1
|
Yu W, Yuan X, Zhai P, Li X, Han C. Effects of Trichinella spiralis excretory-secretory antigens on expression of indoleamine 2, 3-dioxygenase on dendritic cells in vitro. Parasite 2025; 32:26. [PMID: 40239041 PMCID: PMC12002673 DOI: 10.1051/parasite/2025018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 03/27/2025] [Indexed: 04/18/2025] Open
Abstract
Indoleamine 2, 3-dioxygenase (IDO) is a potent immunoenzyme found in dendritic cells (DCs). Research has demonstrated that Trichinella spiralis induces IDO expression in the host immune response through its excretory-secretory (ES) antigens. However, the role of IDO in the immune response to T. spiralis remains unclear. To examine the effects of T. spiralis ES antigens on IDO expression in DCs in vitro, assessments were conducted using qRT-PCR, Western blotting (WB), flow cytometry, and siRNA transfer. The findings indicated that ES antigen stimulation upregulated IDO expression in DCs in vitro. Furthermore, ES antigen significantly enhanced the expression of the proinflammatory cytokines TNF-α and IFN-γ, along with the anti-inflammatory cytokine IL-10, downstream of IDO in DCs. Flow cytometry analysis confirmed that surface molecules CD40, MHC-II, CD80, and CD86 on DCs were upregulated following stimulation with ES antigen and lipopolysaccharide (LPS). Compared to the ES antigen alone, siRNA620 effectively inhibited IDO levels, demonstrating a statistically significant reduction. Continuous stimulation of DCs by ES antigens may lead to immune tolerance through the activation of IDO-mediated inflammation-associated factors. These results suggest that IDO expression in DCs plays a crucial role in T. spiralis infection.
Collapse
Affiliation(s)
- Wenhao Yu
- College of Veterinary Medicine, Northeast Agricultural University Harbin China
| | - Xuhong Yuan
- College of Veterinary Medicine, Northeast Agricultural University Harbin China
| | - Peng Zhai
- College of Veterinary Medicine, Northeast Agricultural University Harbin China
| | - Xiaoyun Li
- College of Veterinary Medicine, Northeast Agricultural University Harbin China
| | - Caixia Han
- College of Veterinary Medicine, Northeast Agricultural University Harbin China
| |
Collapse
|
2
|
Wang R, Lan C, Benlagha K, Camara NOS, Miller H, Kubo M, Heegaard S, Lee P, Yang L, Forsman H, Li X, Zhai Z, Liu C. The interaction of innate immune and adaptive immune system. MedComm (Beijing) 2024; 5:e714. [PMID: 39286776 PMCID: PMC11401974 DOI: 10.1002/mco2.714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 08/11/2024] [Accepted: 08/11/2024] [Indexed: 09/19/2024] Open
Abstract
The innate immune system serves as the body's first line of defense, utilizing pattern recognition receptors like Toll-like receptors to detect pathogens and initiate rapid response mechanisms. Following this initial response, adaptive immunity provides highly specific and sustained killing of pathogens via B cells, T cells, and antibodies. Traditionally, it has been assumed that innate immunity activates adaptive immunity; however, recent studies have revealed more complex interactions. This review provides a detailed dissection of the composition and function of the innate and adaptive immune systems, emphasizing their synergistic roles in physiological and pathological contexts, providing new insights into the link between these two forms of immunity. Precise regulation of both immune systems at the same time is more beneficial in the fight against immune-related diseases, for example, the cGAS-STING pathway has been found to play an important role in infections and cancers. In addition, this paper summarizes the challenges and future directions in the field of immunity, including the latest single-cell sequencing technologies, CAR-T cell therapy, and immune checkpoint inhibitors. By summarizing these developments, this review aims to enhance our understanding of the complexity interactions between innate and adaptive immunity and provides new perspectives in understanding the immune system.
Collapse
Affiliation(s)
- Ruyuan Wang
- Department of Thyroid and Breast Surgery Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Caini Lan
- Cancer Center Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Kamel Benlagha
- Alloimmunity, Autoimmunity and Transplantation Université de Paris, Institut de Recherche Saint-Louis, EMiLy, INSERM U1160 Paris France
| | - Niels Olsen Saraiva Camara
- Department of Immunology Institute of Biomedical Sciences University of São Paulo (USP) São Paulo São Paulo Brazil
| | - Heather Miller
- Coxiella Pathogenesis Section, Laboratory of Bacteriology Rocky Mountain Laboratories National Institute of Allergy and Infectious Diseases, National Institutes of Health Hamilton Montana USA
| | - Masato Kubo
- Division of Molecular Pathology Research Institute for Biomedical Sciences (RIBS) Tokyo University of Science Noda Chiba Japan
| | - Steffen Heegaard
- Department of Ophthalmology Rigshospitalet Hospital Copenhagen University Copenhagen Denmark
| | - Pamela Lee
- Department of Paediatrics and Adolescent Medicine Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong China
| | - Lu Yang
- Department of Pathogen Biology School of Basic Medicine Tongji Medical College and State Key Laboratory for Diagnosis and treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology Wuhan Hubei China
| | - Huamei Forsman
- Department of Laboratory Medicine Institute of Biomedicine, University of Gothenburg Gothenburg Sweden
| | - Xingrui Li
- Department of Thyroid and Breast Surgery Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Zhimin Zhai
- Department of Hematology The Second Hospital of Anhui Medical University Hefei China
| | - Chaohong Liu
- Department of Pathogen Biology School of Basic Medicine Tongji Medical College and State Key Laboratory for Diagnosis and treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology Wuhan Hubei China
| |
Collapse
|
3
|
Yi M, Li T, Niu M, Mei Q, Zhao B, Chu Q, Dai Z, Wu K. Exploiting innate immunity for cancer immunotherapy. Mol Cancer 2023; 22:187. [PMID: 38008741 PMCID: PMC10680233 DOI: 10.1186/s12943-023-01885-w] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/23/2023] [Indexed: 11/28/2023] Open
Abstract
Immunotherapies have revolutionized the treatment paradigms of various types of cancers. However, most of these immunomodulatory strategies focus on harnessing adaptive immunity, mainly by inhibiting immunosuppressive signaling with immune checkpoint blockade, or enhancing immunostimulatory signaling with bispecific T cell engager and chimeric antigen receptor (CAR)-T cell. Although these agents have already achieved great success, only a tiny percentage of patients could benefit from immunotherapies. Actually, immunotherapy efficacy is determined by multiple components in the tumor microenvironment beyond adaptive immunity. Cells from the innate arm of the immune system, such as macrophages, dendritic cells, myeloid-derived suppressor cells, neutrophils, natural killer cells, and unconventional T cells, also participate in cancer immune evasion and surveillance. Considering that the innate arm is the cornerstone of the antitumor immune response, utilizing innate immunity provides potential therapeutic options for cancer control. Up to now, strategies exploiting innate immunity, such as agonists of stimulator of interferon genes, CAR-macrophage or -natural killer cell therapies, metabolic regulators, and novel immune checkpoint blockade, have exhibited potent antitumor activities in preclinical and clinical studies. Here, we summarize the latest insights into the potential roles of innate cells in antitumor immunity and discuss the advances in innate arm-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Ming Yi
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, People's Republic of China
- Department of Breast Surgery, College of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, 310000, People's Republic of China
| | - Tianye Li
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310000, People's Republic of China
| | - Mengke Niu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China
| | - Qi Mei
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, People's Republic of China
| | - Bin Zhao
- Department of Breast Surgery, College of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, 310000, People's Republic of China
| | - Qian Chu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China.
| | - Zhijun Dai
- Department of Breast Surgery, College of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, 310000, People's Republic of China.
| | - Kongming Wu
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, People's Republic of China.
