred on the protein secondary structure. In our future plan, the vaccine candidate that was confirmed by in silico tools should be validated by further in vitro and in vivo experimental studies.
Collapse
Affiliation(s)
- Mahmoud Gharbavi
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran; Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hossein Danafar
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Jafar Amani
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Ali Sharafi
- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
| |
Collapse
|
11
|
Ni G, Zhang L, Yang X, Li H, Ma B, Walton S, Wu X, Yuan J, Wang T, Liu X. Targeting interleukin-10 signalling for cancer immunotherapy, a promising and complicated task. Hum Vaccin Immunother 2020; 16:2328-2332. [PMID: 32159421 DOI: 10.1080/21645515.2020.1717185] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Interleukin 10 (IL-10) belongs to IL-10 family cytokines that are critical for maintaining the integrity of epithelial tissues, protecting pathogenic infection, and preventing excessive immune responses to damage self. Temporal IL-10 signaling blockade enhances vaccine-induced tumor regression by CD8 + T cells. IL-10, especially pegylated IL-10, mediates tumor regression by expanding tumor-infiltrating CD8 + T cells. Moreover, targeting IL-10 enhances immune checkpoint inhibitor mediated tumor regression. In the current paper, we will review recent advances in this area and discuss the complexity of IL-10 manipulation for cancer therapy.
Collapse
Affiliation(s)
- Guoying Ni
- Department of Nuclear Medicine, The First Affiliated Hospital/School of Clinical Medicine of Guangdong Pharmaceutical University , Guangzhou, China.,Genecology Research Centre, University of the Sunshine Coast , Maroochydore, Australia.,School of Medical Science, Griffith Health Institute, Griffith University , Gold Coast, Australia
| | - Lu Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital/School of Clinical Medicine of Guangdong Pharmaceutical University , Guangzhou, China
| | - Xiaodan Yang
- Department of Nuclear Medicine, The First Affiliated Hospital/School of Clinical Medicine of Guangdong Pharmaceutical University , Guangzhou, China
| | - Hejie Li
- Genecology Research Centre, University of the Sunshine Coast , Maroochydore, Australia.,Institute of Industrial Science, Department of Mechanical and Bio-functional System, The University of Tokyo , Tokyo, Japan
| | - Bowei Ma
- Department of Nuclear Medicine, The First Affiliated Hospital/School of Clinical Medicine of Guangdong Pharmaceutical University , Guangzhou, China
| | - Shelley Walton
- Genecology Research Centre, University of the Sunshine Coast , Maroochydore, Australia
| | - Xiaolian Wu
- Cancer Research Institute, First People's Hospital of Foshan , Foshan, Guangdong, China
| | - Jianwei Yuan
- Department of Nuclear Medicine, The First Affiliated Hospital/School of Clinical Medicine of Guangdong Pharmaceutical University , Guangzhou, China
| | - Tianfang Wang
- Genecology Research Centre, University of the Sunshine Coast , Maroochydore, Australia
| | - Xiaosong Liu
- Department of Nuclear Medicine, The First Affiliated Hospital/School of Clinical Medicine of Guangdong Pharmaceutical University , Guangzhou, China.,Genecology Research Centre, University of the Sunshine Coast , Maroochydore, Australia.,Cancer Research Institute, First People's Hospital of Foshan , Foshan, Guangdong, China.,School of Health and Sport Sciences, University of the Sunshine Coast , Maroochydore, Australia
| |
Collapse
|
12
|
Pan X, Ma B, You X, Chen S, Wu J, Wang T, Walton SF, Yuan J, Wu X, Chen G, Wang Y, Ni G, Liu X. Synthesized natural peptides from amphibian skin secretions increase the efficacy of a therapeutic vaccine by recruiting more T cells to the tumour site. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 19:163. [PMID: 31277636 PMCID: PMC6612097 DOI: 10.1186/s12906-019-2571-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 06/19/2019] [Indexed: 02/01/2023]
Abstract
Background Therapeutic vaccines against cervical cancer remain ineffective. Previously, we demonstrated that blocking the signalling of a cytokine, interleukin 10, at the time of immunisation elicited significantly higher numbers of antigen specific T cells and inhibited tumour growth in mice. Results In the current paper, we demonstrate, in a HPV16 E6/E7 transformed TC-1 tumour mouse model, that despite increased antigen specific T cell numbers, blocking IL-10 signalling at the time of immunisation does not increase the survival time of the TC-1 tumour bearing mice compared to mice receiving the same immunisation with no IL-10 signalling blockade. Moreover, the function of tumour infiltrating T cells isolated 3 weeks post TC-1 transplantation is more suppressed than those isolated 2 weeks after tumour inoculation. We demonstrate that synthesized caerin peptides, derived from amphibian skin secretions, 1) were able to inhibit TC-1 tumour growth both in vitro and in vivo; 2) are environmentally stable; and 3) promote the secretion of pro-inflammatory interlukine-6 by TC-1 cells. Notably caerin peptides were able to increase the survival time of TC-1 tumour bearing mice after therapeutic vaccination with a HPV16E7 peptide-based vaccine containing IL-10 inhibitor, via recruiting increased levels of T cells to the tumour site. Conclusion Caerin peptides increase the efficacy of a therapeutic vaccine by recruiting more T cells to the tumour site.
