1
|
Jackson Hoffman BA, Pumford EA, Enueme AI, Fetah KL, Friedl OM, Kasko AM. Engineered macromolecular Toll-like receptor agents and assemblies. Trends Biotechnol 2023; 41:1139-1154. [PMID: 37068999 DOI: 10.1016/j.tibtech.2023.03.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 03/06/2023] [Accepted: 03/13/2023] [Indexed: 04/19/2023]
Abstract
Macromolecular Toll-like receptor (TLR) agents have been utilized as agonists and inhibitors in preclinical and clinical settings. These agents interface with the TLR class of innate immune receptors which recognize macromolecular ligands that are characteristic of pathogenic material. As such, many agents that have been historically investigated are derived from the natural macromolecules which activate or inhibit TLRs. This review covers recent research and clinically available TLR agents that are macromolecular or polymeric. Synthetic materials that have been found to interface with TLRs are also discussed. Assemblies of these materials are investigated in the context of improving stability or efficacy of ligands. Attention is given to strategies which modify or enhance the current agents and to future outlooks on the development of these agents.
Collapse
Affiliation(s)
| | - Elizabeth A Pumford
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Amaka I Enueme
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Kirsten L Fetah
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Olivia M Friedl
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Andrea M Kasko
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA; California Nanosystems Institute, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
2
|
Del Prete A, Salvi V, Soriani A, Laffranchi M, Sozio F, Bosisio D, Sozzani S. Dendritic cell subsets in cancer immunity and tumor antigen sensing. Cell Mol Immunol 2023; 20:432-447. [PMID: 36949244 PMCID: PMC10203372 DOI: 10.1038/s41423-023-00990-6] [Citation(s) in RCA: 227] [Impact Index Per Article: 113.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 02/14/2023] [Indexed: 03/24/2023] Open
Abstract
Dendritic cells (DCs) exhibit a specialized antigen-presenting function and play crucial roles in both innate and adaptive immune responses. Due to their ability to cross-present tumor cell-associated antigens to naïve T cells, DCs are instrumental in the generation of specific T-cell-mediated antitumor effector responses in the control of tumor growth and tumor cell dissemination. Within an immunosuppressive tumor microenvironment, DC antitumor functions can, however, be severely impaired. In this review, we focus on the mechanisms of DC capture and activation by tumor cell antigens and the role of the tumor microenvironment in shaping DC functions, taking advantage of recent studies showing the phenotype acquisition, transcriptional state and functional programs revealed by scRNA-seq analysis. The therapeutic potential of DC-mediated tumor antigen sensing in priming antitumor immunity is also discussed.
Collapse
Affiliation(s)
- Annalisa Del Prete
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Humanitas Clinical and Research Center-IRCCS Rozzano, Milano, Italy
| | - Valentina Salvi
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Alessandra Soriani
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Mattia Laffranchi
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Francesca Sozio
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Daniela Bosisio
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Silvano Sozzani
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy.
- IRCCS Neuromed, Pozzilli, IS, Italy.
| |
Collapse
|
3
|
Xie D, Wang Q, Wu G. Research progress in inducing immunogenic cell death of tumor cells. Front Immunol 2022; 13:1017400. [PMID: 36466838 PMCID: PMC9712455 DOI: 10.3389/fimmu.2022.1017400] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 11/02/2022] [Indexed: 08/29/2023] Open
Abstract
Immunogenic cell death (ICD) is a regulated cell death (RCD) pathway. In response to physical and chemical signals, tumor cells activate specific signaling pathways that stimulate stress responses in the endoplasmic reticulum (ER) and expose damage-associated molecular patterns (DAMPs), which promote antitumor immune responses. As a result, the tumor microenvironment is altered, and many tumor cells are killed. The ICD response in tumor cells requires inducers. These inducers can be from different sources and contribute to the development of the ICD either indirectly or directly. The combination of ICD inducers with other tumor treatments further enhances the immune response in tumor cells, and more tumor cells are killed; however, it also produces side effects of varying severity. New induction methods based on nanotechnology improve the antitumor ability and significantly reduces side effects because they can target tumor cells precisely. In this review, we introduce the characteristics and mechanisms of ICD responses in tumor cells and the DAMPs associated with ICD responses, summarize the current methods of inducing ICD response in tumor cells in five distinct categories: chemical sources, physical sources, pathogenic sources, combination therapies, and innovative therapies. At the same time, we introduce the limitations of current ICD inducers and make a summary of the use of ICD responses in clinical trials. Finally, we provide an outlook on the future of ICD inducer development and provide some constructive suggestions.