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China.
| |
Collapse
|
4
|
Sioud M, Olberg A. Antibody Surface Profiling Identifies Glycoforms in Multiple Myeloma as Targets for Immunotherapy: From Antibody Derivatives to Mimetic Peptides for Killing Tumor Cells. Cancers (Basel) 2023; 15:cancers15071934. [PMID: 37046595 PMCID: PMC10093763 DOI: 10.3390/cancers15071934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/15/2023] [Accepted: 03/18/2023] [Indexed: 04/14/2023] Open
Abstract
Despite therapeutic advances in recent years, there are still unmet medical needs for patients with multiple myeloma (MM). Hence, new therapeutic strategies are needed. Using phage display for screening a large repertoire of single chain variable fragments (scFvs), we isolated several candidates that recognize a heavily sulfated MM-specific glycoform of the surface antigen syndecan-1 (CD138). One of the engineered scFv-Fc antibodies, named MM1, activated NK cells and induced antibody-dependent cellular cytotoxicity against MM cells. Analysis of the binding specificity by competitive binding assays with various glycan ligands identified N-sulfation of glucosamine units as essential for binding. Additionally, site-directed mutagenesis revealed that the amino acids arginine and histidine in the complementarily determining regions (CDRs) 2 and 3 of the heavy chain are important for binding. Based on this observation, a heavy-chain antibody, known as a nanobody, and a peptide mimicking the CDR loop sequences were designed. Both variants exhibited high affinity and specificity to MM cells as compared to blood lymphocytes. Specific killing of MM cells was achieved by conjugating the CDR2/3 mimic peptide to a pro-apoptotic peptide (KLAKLAK)2. In a co-culture model, the fusion peptide killed MM cells, while leaving normal peripheral blood mononuclear cells unaffected. Collectively, the development of antibodies and peptides that detect tumor-specific glycoforms of therapeutic targets holds promise for improving targeted therapies and tumor imaging.
Collapse
Affiliation(s)
- Mouldy Sioud
- Department of Cancer Immunology, Division of Cancer Medicine, Oslo University Hospital-Radiumhospitalet, Ullernchausseen 70, 0379 Oslo, Norway
| | - Anniken Olberg
- Department of Cancer Immunology, Division of Cancer Medicine, Oslo University Hospital-Radiumhospitalet, Ullernchausseen 70, 0379 Oslo, Norway
| |
Collapse
|
5
|
Shadbad MA, Hajiasgharzadeh K, Derakhshani A, Silvestris N, Baghbanzadeh A, Racanelli V, Baradaran B. From Melanoma Development to RNA-Modified Dendritic Cell Vaccines: Highlighting the Lessons From the Past. Front Immunol 2021; 12:623639. [PMID: 33692796 PMCID: PMC7937699 DOI: 10.3389/fimmu.2021.623639] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/26/2021] [Indexed: 12/11/2022] Open
Abstract
Although melanoma remains the deadliest skin cancer, the current treatment has not resulted in the desired outcomes. Unlike chemotherapy, immunotherapy has provided more tolerable approaches and revolutionized cancer therapy. Although dendritic cell-based vaccines have minor side effects, the undesirable response rates of traditional approaches have posed questions about their clinical translation. The immunosuppressive tumor microenvironment can be the underlying reason for their low response rates. Immune checkpoints and indoleamine 2,3-dioxygenase have been implicated in the induction of immunosuppressive tumor microenvironment. Growing evidence indicates that the mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase/Protein kinase B (PKB) (PI3K/AKT) pathways, as the main oncogenic pathways of melanoma, can upregulate the tumoral immune checkpoints, like programmed death-ligand 1. This study briefly represents the main oncogenic pathways of melanoma and highlights the cross-talk between these oncogenic pathways with indoleamine 2,3-dioxygenase, tumoral immune checkpoints, and myeloid-derived suppressor cells. Moreover, this study sheds light on a novel tumor antigen on melanoma, which has substantial roles in tumoral immune checkpoints expression, indoleamine 2,3-dioxygenase secretion, and stimulating the oncogenic pathways. Finally, this review collects the lessons from the previous unsuccessful trials and integrates their lessons with new approaches in RNA-modified dendritic cell vaccines. Unlike traditional approaches, the advances in single-cell RNA-sequencing techniques and RNA-modified dendritic cell vaccines along with combined therapy of the immune checkpoint inhibitors, indoleamine 2,3-dioxygenase inhibitor, and RNA-modified dendritic cell-based vaccine can overcome these auto-inductive loops and pave the way for developing robust dendritic cell-based vaccines with the most favorable response rate and the least side effects.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/adverse effects
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/therapeutic use
- Cancer Vaccines/adverse effects
- Cancer Vaccines/genetics
- Cancer Vaccines/immunology
- Cancer Vaccines/therapeutic use
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Dendritic Cells/transplantation
- Humans
- Immune Checkpoint Proteins/metabolism
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- Melanoma/genetics
- Melanoma/immunology
- Melanoma/metabolism
- Melanoma/therapy
- Myeloid-Derived Suppressor Cells/immunology
- Myeloid-Derived Suppressor Cells/metabolism
- RNA, Small Interfering/adverse effects
- RNA, Small Interfering/genetics
- RNA, Small Interfering/immunology
- RNA, Small Interfering/therapeutic use
- Signal Transduction
- Skin Neoplasms/genetics
- Skin Neoplasms/immunology
- Skin Neoplasms/metabolism
- Skin Neoplasms/therapy
- Tumor Escape
- Tumor Microenvironment
- Vaccines, Synthetic/adverse effects
- Vaccines, Synthetic/therapeutic use
- mRNA Vaccines
Collapse
Affiliation(s)
- Mahdi Abdoli Shadbad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Afshin Derakhshani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Tumori “Giovanni Paolo II” of Bari, Bari, Italy
| | - Nicola Silvestris
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Tumori “Giovanni Paolo II” of Bari, Bari, Italy
- Department of Biomedical Sciences and Human Oncology, Aldo Moro University of Bari, Bari, Italy
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vito Racanelli
- Department of Biomedical Sciences and Human Oncology, Aldo Moro University of Bari, Bari, Italy
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
6
|
Wu C, Zhu W, Jin R, Ai H, Xu Y. The MRI-Visible Nanocomposite Facilitates the Delivery and Tracking of siRNA Loaded DC Vaccine in the Breast Cancer Model. Front Oncol 2021; 10:621642. [PMID: 33614503 PMCID: PMC7892972 DOI: 10.3389/fonc.2020.621642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 12/14/2020] [Indexed: 11/24/2022] Open
Abstract
Dendritic cell (DC) vaccines have recently been developed for the treatment of various cancers but often do not function as well as expected, primarily due to the highly complex in vivo immune environment. This proof-of-principle study aimed to test the feasibility of modulating the in vivo behaviors of DC vaccines (DCVs) by introducing siRNA-laden magnetic resonance (MR) imaging nanovectors into cells, while providing visible information on their homing to lymph nodes. The N-alkyl-PEI2k-LAC/SPIO nanocomposites were prepared and characterized, showing favorable properties of siRNA transfection and MRI labeling efficiency in DCs. Cell viability assays revealed no observable effects on the survival and phenotype of DCs if the concentration of the complex was within 8 μg Fe/ml. An orthotopic mouse model of breast cancer was developed. The DCVs transfected with IDO siRNA contained nanocomposites were adoptively transferred to start the treatment. MR imaging clearly visualized the homing of DCVs into lymph nodes. At the end of the treatment, DCVs presented significantly better tumor suppression than DCs or PBS (P < 0.05). Generally, the N-alkyl-PEI2k-LAC/SPIO nanocomposites represent a highly efficient MR imaging platform for siRNA transfection that is potentially useful for in vivo tracking of vaccine cells.