Collapse
|
13
|
The Dosage of the Derivative of Clostridium Ghonii (DCG) Spores Dictates Whether an IFN γ/IL-9 or a Strong IFN γ Response Is Elicited in TC-1 Tumour Bearing Mice. BIOMED RESEARCH INTERNATIONAL 2019; 2019:1395138. [PMID: 31183361 PMCID: PMC6512072 DOI: 10.1155/2019/1395138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 01/04/2019] [Accepted: 01/15/2019] [Indexed: 11/24/2022]
Abstract
Background Anaerobic Clostridial spores (CG) cause significant oncolysis in hypoxic tumour microenvironment and result in tumour regression in both animal models and clinical trials. The immune mediated response plays a critical role in the antitumour effect by the anaerobic spore treatment. Method Human papillomavirus 16 E6/E7 transformed TC-1 tumour bearing mice were intravenously administered with low (1 × 108 CFU/kg) or high dosage (3 × 108 CFU/kg) of Derivative Clostridial spore (DCG). Results Intravenous administration of the derivative of Clostridial ghonii (DCG) spores leads to both tumour and systemic inflammatory responses characterized by increased IFNγ/IL-9 secreting T cells in the spleen and the tumour. Low numbers of antigen specific T cells (<20/106 spleen cells) in the spleen of the tumour bearing mice are also detected after intravenous DCG delivery. Interestingly, our results showed that a mixed IL-9/IFNγ secreting T cell response was induced when the tumour bearing mice received a low dose of DCG spore (1 × 108 CFU/kg), while a strong IFNγ response was elicited with a high dosage of DCG spore (3 × 108 CFU/kg). Conclusion The dosage of DCG spore will determine the types of the DCG induced immune responses.
Collapse
|
14
|
Yang L, Dong Y, Li Y, Wang D, Liu S, Wang D, Gao Q, Ji S, Chen X, Lei Q, Jiang W, Wang L, Zhang B, Yu JJ, Zhang Y. IL‐10 derived from M2 macrophage promotes cancer stemness
via
JAK1/STAT1/NF‐κB/Notch1 pathway in non‐small cell lung cancer. Int J Cancer 2019; 145:1099-1110. [PMID: 30671927 DOI: 10.1002/ijc.32151] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 12/07/2018] [Accepted: 01/16/2019] [Indexed: 12/27/2022]
Affiliation(s)
- Li Yang
- Biotherapy Center The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan People's Republic of China
- Department of Oncology The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan People's Republic of China
- Key Laboratory for Tumor Immunology and Biotherapy of Henan Province Zhengzhou Henan People's Republic of China
| | - Ying Dong
- Biotherapy Center The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan People's Republic of China
- Department of Oncology The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan People's Republic of China
- Key Laboratory for Tumor Immunology and Biotherapy of Henan Province Zhengzhou Henan People's Republic of China
| | - Yanjun Li
- Biotherapy Center The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan People's Republic of China
- Key Laboratory for Tumor Immunology and Biotherapy of Henan Province Zhengzhou Henan People's Republic of China
- School of Life Sciences Zhengzhou University Zhengzhou Henan People's Republic of China
| | - Dong Wang
- Biotherapy Center The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan People's Republic of China
- Department of Oncology The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan People's Republic of China
- Key Laboratory for Tumor Immunology and Biotherapy of Henan Province Zhengzhou Henan People's Republic of China
| | - Shasha Liu
- Biotherapy Center The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan People's Republic of China
- Key Laboratory for Tumor Immunology and Biotherapy of Henan Province Zhengzhou Henan People's Republic of China
- School of Life Sciences Zhengzhou University Zhengzhou Henan People's Republic of China
| | - Dan Wang