Collapse
Affiliation(s)
| | - Qifei Wang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
4
|
Yang J, Liu Y, Lin C, Yan R, Li Z, Chen Q, Zhang H, Xu H, Chen X, Chen Y, Guo A, Hu C. Regularity of Toll-Like Receptors in Bovine Mammary Epithelial Cells Induced by Mycoplasma bovis. Front Vet Sci 2022; 9:846700. [PMID: 35464378 PMCID: PMC9021453 DOI: 10.3389/fvets.2022.846700] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/15/2022] [Indexed: 11/13/2022] Open
Abstract
Mastitis is one of the most common and significant infectious diseases in dairy cattle and is responsible for significant financial losses for the dairy industry globally. An important pathogen of bovine mastitis, Mycoplasma bovis (M. bovis) has a high infection rate, requires a long course of treatment, and is difficult to cure. Bovine mammary epithelial cells (BMECs) are the first line of defense of the mammary gland, and their natural immune system plays a critical role in resisting M. bovis infection. This study aimed to explore and demonstrate the regularity of Toll-like receptors (TLRs) activation during M. bovis infection and their function during M. bovis mastitis. An in vitro model of M. bovis-induced mastitis showed that the expression of IL-6, IL-8, and TNF-α increased significantly following infection. M. bovis infection also upregulated the expression of TLR1/2/6 on the cell membrane and TLR3/9 in the cytoplasm. There is a crosstalk effect between TLR1–TLR2 and TLR2–TLR6. Furthermore, M. bovis infection was found to activate the TLR1/2/6/9/MyD88/NF-κB and TLR3/TRIF/IRF signal transduction pathways, which in turn activate inflammatory factors. These findings lay the theoretical foundation for understanding the pathogenesis of M. bovis, permitting the development of effective measures for preventing and controlling M. bovis mastitis.
Collapse
Affiliation(s)
- Jinghan Yang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yuhui Liu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Changjie Lin
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Rui Yan
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Zhengzhi Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Qiuhui Chen
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Haiyan Zhang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Haojun Xu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xi Chen
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yingyu Chen
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Aizhen Guo
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
| | - Changmin Hu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- *Correspondence: Changmin Hu
| |
Collapse
|
5
|
Mohammadzadeh R, Soleimanpour S, Pishdadian A, Farsiani H. Designing and development of epitope-based vaccines against Helicobacter pylori. Crit Rev Microbiol 2021; 48:489-512. [PMID: 34559599 DOI: 10.1080/1040841x.2021.1979934] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Helicobacter pylori infection is the principal cause of serious diseases (e.g. gastric cancer and peptic ulcers). Antibiotic therapy is an inadequate strategy in H. pylori eradication because of which vaccination is an inevitable approach. Despite the presence of countless vaccine candidates, current vaccines in clinical trials have performed with poor efficacy which makes vaccination extremely challenging. Remarkable advancements in immunology and pathogenic biology have provided an appropriate opportunity to develop various epitope-based vaccines. The fusion of proper antigens involved in different aspects of H. pylori colonization and pathogenesis as well as peptide linkers and built-in adjuvants results in producing epitope-based vaccines with excellent therapeutic efficacy and negligible adverse effects. Difficulties of the in vitro culture of H. pylori, high genetic variation, and unfavourable immune responses against feeble epitopes in the complete antigen are major drawbacks of current vaccine strategies that epitope-based vaccines may overcome. Besides decreasing the biohazard risk, designing precise formulations, saving time and cost, and induction of maximum immunity with minimum adverse effects are the advantages of epitope-based vaccines. The present article is a comprehensive review of strategies for designing and developing epitope-based vaccines to provide insights into the innovative vaccination against H. pylori.