Collapse
Affiliation(s)
- Changqiang Wu
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China.,Sichuan Key Laboratory of Medical Imaging and School of Medical Imaging, North Sichuan Medical College, Nanchong, China
| | - Wencheng Zhu
- National Engineering Research Center for Biomaterials Sichuan University, Chengdu, China.,Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Rongrong Jin
- National Engineering Research Center for Biomaterials Sichuan University, Chengdu, China
| | - Hua Ai
- National Engineering Research Center for Biomaterials Sichuan University, Chengdu, China
| | - Ye Xu
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| |
Collapse
|
7
|
Heidari F, Ramezani A, Erfani N, Razmkhah M. Indoleamine 2, 3-Dioxygenase: A Professional Immunomodulator and Its Potential Functions in Immune Related Diseases. Int Rev Immunol 2020; 41:346-363. [DOI: 10.1080/08830185.2020.1836176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Fahimeh Heidari
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amin Ramezani
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nasrollah Erfani
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahboobeh Razmkhah
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
8
|
Improving Dendritic Cell Cancer Vaccine Potency Using RNA Interference. Methods Mol Biol 2020. [PMID: 32006405 DOI: 10.1007/978-1-0716-0290-4_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Dendritic cell cancer vaccines have already become a treatment modality for patients with various cancer types. However, the curative potential of this immunotherapy is limited by the existence of negative feedback mechanisms that control dendritic cells (DCs) and T-cell function. By inhibiting the expression of inhibitory factors using RNA interference technology, a new generation of DC vaccines was developed. Vaccine-stimulated T cells showed antitumor effects both in vitro and in cancer patients. Here, we describe the development and validation of a fully GMP-compliant production process of ex vivo DC cancer vaccines combined with the blockade of immunosuppressive pathways using small interfering RNAs. The protocol can be used for DC-based therapy for all cancer types.
Collapse
|
9
|
Optimized siRNA Delivery into Primary Immune Cells Using Electroporation. Methods Mol Biol 2020; 2115:119-131. [PMID: 32006398 DOI: 10.1007/978-1-0716-0290-4_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Effective RNA delivery strategies for primary human monocytes and dendritic cells (DCs) are useful tools for both basic research and cancer immunotherapy applications. Compared to viral delivery, electroporation is a relatively safe and simple technique that has been established for most immune cells. This chapter describes the feasibility of introducing small interfering RNAs into human primary monocytes and DCs using either nucleofection or standard electroporation techniques. DC cancer vaccines that integrate siRNA targeting relevant DC-intrinsic immunosuppressive signals induced robust and durable anti-tumor immune responses.
Collapse
|
10
|
Sioud M. Unleashing the Therapeutic Potential of Dendritic and T Cell Therapies Using RNA Interference. Methods Mol Biol 2020; 2115:259-280. [PMID: 32006406 DOI: 10.1007/978-1-0716-0290-4_15] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Therapeutic dendritic cell (DC) cancer vaccines work to boost the body's immune system to fight a cancer. Although this type of immunotherapy often leads to the activation of tumor-specfic T cells, clinical responses are fairly low, arguing for the need to improve the design of DC-based vaccines. Recent studies revealed a promising strategy of combining DC vaccines with small interfering RNAs (siRNAs) targeting immunosuppressive signals such as checkpoint receptors. Similarly, incorporating checkpoint siRNA blockers in adoptive T-cell therapy to amplify cytotoxic T lymphocyte responses is now being tested in the clinic. The development of the next generation of cancer immunotherapies using siRNA technology will hopefuly benefit patients with various cancer types including those who did not respond to current therapies. This review highlights the latest advances in RNA interference technology to improve the therapeutic efficacy of DC cancer vaccines and T cell therapy.
Collapse
Affiliation(s)
- Mouldy Sioud
- Department of Immunology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Ullernchausseen 70, Oslo, Norway.
| |
Collapse
|
11
|
Sioud M. Releasing the Immune System Brakes Using siRNAs Enhances Cancer Immunotherapy. Cancers (Basel) 2019; 11:cancers11020176. [PMID: 30717461 PMCID: PMC6406640 DOI: 10.3390/cancers11020176] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 01/29/2019] [Accepted: 01/31/2019] [Indexed: 12/12/2022] Open
Abstract
Therapeutic dendritic cell (DC) cancer vaccines rely on the immune system to eradicate tumour cells. Although tumour antigen-specific T cell responses have been observed in most studies, clinical responses are fairly low, arguing for the need to improve the design of DC-based vaccines. The incorporation of small interfering RNAs (siRNAs) against immunosuppressive factors in the manufacturing process of DCs can turn the vaccine into potent immune stimulators. Additionally, siRNA modification of ex vivo-expanded T cells for adoptive immunotherapy enhanced their killing potency. Most of the siRNA-targeted immune inhibitory factors have been successful in that their blockade produced the strongest cytotoxic T cell responses in preclinical and clinical studies. Cancer patients treated with the siRNA-modified DC vaccines showed promising clinical benefits providing a strong rationale for further development of these immunogenic vaccine formulations. This review covers the progress in combining siRNAs with DC vaccines or T cell therapy to boost anti-tumour immunity.