- Biotherapy Center The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan People's Republic of China
- Department of Oncology The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan People's Republic of China
- Key Laboratory for Tumor Immunology and Biotherapy of Henan Province Zhengzhou Henan People's Republic of China
| | - Qun Gao
- Biotherapy Center The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan People's Republic of China
- Department of Oncology The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan People's Republic of China
- Key Laboratory for Tumor Immunology and Biotherapy of Henan Province Zhengzhou Henan People's Republic of China
| | - Shaofei Ji
- Department of Radiology Orthopaedic Hospital of Zhengzhou City Zhengzhou Henan People's Republic of China
| | - Xinfeng Chen
- Biotherapy Center The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan People's Republic of China
- Department of Oncology The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan People's Republic of China
- Key Laboratory for Tumor Immunology and Biotherapy of Henan Province Zhengzhou Henan People's Republic of China
| | - Qingyang Lei
- Biotherapy Center The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan People's Republic of China
- Department of Oncology The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan People's Republic of China
- Key Laboratory for Tumor Immunology and Biotherapy of Henan Province Zhengzhou Henan People's Republic of China
| | - Wenyi Jiang
- Biotherapy Center The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan People's Republic of China
- Department of Oncology The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan People's Republic of China
- Key Laboratory for Tumor Immunology and Biotherapy of Henan Province Zhengzhou Henan People's Republic of China
| | - Liping Wang
- Department of Oncology The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan People's Republic of China
- Key Laboratory for Tumor Immunology and Biotherapy of Henan Province Zhengzhou Henan People's Republic of China
| | - Bin Zhang
- Department of Hematology/Oncology, School of Medicine Northwestern University Chicago IL
| | - Jane J. Yu
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, College of Medicine University of Cincinnati Cincinnati OH
| | - Yi Zhang
- Biotherapy Center The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan People's Republic of China
- Department of Oncology The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan People's Republic of China
- Key Laboratory for Tumor Immunology and Biotherapy of Henan Province Zhengzhou Henan People's Republic of China
- School of Life Sciences Zhengzhou University Zhengzhou Henan People's Republic of China
| |
Collapse
|
15
|
Llopiz D, Ruiz M, Silva L, Sarobe P. Enhancement of Antitumor Vaccination by Targeting Dendritic Cell-Related IL-10. Front Immunol 2018; 9:1923. [PMID: 30233565 PMCID: PMC6129595 DOI: 10.3389/fimmu.2018.01923] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 08/06/2018] [Indexed: 12/24/2022] Open
Abstract
Understanding mechanisms associated to dendritic cell (DC) functions has allowed developing new antitumor therapeutic vaccination strategies. However, these vaccines have demonstrated limited clinical results. Although the low immunogenicity of tumor antigens used and the presence of tumor-associated suppressive factors may in part account for these results, intrinsic vaccine-related factors may also be involved. Vaccines modulate DC functions by inducing activating and inhibitory signals that determine ensuing T cell responses. In this mini review, we focus on IL-10, inhibitory cytokine induced in DC upon vaccination, which defines a suppressive cell subset, discussing its implications as a potential target in combined vaccination immunotherapies.