Collapse
Affiliation(s)
- Roghayeh Mohammadzadeh
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Microbiology and Virology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saman Soleimanpour
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Reference Tuberculosis Laboratory, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abbas Pishdadian
- Department of Immunology, School of Medicine, Zabol University of Medical Sciences, Zabol, Iran
| | - Hadi Farsiani
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Microbiology and Virology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
6
|
Nuñez-Reza KJ, Naldi A, Sánchez-Jiménez A, Leon-Apodaca AV, Santana MA, Thomas-Chollier M, Thieffry D, Medina-Rivera A. Logical modelling of in vitro differentiation of human monocytes into dendritic cells unravels novel transcriptional regulatory interactions. Interface Focus 2021; 11:20200061. [PMID: 34123352 PMCID: PMC8193469 DOI: 10.1098/rsfs.2020.0061] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2021] [Indexed: 12/13/2022] Open
Abstract
Dendritic cells (DCs) are the major specialized antigen-presenting cells, thereby connecting innate and adaptive immunity. Because of their role in establishing adaptive immunity, they constitute promising targets for immunotherapy. Monocytes can differentiate into DCs in vitro in the presence of colony-stimulating factor 2 (CSF2) and interleukin 4 (IL4), activating four signalling pathways (MAPK, JAK/STAT, NFKB and PI3K). However, the downstream transcriptional programme responsible for DC differentiation from monocytes (moDCs) remains unknown. By analysing the scientific literature on moDC differentiation, we established a preliminary logical model that helped us identify missing information regarding the activation of genes responsible for this differentiation, including missing targets for key transcription factors (TFs). Using ChIP-seq and RNA-seq data from the Blueprint consortium, we defined active and inactive promoters, together with differentially expressed genes in monocytes, moDCs and macrophages, which correspond to an alternative cell fate. We then used this functional genomic information to predict novel targets for previously identified TFs. By integrating this information, we refined our model and recapitulated the main established facts regarding moDC differentiation. Prospectively, the resulting model should be useful to develop novel immunotherapies targeting moDCs.
Collapse
Affiliation(s)
- Karen J Nuñez-Reza
- Laboratorio Internacional de Investigación sobre el Genoma Humano, Universidad Nacional Autónoma de México, Juriquilla, México
| | - Aurélien Naldi
- Computational Systems Biology team, Institut de Biologie de l'École Normale Supérieure, Inserm, CNRS, Université PSL, Paris, France
| | - Arantza Sánchez-Jiménez
- Laboratorio Internacional de Investigación sobre el Genoma Humano, Universidad Nacional Autónoma de México, Juriquilla, México
| | - Ana V Leon-Apodaca
- Laboratorio Internacional de Investigación sobre el Genoma Humano, Universidad Nacional Autónoma de México, Juriquilla, México
| | - M Angélica Santana
- Centro de Investigación en Dinámica Celular (IICBA), Universidad Autónoma del Estado de Morelos, Cuernavaca, México
| | - Morgane Thomas-Chollier
- Computational Systems Biology team, Institut de Biologie de l'École Normale Supérieure, Inserm, CNRS, Université PSL, Paris, France
| | - Denis Thieffry
- Computational Systems Biology team, Institut de Biologie de l'École Normale Supérieure, Inserm, CNRS, Université PSL, Paris, France
| | - Alejandra Medina-Rivera
- Laboratorio Internacional de Investigación sobre el Genoma Humano, Universidad Nacional Autónoma de México, Juriquilla, México
| |
Collapse
|
7
|
Cho S, Kim SB, Lee Y, Song EC, Kim U, Kim HY, Suh JH, Goughnour PC, Kim Y, Yoon I, Shin NY, Kim D, Kim IK, Kang CY, Jang SY, Kim MH, Kim S. Endogenous TLR2 ligand embedded in the catalytic region of human cysteinyl-tRNA synthetase 1. J Immunother Cancer 2021; 8:jitc-2019-000277. [PMID: 32461342 PMCID: PMC7254149 DOI: 10.1136/jitc-2019-000277] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/24/2020] [Indexed: 12/21/2022] Open
Abstract
Background The generation of antigen-specific cytotoxic T lymphocyte (CTL) responses is required for successful cancer vaccine therapy. In this regard, ligands of Toll-like receptors (TLRs) have been suggested to activate adaptive immune responses by modulating the function of antigen-presenting cells (APCs). Despite their therapeutic potential, the development of TLR ligands for immunotherapy is often hampered due to rapid systemic toxicity. Regarding the safety concerns of currently available TLR ligands, finding a new TLR agonist with potent efficacy and safety is needed. Methods A unique structural domain (UNE-C1) was identified as a novel TLR2/6 in the catalytic region of human cysteinyl-tRNA synthetase 1 (CARS1) using comprehensive approaches, including RNA sequencing, the human embryonic kidney (HEK)-TLR Blue system, pull-down, and ELISA. The potency of its immunoadjuvant properties was analyzed by assessing antigen-specific antibody and CTL responses. In addition, the efficacy of tumor growth inhibition and the presence of the tumor-infiltrating leukocytes were evaluated using E.G7-OVA and TC-1 mouse models. The combined effect of UNE-C1 with an immune checkpoint inhibitor, anti-CTLA-4 antibody, was also evaluated in vivo. The safety of UNE-C1 