Collapse
Affiliation(s)
- Mouldy Sioud
- Department of Immunology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Montebello, N-0310 Oslo, Norway.
| |
Collapse
|
12
|
Liu Y, Yao L, Cao W, Liu Y, Zhai W, Wu Y, Wang B, Gou S, Qin Y, Qi Y, Chen Z, Gao Y. Dendritic Cell Targeting Peptide-Based Nanovaccines for Enhanced Cancer Immunotherapy. ACS APPLIED BIO MATERIALS 2019; 2:1241-1254. [DOI: 10.1021/acsabm.8b00811] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Yating Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Lintong Yao
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Wenpeng Cao
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yajing Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Wenjie Zhai
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yahong Wu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Binglin Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Shanshan Gou
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yaping Qin
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yuanming Qi
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
- Collaborative
Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou, Henan 450001, China
| | - Zhenzhen Chen
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
- Collaborative
Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou, Henan 450001, China
| | - Yanfeng Gao
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
- Collaborative
Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou, Henan 450001, China
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Shenzhen 518107, China
| |
Collapse
|
13
|
Liu YH, Yeh IJ, Lai MD, Liu KT, Kuo PL, Yen MC. Cancer Immunotherapy: Silencing Intracellular Negative Immune Regulators of Dendritic Cells. Cancers (Basel) 2019; 11:cancers11010108. [PMID: 30658461 PMCID: PMC6357062 DOI: 10.3390/cancers11010108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/09/2019] [Accepted: 01/13/2019] [Indexed: 01/26/2023] Open
Abstract
Dendritic cells (DCs) are capable of activating adaptive immune responses, or inducing immune suppression or tolerance. In the tumor microenvironment, the function of DCs is polarized into immune suppression that attenuates the effect of T cells, promoting differentiation of regulatory T cells and supporting tumor progression. Therefore, blocking negative immune regulators in DCs is considered a strategy of cancer immunotherapy. Antibodies can target molecules on the cell surface, but not intracellular molecules of DCs. The delivery of short-hairpin RNAs (shRNA) and small-interfering RNAs (siRNA) should be a strategy to silence specific intracellular targets in DCs. This review provides an overview of the known negative immune regulators of DCs. Moreover, a combination of shRNA/siRNA and DC vaccines, DNA vaccines in animal models, and clinical trials are also discussed.
Collapse
Affiliation(s)
- Yao-Hua Liu
- Department of Emergency Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - I-Jeng Yeh
- Department of Emergency Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Ming-Derg Lai
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan.
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan.
| | - Kuan-Ting Liu
- Department of Emergency Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Po-Lin Kuo
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Meng-Chi Yen
- Department of Emergency Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| |
Collapse
|
14
|
Liu DQ, Lu S, Zhang LX, Ji M, Liu SY, Wang SW, Liu RT. An indoleamine 2, 3-dioxygenase siRNA nanoparticle-coated and Trp2-displayed recombinant yeast vaccine inhibits melanoma tumor growth in mice. J Control Release 2018; 273:1-12. [DOI: 10.1016/j.jconrel.2018.01.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 12/15/2017] [Accepted: 01/16/2018] [Indexed: 10/18/2022]
|
15
|
Xie Y, Yan JF, Ma JY, Li HY, Ye YC, Zhang YS, Zhang H. Evaluation of the toxicity of iron-ion irradiation in murine bone marrow dendritic cells via increasing the expression of indoleamine 2,3-dioxygenase 1. Toxicol Res (Camb) 2017; 6:958-968. [PMID: 30090556 PMCID: PMC6061850 DOI: 10.1039/c7tx00194k] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 09/24/2017] [Indexed: 12/13/2022] Open
Abstract
High linear energy transfer radiation is known to deposit higher energy in tissues and cause greater toxicity compared to low-LET irradiation. Local immunosuppression is frequently observed after irradiation (IR). Dendritic cells (DCs) play important roles in the initiation and maintenance of the immune response. The dysfunction of DCs contributes to tumor evasion and growth. However, molecular mechanisms underlying the establishment of immune tolerance induced by heavy ion IR through this DC population are poorly understood. Therefore, here we report our findings on the dysfunction of bone marrow-derived dendritic cells (BMDCs) induced by 1 Gy iron ion radiation and promotions of expressions of JNK1/2/3, indoleamine 2,3-dioxygenase 1 (IDO1), p-ERK1/2 and p38/MAPK; and decrease of IDO2, MHC class II, CD40, CD80 expressions and IFN-γ and TNF-α secretion after total-body IR in mice. JNK+IDO1+ BMDCs showed up-expression of p-ERK1/2 and p-p38/MAPK, reduced expression of MHC class II and CD80, and were not able to effectively stimulate allogeneic spleen T cells. The inhibition of IDO1 expressions could partly restore the function of BMDCs. In all, our study shows that elevated JNK and IDO1 expression induced by Fe ion IR could result in dysfunction of BMDCs via p-p38/MAPK and p-ERK1/2 signal pathway, and it may represent a new mechanism in radiation-induced immune tolerance.
Collapse
Affiliation(s)
- Yi Xie
- Institute of Modern Physics , Chinese Academy of Sciences , 509 Nanchang Road , Lanzhou 730000 , China . ; ; Tel: +86 931 4969344
- CAS Key Laboratory of Heavy Ion Radiation Biology and Medicine , Institute of Modern Physics , Lanzhou 730000 , Gansu , China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine , Gansu Province , Lanzhou 730000 , China
| | - Jun-Fang Yan
- Institute of Modern Physics , Chinese Academy of Sciences , 509 Nanchang Road , Lanzhou 730000 , China . ; ; Tel: +86 931 4969344
- CAS Key Laboratory of Heavy Ion Radiation Biology and Medicine , Institute of Modern Physics , Lanzhou 730000 , Gansu , China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine , Gansu Province , Lanzhou 730000 , China
- Graduate School of University of Chinese Academy of Sciences , Beijing 100039 , China
| | - Jing-Yi Ma
- School of Pharmaceutical Sciences , Sun Yat-sen University , Guangzhou 510006 , China
| | - Hong-Yan Li
- Institute of Modern Physics , Chinese Academy of Sciences , 509 Nanchang Road , Lanzhou 730000 , China . ; ; Tel: +86 931 4969344
- CAS Key Laboratory of Heavy Ion Radiation Biology and Medicine , Institute of Modern Physics , Lanzhou 730000 , Gansu , China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine , Gansu Province , Lanzhou 730000 , China
| | - Yan-Cheng Ye
- Gansu Wuwei Tumor Hospital , Wuwei , 733000 , China
| | | | - Hong Zhang
- Institute of Modern Physics , Chinese Academy of Sciences , 509 Nanchang Road , Lanzhou 730000 , China . ; ; Tel: +86 931 4969344
- CAS Key Laboratory of Heavy Ion Radiation Biology and Medicine , Institute of Modern Physics , Lanzhou 730000 , Gansu , China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine , Gansu Province , Lanzhou 730000 , China
- Gansu Wuwei Tumor Hospital , Wuwei , 733000 , China
| |
Collapse
|
16
|
Meng Z, Lu M. RNA Interference-Induced Innate Immunity, Off-Target Effect, or Immune Adjuvant? Front Immunol 2017; 8:331. [PMID: 28386261 PMCID: PMC5362589 DOI: 10.3389/fimmu.2017.00331] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 03/08/2017] [Indexed: 12/24/2022] Open
Abstract
RNA interference (RNAi) is a natural cellular mechanism that inhibits gene expression in a sequence-specific manner. In the last decade, RNAi has become a cornerstone in basic biological systems research and drug development efforts. The RNAi-based manipulation of mammalian cells facilitates target identification and validation; assists in identifying human disease etiologies; and expedites the development of treatments for infectious diseases, cancer, and other conditions. Several RNAi-based approaches are currently undergoing assessment in phase I and II clinical trials. However, RNAi-associated immune stimulation might act as a hurdle to safe and effective RNAi, particularly in clinical applications. The induction of innate immunity may originate from small interfering RNA (siRNA) sequence-dependent delivery vehicles and even the RNAi process itself. However, in the case of antagonistic cancers and viral infection, immune activation is beneficial; thus, immunostimulatory small interfering RNAs were designed to create bifunctional small molecules with RNAi and immunostimulatory activities. This review summarizes the research studies of RNAi-associated immune stimulation and the approaches for manipulating immunostimulatory activities.