Collapse
Affiliation(s)
- Diana Llopiz
- Programa de Inmunología e Inmunoterapia, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain.,Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Marta Ruiz
- Programa de Inmunología e Inmunoterapia, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain.,Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Leyre Silva
- Programa de Inmunología e Inmunoterapia, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain.,Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Pablo Sarobe
- Programa de Inmunología e Inmunoterapia, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain.,Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| |
Collapse
|
16
|
Ni G, Wang T, Yang L, Wang Y, Liu X, Wei MQ. Combining anaerobic bacterial oncolysis with vaccination that blocks interleukin-10 signaling may achieve better outcomes for late stage cancer management. Hum Vaccin Immunother 2017; 12:599-606. [PMID: 26367244 DOI: 10.1080/21645515.2015.1089008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Late stage solid tumors cause significant cancer mortality rates worldwide and effective therapy remains a big challenge. Cancer therapeutic vaccines elicit tumor specific T cells that kill tumor cells yet often fail to result in tumor destruction because of the limited T cell response and the local immune-suppressive environment. Blocking interleukin 10 (IL-10) signaling at the time of therapeutic vaccination elicits much stronger T cell responses than vaccination without IL-10 blocking. Anaerobic oncolytic bacteria target hypoxic regions of the late stage tumor tissues which not only stops tumor growth but also provides a pro-inflammatory environment that may increase the effectiveness of a therapeutic vaccine by recruiting more effector T cells to tumor site. In this review, we argue that combining both bacterial and vaccine therapies may improve the efficiency of late stage cancer management.
Collapse
Affiliation(s)
- Guoying Ni
- a School of Medical Science and Griffith Health Institute, Griffith University , Gold Coast , QLD , Australia.,d Tangshan Supervision Institute of Health , Tangshan , China
| | - Tianfang Wang
- c Genecology Research Center, University of the Sunshine Coast , Maroochydore DC , QLD , Australia
| | - Lin Yang
- f Department of Surgical Oncology , Tangshan Gongren Hospital , Tangshan , Hebei , China
| | - Yuejian Wang
- e Cancer Research Institute, Foshan First People's Hospital , Foshan, Guangdong , China
| | - Xiaosong Liu
- b Inflammation and Healing Research Cluster, University of the Sunshine Coast , Maroochydore DC , QLD , Australia.,e Cancer Research Institute, Foshan First People's Hospital , Foshan, Guangdong , China
| | - Ming Q Wei
- a School of Medical Science and Griffith Health Institute, Griffith University , Gold Coast , QLD , Australia
| |
Collapse
|
17
|
Ni G, Liao Z, Chen S, Wang T, Yuan J, Pan X, Mounsey K, Cavezza S, Liu X, Wei MQ. Blocking IL-10 signalling at the time of immunization does not increase unwanted side effects in mice. BMC Immunol 2017; 18:40. [PMID: 28810829 PMCID: PMC5557397 DOI: 10.1186/s12865-017-0224-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 08/01/2017] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Cancer therapeutic vaccine induced cytotoxic T cell (CTL) responses are pivotal for the killing of tumour cells. Blocking interleukin 10 (IL-10) signalling at the time of immunization increases vaccine induced CTL responses and improves prevention of tumour growth in animal models compared to immunization without an IL-10 signalling blockade. Therefore, this immunization strategy may have potential to curtail cancer in a clinical setting. However, IL-10 deficiency leads to autoimmune disease in the gut. Blocking IL-10 at the time of immunization may result in unwanted side effects, especially immune-pathological diseases in the intestine. METHODS We investigated whether blocking IL-10 at the time of immunization results in intestinal inflammation responses in a mouse TC-1 tumour model and in a NOD autoimmune disease prone mouse model. RESULTS We now show that blocking IL-10 at the time of immunization increases IL-10 production by CD4+ T cells in the spleen and draining lymph nodes, and does not result in blood cell infiltration to the intestines leading to intestinal pathological changes. Moreover, immunization with papillomavirus like particles combined with simultaneously blocking IL-10 signalling does not increase the incidence of autoimmune disease in Non-obese diabetic (NOD) mice. CONCLUSIONS Our results indicate that immunization with an IL-10 inhibitor may facilitate the generation of safe, effective therapeutic vaccines against chronic viral infection and cancer.