immunization was determined by monitoring splenomegaly and cytokine production in the blood. Results Here, we report that CARS1 can be secreted from cancer cells to activate immune responses via specific interactions with TLR2/6 of APCs. A unique domain (UNE-C1) inserted into the catalytic region of CARS1 was determined to activate dendritic cells, leading to the stimulation of robust humoral and cellular immune responses in vivo. UNE-C1 also showed synergistic efficacy with cancer antigens and checkpoint inhibitors against different cancer models in vivo. Further, the safety assessment of UNE-C1 showed lower systemic cytokine levels than other known TLR agonists. Conclusions We identified the endogenous TLR2/6 activating domain from human cysteinyl-tRNA synthetase CARS1. This novel TLR2/6 ligand showed potent immune-stimulating activity with little toxicity. Thus, the UNE-C1 domain can be developed as an effective immunoadjuvant with checkpoint inhibitors or cancer antigens to boost antitumor immunity.
Collapse
Affiliation(s)
- Seongmin Cho
- Medicinal Bioconvergence Research Center and College of Pharmacy, Seoul National University, Suwon, South Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, and College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Sang Bum Kim
- Medicinal Bioconvergence Research Center and College of Pharmacy, Seoul National University, Suwon, South Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, and College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Youngjin Lee
- Infection and Immunity Research Laboratory, Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Ee Chan Song
- Medicinal Bioconvergence Research Center and College of Pharmacy, Seoul National University, Suwon, South Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, and College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Uijoo Kim
- Medicinal Bioconvergence Research Center and College of Pharmacy, Seoul National University, Suwon, South Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, and College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Hyeong Yun Kim
- Medicinal Bioconvergence Research Center and College of Pharmacy, Seoul National University, Suwon, South Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, and College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Ji Hun Suh
- Medicinal Bioconvergence Research Center and College of Pharmacy, Seoul National University, Suwon, South Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, and College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Peter C Goughnour
- Medicinal Bioconvergence Research Center and College of Pharmacy, Seoul National University, Suwon, South Korea
| | - YounHa Kim
- Medicinal Bioconvergence Research Center and College of Pharmacy, Seoul National University, Suwon, South Korea
| | - Ina Yoon
- Medicinal Bioconvergence Research Center and College of Pharmacy, Seoul National University, Suwon, South Korea
| | - Na Young Shin
- Medicinal Bioconvergence Research Center and College of Pharmacy, Seoul National University, Suwon, South Korea
| | - Doyeun Kim
- Medicinal Bioconvergence Research Center and College of Pharmacy, Seoul National University, Suwon, South Korea
| | - Il-Kyu Kim
- Laboratory of Immunology, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Chang-Yuil Kang
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, and College of Pharmacy, Seoul National University, Seoul, South Korea.,Laboratory of Immunology, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Song Yee Jang
- Infection and Immunity Research Laboratory, Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Myung Hee Kim
- Infection and Immunity Research Laboratory, Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Sunghoon Kim
- Medicinal Bioconvergence Research Center and College of Pharmacy, Seoul National University, Suwon, South Korea .,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, and College of Pharmacy, Seoul National University, Seoul, South Korea
| |
Collapse
|
8
|
Bunch BL, Kodumudi KN, Scott E, Morse J, Weber AM, Berglund AE, Pilon-Thomas S, Markowitz J. Anti-tumor efficacy of plasmid encoding emm55 in a murine melanoma model. Cancer Immunol Immunother 2020; 69:2465-2476. [PMID: 32556443 PMCID: PMC7680263 DOI: 10.1007/s00262-020-02634-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 06/04/2020] [Indexed: 12/31/2022]
Abstract
Emm55 is a bacterial gene derived from Streptococcus pyogenes (S. pyogenes) that was cloned into a plasmid DNA vaccine (pAc/emm55). In this study, we investigated the anti-tumor efficacy of pAc/emm55 in a B16 murine melanoma model. Intralesional (IL) injections of pAc/emm55 significantly delayed tumor growth compared to the pAc/Empty group. There was a significant increase in the CD8+ T cells infiltrating into the tumors after pAc/emm55 treatment compared to the control group. In addition, we observed that IL injection of pAc/emm55 increased antigen-specific T cell infiltration into tumors. Depletion of CD4+ or CD8+ T cells abrogated the anti-tumor effect of pAc/emm55. Combination treatment of IL injection of pAc/emm55 with anti-PD-1 antibody significantly delayed tumor growth compared to either monotherapy. pAc/emm55 treatment combined with PD-1 blockade enhanced anti-tumor immune response and improved systemic anti-tumor immunity. Together, these strategies may lead to improvements in the treatment of patients with melanoma.