Collapse
Affiliation(s)
- Zhongji Meng
- Department of Infectious Diseases, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Mengji Lu
- Institute of Virology, University Hospital of Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
17
|
Zhang LX, Liu DQ, Wang SW, Yu XL, Ji M, Xie XX, Liu SY, Liu RT. MgAl-layered double hydroxide nanoparticles co-delivering siIDO and Trp2 peptide effectively reduce IDO expression and induce cytotoxic T-lymphocyte responses against melanoma tumor in mice. J Mater Chem B 2017; 5:6266-6276. [DOI: 10.1039/c7tb00819h] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The co-delivery of Trp2 and siIDO by LDH nanoparticles alleviates immune tolerance and promotes CTL responses in vivo.
Collapse
Affiliation(s)
- Ling-xiao Zhang
- State Key Laboratory of Biochemical Engineering
- Institute of Process Engineering
- Chinese Academy of Sciences
- Beijing
- China
| | - Dong-qun Liu
- State Key Laboratory of Biochemical Engineering
- Institute of Process Engineering
- Chinese Academy of Sciences
- Beijing
- China
| | - Shao-wei Wang
- State Key Laboratory of Biochemical Engineering
- Institute of Process Engineering
- Chinese Academy of Sciences
- Beijing
- China
| | - Xiao-lin Yu
- State Key Laboratory of Biochemical Engineering
- Institute of Process Engineering
- Chinese Academy of Sciences
- Beijing
- China
| | - Mei Ji
- State Key Laboratory of Biochemical Engineering
- Institute of Process Engineering
- Chinese Academy of Sciences
- Beijing
- China
| | - Xi-xiu Xie
- State Key Laboratory of Biochemical Engineering
- Institute of Process Engineering
- Chinese Academy of Sciences
- Beijing
- China
| | - Shu-ying Liu
- State Key Laboratory of Biochemical Engineering
- Institute of Process Engineering
- Chinese Academy of Sciences
- Beijing
- China
| | - Rui-tian Liu
- State Key Laboratory of Biochemical Engineering
- Institute of Process Engineering
- Chinese Academy of Sciences
- Beijing
- China
| |
Collapse
|
18
|
Prolongation of kidney allograft survival regulated by indoleamine 2, 3-dioxygenase in immature dendritic cells generated from recipient type bone marrow progenitors. Mol Immunol 2016; 79:22-31. [PMID: 27689750 DOI: 10.1016/j.molimm.2016.09.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 09/02/2016] [Accepted: 09/07/2016] [Indexed: 12/13/2022]
Abstract
Immature dendritic cells (iDCs) are bone marrow-derived professional antigen-presenting cells, exhibit very low levels of the co-stimulatory molecules CD80 (B7-1), CD86 (B7-2), and CD40 and major histocompatibility complex (MHC) class II and play a critical role in triggering antigen-specific immunotolerance. The enzyme indoleamine 2, 3-dioxygenase (IDO) is a cytosolic tryptophan catabolism rate-limiting step enzyme. IDO secreted by DCs shows an association with the suppression of T-cell responses and promotion of tolerance. In this study, BN rat recipients were pre-injected with donor renal alloantigen-treated recipient iDCs before kidney transplantation. The renal allograft exhibited a lighter renal rejection response, prolonged graft survival time, and an increasing content of CD4+CD25+Foxp3+ regulatory T cells (Tregs). Additionally, up-regulated secretion of Th2 cytokines were found in recipient sera post-transplantation. Transfection of si-IDO1 RNA into renal-antigen-treated recipient iDCs reversed these changes, which suggested that IDO channel signaling may be involved in iDC-induced allograft immunotolerance. These results suggested that iDC-induced and IDO-mediated allograft immunotolerance might be a potentially feasible tactic to prolong allograft survival, in addition to immunosuppressive drugs.
Collapse
|
19
|
Diversification of Antitumour Immunity in a Patient with Metastatic Melanoma Treated with Ipilimumab and an IDO-Silenced Dendritic Cell Vaccine. Case Rep Med 2016; 2016:9639585. [PMID: 27504122 PMCID: PMC4967686 DOI: 10.1155/2016/9639585] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 06/13/2016] [Indexed: 01/17/2023] Open
Abstract
Indoleamine 2,3-dioxygenase (IDO) expression in dendritic cells (DCs) inhibits T-cell activation and promotes T-cell differentiation into regulatory T-cells. Moreover, IDO expression promotes resistance to immunotherapies targeting immune checkpoints such as the cytotoxic T lymphocyte antigen-4 (CTLA-4). Here, a patient with metastatic melanoma pretreated with ipilimumab, an anti-CTLA-4 blocking antibody, was vaccinated with IDO-silenced DCs cotransfected with mRNA for survivin or hTERT tumour antigens. During vaccination, T-cell responses to survivin and hTERT tumour antigens were generated, and a certain degree of clinical benefit was achieved, with a significant reduction in lung, liver, and skin metastases, along with a better performance status. T-cell responses against MART-1 and NY-ESO-1 tumour antigens were also detected in the peripheral blood. The patient also mounted an antibody response to several melanoma proteins, indicating diversification of the antitumour immunity in this patient. The identification of such serum antibody-reacting proteins could facilitate the discovery of tumour neoantigens.
Collapse
|
20
|
Hanks BA. Immune evasion pathways and the design of dendritic cell-based cancer vaccines. DISCOVERY MEDICINE 2016; 21:135-142. [PMID: 27011049 PMCID: PMC4934601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Emerging data is suggesting that the process of dendritic cell (DC) tolerization is an important step in tumorigenesis. Our understanding of the networks within the tumor microenvironment that functionally tolerize DC function is evolving while methods for genetically manipulating DC populations in situ continue to develop. A more intimate understanding of the paracrine signaling pathways which mediate immune evasion by subverting DC function promises to provide novel strategies for improving the clinical efficacy of DC-based cancer vaccines. This will likely require a better understanding of both the antigen expression profile and the immune evasion network of the tumor and its associated stromal tissues.