Collapse
Affiliation(s)
- Guoying Ni
- School of Medical Science, Griffith Health Institute, Griffith University, Gold Coast, QLD 4333 Australia
| | - Zaowen Liao
- Cancer Research Institute, The First People’s Hospital of Foshan, Foshan, Guangdong 528000 China
| | - Shu Chen
- Cancer Research Institute, The First People’s Hospital of Foshan, Foshan, Guangdong 528000 China
| | - Tianfang Wang
- Inflammation and Healing Research Cluster, University of the Sunshine Coast, Maroochydore DC, QLD 4558 Australia
| | - Jianwei Yuan
- Molecular diagnosis and Target Therapy Laboratory, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong China
| | - Xuan Pan
- Molecular diagnosis and Target Therapy Laboratory, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong China
| | - Kate Mounsey
- Inflammation and Healing Research Cluster, University of the Sunshine Coast, Maroochydore DC, QLD 4558 Australia
| | - Shelley Cavezza
- Inflammation and Healing Research Cluster, University of the Sunshine Coast, Maroochydore DC, QLD 4558 Australia
| | - Xiaosong Liu
- Cancer Research Institute, The First People’s Hospital of Foshan, Foshan, Guangdong 528000 China
- Inflammation and Healing Research Cluster, University of the Sunshine Coast, Maroochydore DC, QLD 4558 Australia
- Molecular diagnosis and Target Therapy Laboratory, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong China
| | - Ming Q. Wei
- School of Medical Science, Griffith Health Institute, Griffith University, Gold Coast, QLD 4333 Australia
| |
Collapse
|
18
|
Wu Z, Hu T, Rothwell L, Vervelde L, Kaiser P, Boulton K, Nolan MJ, Tomley FM, Blake DP, Hume DA. Analysis of the function of IL-10 in chickens using specific neutralising antibodies and a sensitive capture ELISA. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 63:206-12. [PMID: 27108075 PMCID: PMC4947970 DOI: 10.1016/j.dci.2016.04.016] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 04/18/2016] [Accepted: 04/18/2016] [Indexed: 05/19/2023]
Abstract
In mammals, the inducible cytokine interleukin 10 is a feedback negative regulator of inflammation. To determine the extent to which this function is conserved in birds, recombinant chicken IL-10 was expressed as a secreted human Ig Fc fusion protein (chIL-10-Fc) and used to immunise mice. Five monoclonal antibodies (mAb) which specifically recognise chicken IL-10 were generated and characterised. Two capture ELISA assays were developed which detected native chIL-10 secreted from chicken bone marrow-derived macrophages (chBMMs) stimulated with lipopolysaccharide (LPS). Three of the mAbs detected intracellular IL-10. This was detected in only a subset of the same LPS-stimulated chBMMs. The ELISA assay also detected massive increases in circulating IL-10 in chickens challenged with the coccidial parasite, Eimeria tenella. The same mAbs neutralised the bioactivity of recombinant chIL-10. The role of IL-10 in feedback control was tested in vitro. The neutralising antibodies prevented IL-10-induced inhibition of IFN-γ synthesis by mitogen-activated lymphocytes and increased nitric oxide production in LPS-stimulated chBMMs. The results confirm that IL-10 is an inducible feedback regulator of immune response in chickens, and could be the target for improved vaccine efficacy or breeding strategies.
Collapse
Affiliation(s)
- Zhiguang Wu
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| | - Tuanjun Hu
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| | - Lisa Rothwell
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| | - Lonneke Vervelde
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| | - Pete Kaiser
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| | - Kay Boulton
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| | - Matthew J Nolan
- Department of Pathology and Pathogen Biology, Royal Veterinary College, University of London, Hatfield AL9 7TA, UK
| | - Fiona M Tomley
- Department of Pathology and Pathogen Biology, Royal Veterinary College, University of London, Hatfield AL9 7TA, UK
| | - Damer P Blake
- Department of Pathology and Pathogen Biology, Royal Veterinary College, University of London, Hatfield AL9 7TA, UK
| | - David A Hume
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK.