Collapse
Affiliation(s)
- Brittany L Bunch
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Krithika N Kodumudi
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Ellen Scott
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Jennifer Morse
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Amy Mackay Weber
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Anders E Berglund
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
- Department of Oncologic Sciences, University of South Florida, Morsani College of Medicine, Tampa, FL, USA
| | - Shari Pilon-Thomas
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA.
- Department of Oncologic Sciences, University of South Florida, Morsani College of Medicine, Tampa, FL, USA.
- Cutaneous Oncology Program, H. Lee Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, SRB-4, Tampa, FL, 33612, USA.
- Center for Immunization and Infection Research in Cancer (CIIRC), H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| | - Joseph Markowitz
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA.
- Department of Oncologic Sciences, University of South Florida, Morsani College of Medicine, Tampa, FL, USA.
- Immunology Program, H. Lee Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, SRB-3, Tampa, FL, 33606, USA.
| |
Collapse
|
9
|
Detection of immunogenic cell death and its relevance for cancer therapy. Cell Death Dis 2020; 11:1013. [PMID: 33243969 PMCID: PMC7691519 DOI: 10.1038/s41419-020-03221-2] [Citation(s) in RCA: 607] [Impact Index Per Article: 121.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/06/2020] [Accepted: 11/09/2020] [Indexed: 02/07/2023]
Abstract
Chemotherapy, radiation therapy, as well as targeted anticancer agents can induce clinically relevant tumor-targeting immune responses, which critically rely on the antigenicity of malignant cells and their capacity to generate adjuvant signals. In particular, immunogenic cell death (ICD) is accompanied by the exposure and release of numerous damage-associated molecular patterns (DAMPs), which altogether confer a robust adjuvanticity to dying cancer cells, as they favor the recruitment and activation of antigen-presenting cells. ICD-associated DAMPs include surface-exposed calreticulin (CALR) as well as secreted ATP, annexin A1 (ANXA1), type I interferon, and high-mobility group box 1 (HMGB1). Additional hallmarks of ICD encompass the phosphorylation of eukaryotic translation initiation factor 2 subunit-α (EIF2S1, better known as eIF2α), the activation of autophagy, and a global arrest in transcription and translation. Here, we outline methodological approaches for measuring ICD markers in vitro and ex vivo for the discovery of next-generation antineoplastic agents, the development of personalized anticancer regimens, and the identification of optimal therapeutic combinations for the clinical management of cancer.