Collapse
Affiliation(s)
- Brent A Hanks
- Department of Medicine Division of Medical Oncology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
21
|
Abstract
The therapeutic potential of dendritic cell (DC) cancer vaccines has gained momentum in recent years. However, clinical data indicate that antitumor immune responses generally fail to translate into measurable tumor regression. This has been ascribed to a variety of tolerance mechanisms, one of which is the expression of immunosuppressive factors by DCs and T cells. With respect to cancer immunotherapies, these factors antagonise the ability to induce robust and sustained immunity required for tumor cell eradication. Gene silencing of immunosuppressive factors in either DCs or adoptive transferred T cells enhanced anti-tumor immune responses and significantly inhibited tumor growth. Therefore, engineered next generation of DC vaccines or adoptive T-cell therapy should include immunomodulatory siRNAs to release the "brakes" imposed by the immune system. Moreover, the combination of gene silencing, antigen targeting to DCs and cytoplasmic cargo delivery will improve clinical benefits.
Collapse
Key Words
- AML, acute myeloid leukemia
- CMV, human cytomegalovirus
- CTLA4, T-lymphocyte-associated antigen 4
- DC, Dendritic cells
- Gal, galectin hTERT, human telomerase reverse transcriptase
- IDO, indoleamine 2,3-dioxygenase
- IL, interleukin
- INF, interferon
- NK, natural killer
- PD1, programmed cell death
- RNA interference
- RNAi, RNA interference
- SOCS1, suppressor of cytokine signaling
- STAT, Signal transducer and activator of transcription
- T-cell therapy
- TCR, T cell receptor
- TLR, toll like receptor
- Treg, Regulatory T
- cancer vaccine
- gene silencing
- immunotherapy
- siRNA, small interfering RNA
- targeted therapies
Collapse
Affiliation(s)
- Mouldy Sioud
- a Department of Immunology; Institute for Cancer Research ; Oslo University Hospital ; Montebello , Norway
| |
Collapse
|
22
|
Martínez T, Jiménez AI, Pañeda C. Short-interference RNAs: becoming medicines. EXCLI JOURNAL 2015; 14:714-46. [PMID: 26648823 PMCID: PMC4669907 DOI: 10.17179/excli2015-297] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 05/12/2015] [Indexed: 12/30/2022]
Abstract
RNA interference is a cellular mechanism by which small molecules of double stranded RNA modulate gene expression acting on the concentration and/or availability of a given messenger RNA. Almost 10 years after Fire and Mello received the Nobel Prize for the discovery of this mechanism in flat worms, RNA interference is on the edge of becoming a new class of therapeutics. With various phase III studies underway, the following years will determine whether RNAi-therapeutics can rise up to the challenge and become mainstream medicines. The present review gives a thorough overview of the current status of this technology focusing on the path to the clinic of this new class of compounds.
Collapse
Affiliation(s)
- Tamara Martínez
- Sylentis, R&D department c/Santiago Grisolía, Tres Cantos, Madrid, Spain
| | - Ana Isabel Jiménez
- Sylentis, R&D department c/Santiago Grisolía, Tres Cantos, Madrid, Spain
| | - Covadonga Pañeda
- Sylentis, R&D department c/Santiago Grisolía, Tres Cantos, Madrid, Spain
| |
Collapse
|
23
|
Versatile polyion complex micelles for peptide and siRNA vectorization to engineer tolerogenic dendritic cells. Eur J Pharm Biopharm 2015; 92:216-27. [DOI: 10.1016/j.ejpb.2015.03.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 03/06/2015] [Accepted: 03/10/2015] [Indexed: 11/20/2022]
|
24
|
Sioud M. Overcoming the challenges of siRNA activation of innate immunity: design better therapeutic siRNAs. Methods Mol Biol 2015; 1218:301-319. [PMID: 25319660 DOI: 10.1007/978-1-4939-1538-5_19] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
RNA interference (RNAi) is a conserved regulatory mechanism of posttranscriptional gene silencing triggered by either endogenously (e.g. microRNAs) or exogenously double-stranded RNA as small interfering (si) RNAs. To date, the use of siRNA (21-nt) has become a standard laboratory tool to silence gene expression in mammalian cells in-vitro and in-vivo. The methodology also holds promise for treating a diversity of human diseases. However, one of the challenges of making siRNAs as therapeutic drugs includes the activation of innate immunity and silencing of unwanted genes. Therefore, the use of siRNAs in functional genomics and human therapies depends on the development of strategies to overcome siRNA unwanted effects. This chapter highlights some efficient strategies aimed at separating gene silencing from immunostimulation and improving siRNA gene silencing specificity.
Collapse
MESH Headings
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/genetics
- Cancer Vaccines/immunology
- Dendritic Cells/immunology
- Dendritic Cells/transplantation
- Gene Expression Regulation, Neoplastic
- Genetic Engineering/methods
- Humans
- Immunity, Innate
- Immunotherapy/methods
- Neoplasms/genetics
- Neoplasms/immunology
- Neoplasms/therapy
- RNA, Double-Stranded
- RNA, Small Interfering/chemical synthesis
- RNA, Small Interfering/genetics
- RNA, Small Interfering/immunology
- Receptors, Retinoic Acid/genetics
- Receptors, Retinoic Acid/immunology
- Ribose/metabolism
- Toll-Like Receptor 7/genetics
- Toll-Like Receptor 7/immunology
- Toll-Like Receptor 8/genetics
- Toll-Like Receptor 8/immunology
- Toll-Like Receptors/genetics
- Toll-Like Receptors/immunology
- Transfection
Collapse
Affiliation(s)
- Mouldy Sioud
- Department of Immunology, Institute for Cancer Research, Oslo University Hospital, Montebello, Oslo, N-310, Norway,
| |
Collapse
|
25
|
Sioud M, Mobergslien A, Sæbøe-Larssen S. Immunosuppressive factor blockade in dendritic cells via siRNAs results in objective clinical responses. Methods Mol Biol 2015; 1218:269-76. [PMID: 25319657 DOI: 10.1007/978-1-4939-1538-5_16] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Over the past decade, immunotherapy has emerged as a promising new form of cancer treatment with the potential to eradicate tumor metastasis. However, its curative potential is in general limited by the existence of negative feedback mechanisms that control dendritic cells (DCs) and T-cell activation. For clinically effective immunity, there is a need of inhibiting the expression of these immune suppressors. This could enhance the activation of DCs, T cells, and natural killer cells, and might be beneficial for cancer immunotherapy. Among the immune inhibitory molecules expressed by DCs is indoleamine 2,3-dioxygenase (IDO), an enzyme that conveys immunosuppressive effects by degrading tryptophan, an essential amino acid required for T-cell proliferation and survival. Depletion of tryptophan by IDO-positive DCs induces T-cell apoptosis and the conversion of naïve CD4+ T cells into regulatory T cells that further suppress antitumor immunity. Herein, we describe a protocol for in vitro synthesis of small interfering RNA against IDO and other immunosuppressive factors such as interleukin-10 and programmed cell death-1 ligands in order to reverse immune suppression mediated by DCs. Vaccination with IDO-silenced DC vaccines enhanced immune responses and antitumor immunity in cancer patients.