| |
Collapse
|
19
|
Ni G, Wang Y, Cummins S, Walton S, Mounsey K, Liu X, Wei MQ, Wang T. Inhibitory mechanism of peptides with a repeating hydrophobic and hydrophilic residue pattern on interleukin-10. Hum Vaccin Immunother 2016; 13:518-527. [PMID: 27686406 DOI: 10.1080/21645515.2016.1238537] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Interleukin 10 (IL-10) is a cytokine that is able to downregulate inflammation. Its overexpression is directly associated with the difficulty in the clearance of chronic viral infections, such as chronic hepatitis B, hepatitis C and HIV infection, and infection-related cancer. IL-10 signaling blockade has been proposed as a promising way of clearing chronic viral infection and preventing tumor growth in animal models. Recently, we have reported that peptides with a helical repeating pattern of hydrophobic and hydrophilic residues are able to inhibit IL-10 significantly both in vitro and in vivo. 1 In this work, we seek to further study the inhibiting mechanism of these peptides using sequence-modified peptides. As evidenced by both experimental and molecular dynamics simulation in concert the N-terminal hydrophobic peptide constructed with repeating hydrophobic and hydrophilic pattern of residues is more likely to inhibit IL10. In addition, the sequence length and the ability of protonation are also important for inhibition activity.
Collapse
Affiliation(s)
- Guoying Ni
- a Genecology Research Centre , University of the Sunshine Coast , Maroochydore , DC , Australia.,b School of Medical Science, Griffith Health Institute , Griffith University , Gold Coast , Australia
| | - Yuejian Wang
- c Cancer Research Institute, Foshan First People's Hospital , Foshan , Guangdong , China
| | - Scott Cummins
- a Genecology Research Centre , University of the Sunshine Coast , Maroochydore , DC , Australia
| | - Shelley Walton
- d Inflammation and Healing Research Cluster, School of Health and Sport Sciences , University of Sunshine Coast , Maroochydore , DC , Australia
| | - Kate Mounsey
- d Inflammation and Healing Research Cluster, School of Health and Sport Sciences , University of Sunshine Coast , Maroochydore , DC , Australia
| | - Xiaosong Liu
- c Cancer Research Institute, Foshan First People's Hospital , Foshan , Guangdong , China.,d Inflammation and Healing Research Cluster, School of Health and Sport Sciences , University of Sunshine Coast , Maroochydore , DC , Australia
| | - Ming Q Wei
- b School of Medical Science, Griffith Health Institute , Griffith University , Gold Coast , Australia
| | - Tianfang Wang
- a Genecology Research Centre , University of the Sunshine Coast , Maroochydore , DC , Australia
| |
Collapse
|
20
|
Investigation the Possibility of Using Peptides with a Helical Repeating Pattern of Hydro-Phobic and Hydrophilic Residues to Inhibit IL-10. PLoS One 2016; 11:e0153939. [PMID: 27100390 PMCID: PMC4839630 DOI: 10.1371/journal.pone.0153939] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 04/06/2016] [Indexed: 01/26/2023] Open
Abstract
Blockade of IL-10 signalling clears chronic viral and bacterial infections. Immunization together with blockade of IL-10 signalling or relatively low level of IL-10 further enhances viral and bacterial clearance. IL-10 functions through binding to interleukin 10 receptor (IL-10R). Here we showed that peptides P1 and P2 with the hydrophobic and hydrophilic pattern of the IL10R-binding helix in IL-10 could bind with either IL-10R1 or IL-10, and inhibit inflammatory signals with long duration and negligible cytotoxicity in vitro. Furthermore, P2 can enhance antigen specific CD8+ T cell responses in mice induced by the vaccine based on a long peptide of protein E7 in a human papillomavirus type 16.
Collapse
|
21
|
Chen S, Ni G, Wu X, Zhu B, Liao Z, Wang Y, Liu X. Blocking IL-10 signalling at the time of immunization renders the tumour more accessible to T cell infiltration in mice. Cell Immunol 2015; 300:9-17. [PMID: 26607604 DOI: 10.1016/j.cellimm.2015.11.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 11/05/2015] [Accepted: 11/11/2015] [Indexed: 12/15/2022]
Abstract
We recently reported that blockade of IL-10 signalling at the time of a human papillomavirus (HPV) long E7 peptide/LPS immunization leads to the regression of established HPV-16 immortalized tumours in mice similar to that induced by long E7 peptide/incomplete Freund's adjuvant (IFA)-based vaccination. In this paper, we demonstrated that blockade of IL-10 signalling at the time of long E7 peptide/LPS could elicit stronger T cells responses and render the tumour more accessible for immune cell infiltration than vaccination with long E7 peptide/IFA. Furthermore, priming with long E7 peptide/LPS and IL10 signalling blockade then boosting with long E7 peptide/IFA elicits stronger CD8+ T cell responses than long E7 peptide/IFA immunization. The results suggest that priming with long E7 peptide/LPS and IL10 signalling inhibitor, then boosting with long E7 peptide/IFA elicits may lead to better HPV infection related tumour regression in clinic.