Collapse
|
10
|
Banstola A, Jeong JH, Yook S. Immunoadjuvants for cancer immunotherapy: A review of recent developments. Acta Biomater 2020; 114:16-30. [PMID: 32777293 DOI: 10.1016/j.actbio.2020.07.063] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/14/2020] [Accepted: 07/31/2020] [Indexed: 02/07/2023]
Abstract
Cancer immunotherapy evolved as a new treatment modality to eradicate tumor cells and has gained in popularity after its successful clinical transition. By activating antigen-presenting cells (APCs), and thus, inducing innate or adaptive immune responses, immunoadjuvants have become promising tools for cancer immunotherapy. Different types of immunoadjuvants such as toll-like receptor (TLR) agonists, exosomes, and metallic and plant-derived immunoadjuvants have been studied for their immunological effects. However, the clinical use of immunoadjuvants is limited by short response rates and various side-effects. The rapid progress made in the development of nanoparticle systems as immunoadjuvant carrier vehicles has provided potential carriers for cancer immunotherapy. In this review article, we describe different types of immunoadjuvants, their limitations, modes of action, and the reasons for their clinical adoption. In addition, we review recent progress made in the nanoparticle-based immunoadjuvant field and on the combined use of nanoparticle-based immunoadjuvants and chemotherapy, phototherapy, radiation therapy, and immune checkpoint inhibitor-based therapy. STATEMENT OF SIGNIFICANCE: Cancer immunotherapy emerged as a new hope for treating malignant tumors. Different types of immunoadjuvants serve as an important tool for cancer immunotherapy by activating an innate or adaptive immune response. Limitation of free immunoadjuvant has paved the path for the development of nanoparticle-based immunoadjuvant therapy with the hope of prolonging the therapeutic efficacy. This review highlights the recent advancement made in nanoparticle-based immunoadjuvant therapy in modulating the adaptive and innate immune system. The application of the combinatorial approach of chemotherapy, phototherapy, radiation therapy adds synergy in nanoparticle-based immunoadjuvant therapy. It will broaden the reader's understanding on the recent progress made in immunotherapy with the aid of immunoadjuvant-based nanosystem.
Collapse
Affiliation(s)
- Asmita Banstola
- College of Pharmacy, Keimyung University, Daegu, 42601, Republic of Korea
| | - Jee-Heon Jeong
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea.
| | - Simmyung Yook
- College of Pharmacy, Keimyung University, Daegu, 42601, Republic of Korea.
| |
Collapse
|
11
|
Zhao W, Bendickson L, Nilsen-Hamilton M. The Lipocalin2 Gene is Regulated in Mammary Epithelial Cells by NFκB and C/EBP In Response to Mycoplasma. Sci Rep 2020; 10:7641. [PMID: 32376831 PMCID: PMC7203223 DOI: 10.1038/s41598-020-63393-x] [Citation(s) in RCA: 5] [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: 08/27/2019] [Accepted: 03/25/2020] [Indexed: 02/02/2023] Open
Abstract
Lcn2 gene expression increases in response to cell stress signals, particularly in cells involved in the innate immune response. Human Lcn2 (NGAL) is increased in the blood and tissues in response to many stressors including microbial infection and in response to LPS in myeloid and epithelial cells. Here we extend the microbial activators of Lcn2 to mycoplasma and describe studies in which the mechanism of Lcn2 gene regulation by MALP-2 and mycoplasma infection was investigated in mouse mammary epithelial cells. As for the LPS response of myeloid cells, Lcn2 expression in epithelial cells is preceded by increased TNFα, IL-6 and IκBζ expression and selective reduction of IκBζ reduces Lcn2 promoter activity. Lcn2 promoter activation remains elevated well beyond the period of exposure to MALP-2 and is persistently elevated in mycoplasma infected cells. Activation of either the human or the mouse Lcn2 promoter requires both NFκB and C/EBP for activation. Thus, Lcn2 is strongly and enduringly activated by mycoplasma components that stimulate the innate immune response with the same basic regulatory mechanism for the human and mouse genes.
Collapse
Affiliation(s)
- Wei Zhao
- Roy J Carver Department of Biochemistry, Biophysics and Molecular Biology and the Interdepartmental Molecular, Cellular and Developmental Biology Program, Iowa State University, Ames, IA, 50011, USA
- Interdepartmental Molecular, Cellular and Developmental Biology Program, Iowa State University, Ames, IA, 50011, USA
- Bayview Physicians Group, Battlefield Medical association, 675 North Battlefield Boulevard, Chesapeake, VA, 23320, USA
| | - Lee Bendickson
- Roy J Carver Department of Biochemistry, Biophysics and Molecular Biology and the Interdepartmental Molecular, Cellular and Developmental Biology Program, Iowa State University, Ames, IA, 50011, USA
| | - Marit Nilsen-Hamilton
- Roy J Carver Department of Biochemistry, Biophysics and Molecular Biology and the Interdepartmental Molecular, Cellular and Developmental Biology Program, Iowa State University, Ames, IA, 50011, USA.