Collapse
Affiliation(s)
- Mouldy Sioud
- Department of Immunology, Institute for Cancer Research, Oslo University Radium Hospital, Norway, 0310, Montebello, Oslo, Norway,
| | | | | |
Collapse
|
26
|
Ballarín-González B, Ebbesen MF, Howard KA. Polycation-based nanoparticles for RNAi-mediated cancer treatment. Cancer Lett 2014; 352:66-80. [DOI: 10.1016/j.canlet.2013.09.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 09/16/2013] [Accepted: 09/18/2013] [Indexed: 12/19/2022]
|
27
|
Shahzidi S, Brech A, Sioud M, Li X, Suo Z, Nesland JM, Peng Q. Lamin A/C cleavage by caspase-6 activation is crucial for apoptotic induction by photodynamic therapy with hexaminolevulinate in human B-cell lymphoma cells. Cancer Lett 2013; 339:25-32. [PMID: 23916608 DOI: 10.1016/j.canlet.2013.07.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 07/24/2013] [Indexed: 01/16/2023]
Abstract
Photodynamic therapy (PDT) with a light-activated drug is an approved modality for cancer treatment. Hexaminolevulinate (HAL), a hexylester of 5-aminolevulinic acid as the photosensitising protoporphyrin IX (PpIX) precursor, is clinically used for both PDT and photodetection. Our previous studies have shown that HAL-PDT can effectively induce apoptosis in several human blood malignant cell lines. However, the mechanisms involved in the apoptotic induction are still not fully elucidated. In this study we have focused on the role of cellular lamin A/C in the apoptotic induction. HAL-PDT-mediated apoptosis was confirmed by various techniques including fluorescence microscopy and electron microscopy in both human B-cell lymphoma Ramos and Daudi cell lines. The lamin A/C, together with caspases-6 and -3, was cleaved during the apoptosis. Western blots, immunocytochemistry, fluorescence microscopy and electron microscopy demonstrated that the specific caspase-6 inhibitor abrogated the HAL-PDT-mediated cleavages of both caspase-6 and lamin A/C and subsequent apoptosis in these two cell lines, suggesting that the cleavage of lamin A/C by the caspase-6 activation is crucial for such apoptotic induction.
Collapse
Affiliation(s)
- Susan Shahzidi
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital and Medical Faculty, University of Oslo, Oslo, Norway
| | | | | | | | | | | | | |
Collapse
|
28
|
Olesen MTJ, Ballarín-González B, Howard KA. The application of RNAi-based treatments for inflammatory bowel disease. Drug Deliv Transl Res 2013; 4:4-18. [DOI: 10.1007/s13346-013-0156-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
29
|
Sioud M, Skorstad G, Mobergslien A, Sæb⊘e‐Larssen S. A novel peptide carrier for efficient targeting of antigens and nucleic acids to dendritic cells. FASEB J 2013; 27:3272-83. [DOI: 10.1096/fj.12-224758] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Mouldy Sioud
- Department of ImmunologyOslo University Radium HospitalOsloNorway
| | | | - Anne Mobergslien
- Department of ImmunologyOslo University Radium HospitalOsloNorway
| | | |
Collapse
|
30
|
Wang J, Liao L, Tan J. Dendritic cell-based vaccination for renal cell carcinoma: challenges in clinical trials. Immunotherapy 2013; 4:1031-42. [PMID: 23148755 DOI: 10.2217/imt.12.107] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
After decades of research, dendritic cell (DC)-based vaccines for renal cell carcinoma have progressed from preclinical rodent models and safety assessments to Phase I/II clinical trials. DC vaccines represent a promising therapy that has produced measurable immunological responses and prolonged survival rates. However, there is still much room to improve in terms of therapeutic efficacy. The key issues that affect the efficiency and reliability of DC therapy include the selection of patients who will respond best to treatment, the proper preparation and administration of DC vaccines, and a combination of DC vaccination with other immune-enhancing therapies (e.g., removal of Tregs, CTLA-4 blockade and lymphodepletion). Additional antiangiogenic agents will hopefully lead to greater survival benefits for patients in early disease stages. This review focuses on the different approaches of DC-based vaccination against renal cell carcinoma and potential strategies to enhance the efficacy of DC vaccination.
Collapse
Affiliation(s)
- Jin Wang
- Organ Transplant Institute, Fuzhou General Hospital, Xiamen University, Fuzhou, China
| | | | | |
Collapse
|
31
|
Sioud M, Saebøe-Larssen S, Hetland TE, Kaern J, Mobergslien A, Kvalheim G. Silencing of indoleamine 2,3-dioxygenase enhances dendritic cell immunogenicity and antitumour immunity in cancer patients. Int J Oncol 2013; 43:280-8. [PMID: 23620105 DOI: 10.3892/ijo.2013.1922] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Accepted: 02/20/2013] [Indexed: 11/05/2022] Open
Abstract
Dendritic cells (DCs) are being explored as a therapeutic vaccine for cancers. However, their immunogenic potential is limited by the presence of immunosuppressive factors. Among these factors is the tryptophan-degrading enzyme indoleamine 2,3-dioxygenase (IDO). In this study, we have investigated the safety, immunogenicity and clinical response of IDO-silenced DC vaccine in four patients with gynecological cancers. DCs were transfected with IDO small interfering RNA and mRNA encoding human telomerase reverse transcriptase (hTERT) or survivin, two universal tumour antigens. Silencing of IDO in DCs did not affect the expression of the co-stimulatory molecules CD80 and CD86, but enhanced the expression of the CCR7 and CD40 molecules. IDO-silenced DCs showed superior potency to activate allogeneic T cells compared to their IDO-positive counterparts. The immunisation with this novel DC cancer vaccine was well tolerated and all patients developed delayed-type hypersensitivity skin reaction and specific T-cell response against hTERT and survivin tumour antigens. Perhaps most importantly, the immune response seen in the patients was related to objective clinical response. Thus, IDO silencing can enhance the immunogenic function of DCs in vitro and in vivo. Overall, the data provide proof-of-principle that immunisation with IDO-silenced DC vaccine is safe and effective in inducing antitumour immunity.
Collapse
Affiliation(s)
- Mouldy Sioud
- Department of Immunology, Oslo University Radium Hospital, Montebello, N-0310 Oslo, Norway.
| | | | | | | | | | | |
Collapse
|
32
|
Harden JL, Egilmez NK. Indoleamine 2,3-dioxygenase and dendritic cell tolerogenicity. Immunol Invest 2013; 41:738-64. [PMID: 23017144 DOI: 10.3109/08820139.2012.676122] [Citation(s) in RCA: 135] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
This article summarizes the molecular and cellular mechanisms that regulate the activity of indoleamine 2,3-dioxygenase (IDO), a potent immune-suppressive enzyme, in dendritic cells (DCs). Specific attention is given to differential up-regulation of IDO in distinct DC subsets, its function in immune homeostasis/autoimmunity, infection and cancer; and the associated immunological outcomes. The review will conclude with a discussion of the poorly defined mechanisms that mediate the long-term maintenance of IDO-expression in response to inflammatory stimuli and how selective modulation of IDO activity may be used in the treatment of disease.