Collapse
Affiliation(s)
- Shu Chen
- Cancer Research Institute, Foshan First People's Hospital, Foshan, Guangdong 528000, China
| | - Guoying Ni
- School of Medical Science, Griffith Health Institute, Griffith University, Gold Coast, QLD 4222, Australia
| | - Xiaolian Wu
- Cancer Research Institute, Foshan First People's Hospital, Foshan, Guangdong 528000, China
| | - Bin Zhu
- Cancer Research Institute, Foshan First People's Hospital, Foshan, Guangdong 528000, China
| | - Zaowen Liao
- Cancer Research Institute, Foshan First People's Hospital, Foshan, Guangdong 528000, China
| | - Yuejian Wang
- Cancer Research Institute, Foshan First People's Hospital, Foshan, Guangdong 528000, China.
| | - Xiaosong Liu
- Cancer Research Institute, Foshan First People's Hospital, Foshan, Guangdong 528000, China; Inflammation and Healing Research Cluster, School of Health and Sport Sciences, University of Sunshine Coast, Maroochydore DC, QLD 4558, Australia.
| |
Collapse
|
22
|
Immunogénicité de la chimiothérapie. ONCOLOGIE 2015. [DOI: 10.1007/s10269-015-2543-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
23
|
Ni G, Wang T, Walton S, Zhu B, Chen S, Wu X, Wang Y, Wei MQ, Liu X. Manipulating IL-10 signalling blockade for better immunotherapy. Cell Immunol 2015; 293:126-9. [PMID: 25596475 DOI: 10.1016/j.cellimm.2014.12.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 12/29/2014] [Indexed: 01/21/2023]
Abstract
Interleukin 10 is a cytokine with the ability to reduce or terminate inflammation. Chronic viral infection, such as infection of chronic hepatitis B, hepatitis C and HIV, has increased levels of interleukin 10 in peripheral blood. Serum IL-10 levels are also high in certain cancers. Blocking IL-10 signalling at the time of immunisation clears chronic viral infection and prevents tumour growth in animal models. We review recent advances in this area, with the emphasis on potential use of this novel strategy to treat chronic viral infection and cancer in human.
Collapse
Affiliation(s)
- Guoying Ni
- School of Medical Science, Griffith Health Institute, Griffith University, Gold Coast, QLD 4222, Australia
| | - Tianfang Wang
- Genecology Research Centre, University of Sunshine Coast, Sippy Downs 4556, QLD, Australia
| | - Shelley Walton
- Inflammation and Healing Research Cluster, University of Sunshine Coast, Sippy Downs 4556, Australia
| | - Bin Zhu
- Cancer Research Institute, Foshan First People's Hospital, Foshan, Guangdong 528000, China
| | - Shu Chen
- Cancer Research Institute, Foshan First People's Hospital, Foshan, Guangdong 528000, China
| | - Xiaolian Wu
- Cancer Research Institute, Foshan First People's Hospital, Foshan, Guangdong 528000, China
| | - Yuejian Wang
- Cancer Research Institute, Foshan First People's Hospital, Foshan, Guangdong 528000, China.
| | - Ming Q Wei
- School of Medical Science, Griffith Health Institute, Griffith University, Gold Coast, QLD 4222, Australia.
| | - Xiaosong Liu
- Inflammation and Healing Research Cluster, University of Sunshine Coast, Sippy Downs 4556, Australia; Cancer Research Institute, Foshan First People's Hospital, Foshan, Guangdong 528000, China.
| |
Collapse
|