- Interdepartmental Molecular, Cellular and Developmental Biology Program, Iowa State University, Ames, IA, 50011, USA.
| |
Collapse
|
12
|
Kornspan JD, Kosower NS, Vaisid T, Katzhandler J, Rottem S. Novel synthetic lipopeptides derived from Mycoplasma hyorhinis upregulate calpastatin in SH-SY5Y neuroblastoma cells and induce a neuroprotective effect against amyloid-β-peptide toxicity. FEMS Microbiol Lett 2020; 367:5824629. [PMID: 32329786 DOI: 10.1093/femsle/fnaa073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 04/22/2020] [Indexed: 11/14/2022] Open
Abstract
Previously, we showed that contamination of SH-SY5Y neuroblastoma cells by Mycoplasma hyorhinis strains NDMh and MCLD leads to increased levels of calpastatin (the endogenous, specific inhibitor of the Ca2+-dependent protease calpain), resulting in inhibition of calpain activation. We have found that the increased calpastatin level is promoted by the lipoprotein fraction (MhLpp) of the mycoplasmal membrane. Here, we present MhLpp-based novel synthetic lipopeptides that induce upregulation of calpastatin in SH-SY5Y neuroblastoma cells, leading to protection of the treated cells against Ca2+/amyloid-β-peptide toxicity. These lipopeptides present a new class of promising agents against calpain-induced cell toxicity.
Collapse
Affiliation(s)
- Jonathan D Kornspan
- Department of Microbiology and Molecular Genetics, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Nechama S Kosower
- Department of Human Molecular Genetics & Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Tali Vaisid
- Department of Human Molecular Genetics & Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | | | - Shlomo Rottem
- Department of Microbiology and Molecular Genetics, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| |
Collapse
|
13
|
Yoshida S, Shime H, Matsumoto M, Kasahara M, Seya T. Anti-oxidative Amino Acid L-ergothioneine Modulates the Tumor Microenvironment to Facilitate Adjuvant Vaccine Immunotherapy. Front Immunol 2019; 10:671. [PMID: 31019508 PMCID: PMC6458301 DOI: 10.3389/fimmu.2019.00671] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 03/12/2019] [Indexed: 12/23/2022] Open
Abstract
Cancer vaccines consist of a tumor-associated antigen (TAA) and adjuvant. These vaccines induce and activate proliferation of TAA-specific cytotoxic T lymphocytes (CTLs), suppressing tumor growth. The therapeutic efficacy of TAA-specific CTLs depends on the properties of tumor microenvironment. The environments make immunosuppressive by function of regulatory T cells and tumor-associated myeloid cells; thus, regulation of these cells is important for successful cancer immunotherapy. We report here that L-ergothioneine (EGT) with the adjuvant Toll-like receptor 2 (TLR2) ligand modulated suppressive microenvironments to be immune-enhancing. EGT did not augment DC-mediated CTL priming or affect CTL activation in draining lymph node and spleen. However, EGT decreased the immuno-suppressive function of tumor-associated macrophages (TAMs). TLR2 stimulation accompanied with EGT administration downregulated expression of PD-L1, CSF-1R, arginase-1, FAS ligand, and TRAIL in TAMs, reflecting reduction of CTL suppression. An anti-oxidative thiol-thione residue of EGT was essential to dampening CTL suppression. The effect was specific to the thiol-thione residue of EGT because no effect was observed with another anti-oxidant N-acetyl-L-cysteine (NAC). A CTL-suppressive environment made by TLR2 is relieved to be improved by the addition of EGT, which may ameliorate the efficacy of vaccine immunotherapy.
Collapse
Affiliation(s)
- Sumito Yoshida
- Department of Vaccine Immunology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
- Department of Pathology I, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroaki Shime
- Department of Vaccine Immunology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
- Department of Immunology, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Misako Matsumoto
- Department of Vaccine Immunology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Masanori Kasahara
- Department of Pathology I, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Tsukasa Seya
- Department of Vaccine Immunology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
- Department of Immunology, School of Pharmaceutical Sciences, Aomori University, Aomori, Japan
| |
Collapse
|
14
|
Lei Y, Zhao F, Shao J, Li Y, Li S, Chang H, Zhang Y. Application of built-in adjuvants for epitope-based vaccines. PeerJ 2019; 6:e6185. [PMID: 30656066 PMCID: PMC6336016 DOI: 10.7717/peerj.6185] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 11/29/2018] [Indexed: 12/21/2022] Open
Abstract
Several studies have shown that epitope vaccines exhibit substantial advantages over conventional vaccines. However, epitope vaccines are associated with limited immunity, which can be overcome by conjugating antigenic epitopes with built-in adjuvants (e.g., some carrier proteins or new biomaterials) with special properties, including immunologic specificity, good biosecurity and biocompatibility, and the ability to vastly improve the immune response of epitope vaccines. When designing epitope vaccines, the following types of built-in adjuvants are typically considered: (1) pattern recognition receptor ligands (i.e., toll-like receptors); (2) virus-like particle carrier platforms; (3) bacterial toxin proteins; and (4) novel potential delivery systems (e.g., self-assembled peptide nanoparticles, lipid core peptides, and polymeric or inorganic nanoparticles). This review primarily discusses the current and prospective applications of these built-in adjuvants (i.e., biological carriers) to provide some references for the future design of epitope-based vaccines.
Collapse
Affiliation(s)
- Yao Lei
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Furong Zhao
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Junjun Shao
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yangfan Li
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Shifang Li
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Huiyun Chang
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yongguang Zhang
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| |
Collapse
|
15
|
Takeda Y, Azuma M, Hatsugai R, Fujimoto Y, Hashimoto M, Fukase K, Matsumoto M, Seya T. The second and third amino acids of Pam2 lipopeptides are key for the proliferation of cytotoxic T cells. Innate Immun 2018; 24:323-331. [PMID: 29848176 PMCID: PMC6830919 DOI: 10.1177/1753425918777598] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The TLR2 agonist, dipalmitoyl lipopeptide (Pam2LP), has been used as an immune
adjuvant without much success. Pam2LP is recognised by TLR2/6 receptors in
humans and in mice. This study examined the proliferative activity of cytotoxic
T lymphocytes (CTL) using mouse Ag-presenting dendritic cells (DCs) and OT-I
assay system, where a library of synthetic Pam2LP was utilised from the
Staphylococcus aureus database. Ag-specific CTL expansion
and IFN-γ levels largely depended on the Pam2LP peptide sequence. The first Aa
is cysteine (Cys), which has an active SH residue to bridge fatty acids, and the
second and third Aa are hydrophilic or non-polar. The sequence structurally
adapted to the residual constitution of the reported TLR2/6 pocket. The inactive
sequence contained proline or leucine/isoleucine after the first Cys. Notably,
no direct activation of OT-I cells was detected without DCs by stimulation with
the active Pam2LP having the Cys-Ser sequence. MyD88, but not TICAM-1 or IFN
pathways, in DCs participates in DC maturation characterised by upregulation of
CD40, CD80 and CD86. Hence, the active Pam2LPs appear suitable for dimeric
TLR2/6 on DCs, resulting in induction of DC maturation.
Collapse
Affiliation(s)
- Yohei Takeda
- 1 Department of Vaccine Immunology, Graduate School of Medicine, Hokkaido University, Japan
| | - Masahiro Azuma
- 1 Department of Vaccine Immunology, Graduate School of Medicine, Hokkaido University, Japan
| | - Ryoko Hatsugai
- 1 Department of Vaccine Immunology, Graduate School of Medicine, Hokkaido University, Japan
| | - Yukari Fujimoto
- 2 Faculty of Science and Technology, Keio University, Japan.,3 Department of Chemistry, Graduate School of Science, Osaka University, Japan
| | - Masahito Hashimoto
- 4 Department of Nanostructure and Advanced Materials, Kagoshima University, Japan
| | - Koichi Fukase
- 3 Department of Chemistry, Graduate School of Science, Osaka University, Japan
| | - Misako Matsumoto
- 1 Department of Vaccine Immunology, Graduate School of Medicine, Hokkaido University, Japan
| | - Tsukasa Seya
- 1 Department of Vaccine Immunology, Graduate School of Medicine, Hokkaido University, Japan
| |
Collapse
|