Collapse
Affiliation(s)
- Jamie L Harden
- The State University of New York at Buffalo, Buffalo, New York 14214, USA.
| | | |
Collapse
|
33
|
Abstract
Synthetic small interfering RNAs (siRNAs) have revolutionized functional genomics in mammalian cell cultures due to their reliability, efficiency, and ease of use. This success, however, has not fully translated into siRNA applications in vivo and in siRNA therapeutics where initial optimism has been dampened by a lack of efficient delivery strategies and reports of siRNA off-target effects and immunogenicity. Encouragingly, most aspects of siRNA behavior can be addressed by careful engineering of siRNAs incorporating beneficial chemical modifications into discrete nucleotide positions during siRNA synthesis. Here, we review the literature (Subheadings 1 -3) and provide a quick guide (Subheading 4) to how the performance of siRNA can be improved by chemical modification to suit specific applications in vitro and in vivo.
Collapse
Affiliation(s)
- Jesper B Bramsen
- Department of Molecular Biology and Genetics, Interdisciplinary Nanoscience Center (iNANO), University of Aarhus, Aarhus, Denmark.
| | | |
Collapse
|
34
|
Polycation-based nanoparticle delivery of RNAi therapeutics: adverse effects and solutions. Adv Drug Deliv Rev 2012; 64:1717-29. [PMID: 22800620 DOI: 10.1016/j.addr.2012.07.004] [Citation(s) in RCA: 123] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Revised: 06/27/2012] [Accepted: 07/06/2012] [Indexed: 12/19/2022]
Abstract
Small interfering RNA (siRNA) that silence genes by the process of RNA interference offers a new therapeutic modality for disease treatment. Polycation-based nanoparticles termed polyplexes have been developed to maximise extracellular and intracellular siRNA delivery, a key requirement for enabling the clinical translation of RNAi-based drugs. Medical applications are dependent on safety; therefore, detailed investigation into potential toxicity to the cell or organism is required. This review addresses potential adverse effects arising from cellular and tissue interactions, immune stimulation and altered gene expression that can be associated with the assembled polyplex or the polycation and siRNA component parts. A greater understanding of the cellular mechanisms involved allows design-based solutions for rationale development of safe, effective and clinically relevant polyplex-based RNAi drugs.
Collapse
|
35
|
Sioud M, Mobergslien A. Selective killing of cancer cells by peptide-targeted delivery of an anti-microbial peptide. Biochem Pharmacol 2012; 84:1123-32. [PMID: 22922046 DOI: 10.1016/j.bcp.2012.08.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2012] [Revised: 08/02/2012] [Accepted: 08/03/2012] [Indexed: 10/28/2022]
Abstract
Antimicrobial peptides selectively kill bacteria while maintaining low mammalian cell cytotoxicity. However, they become cytotoxic subsequent to internalization. Here we have conjugated the lytic peptide (KLAKLAK)(2) to either a cancer-cell binding peptide (LTVSPWY) selected from peptide libraries or to a gastrin-releasing peptide (GNHWAVGHLM) in order to direct the lytic peptide to cancer cells. Peptide cytotoxicity was tested in breast MCF-7 and MDA-MB-231 cancer cells. The fusion peptides were internalized by cancer cells, disintegrated the cell membrane and induced rapid killing of the cells with IC50 values as low as 4-7 μM. Peptide cytotoxicity was dependent on the targeting receptor. Indeed, addition of free targeting peptide reduced cell killing. Blood lymphocytes and normal human mammary epithelial cells were less sensitive to the fusion peptides. Although most of the cells were killed by necrosis, fusion peptides branched with DNA oligonucleotides induced apoptosis as assayed by annexin V staining and activation of caspase 3. Therefore, the new designed drug peptides might provide a potent and selective anticancer therapy.
Collapse
Affiliation(s)
- Mouldy Sioud
- Department of Immunology, Molecular Medicine Group, the Norwegian Radium Hospital, Institute for Cancer Research Montebello, N-0310, Oslo, Norway.
| | | |
Collapse
|
36
|
Zheng X, Koropatnick J, Chen D, Velenosi T, Ling H, Zhang X, Jiang N, Navarro B, Ichim TE, Urquhart B, Min W. Silencing IDO in dendritic cells: a novel approach to enhance cancer immunotherapy in a murine breast cancer model. Int J Cancer 2012; 132:967-77. [PMID: 22870862 DOI: 10.1002/ijc.27710] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Accepted: 06/14/2012] [Indexed: 12/15/2022]
Abstract
Cancer immunotherapeutic agents (vaccines) in the form of antigen-loaded dendritic cells (DCs) reached an important milestone with the recent approval of Provenge, the first DC vaccine for treatment of prostate cancer. Although this heralds a new era of tumor immunotherapy, it also highlights the compelling need to optimize such DC-based therapies as they are increasingly tested and used to treat human patients. In this study we sought to augment and enhance the antitumor activity of a DC-based vaccine using siRNA to silence expression of immunosuppressive enzyme indoleamine 2,3-dioxygenase (IDO) in DCs. We report here that DCs loaded with tumor antigens, but with siRNA-silenced IDO expression, were introduced into 4T1 breast tumor-bearing mice, the treatment: (i) lengthened the time required for tumor onset, (ii) decreased tumor size compared to tumors grown for equal lengths of time in mice treated with antigen-loaded DCs without IDO silencing and (iii) reduced CD4(+) and CD8(+) T cell apoptosis. Furthermore, immunization with IDO-silenced DCs enhanced tumor antigen-specific T cell proliferation and CTL activity, and decreased numbers of CD4(+) CD25(+) Foxp3(+) T(reg). This study provides evidence to support silencing of immunosuppressive genes (IDO) as an effective strategy to enhance the efficacy of DC-based cancer immunotherapeutic.
Collapse
Affiliation(s)
- Xiufen Zheng
- Department of Surgery, University of Western Ontario, London, Ontario, Canada.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Li L, Huang L, Lemos HP, Mautino M, Mellor AL. Altered tryptophan metabolism as a paradigm for good and bad aspects of immune privilege in chronic inflammatory diseases. Front Immunol 2012; 3:109. [PMID: 22593757 PMCID: PMC3350084 DOI: 10.3389/fimmu.2012.00109] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Accepted: 04/17/2012] [Indexed: 01/01/2023] Open
Abstract
The term "immune privilege" was coined to describe weak immunogenicity (hypo-immunity) that manifests in some transplant settings. We extended this concept to encompass hypo-immunity that manifests at local sites of inflammation relevant to clinical diseases. Here, we focus on emerging evidence that enhanced tryptophan catabolism is a key metabolic process that promotes and sustains induced immune privilege, and discuss the implications for exploiting this knowledge to improve treatments for hypo-immune and hyper-immune syndromes using strategies to manipulate tryptophan metabolism.
Collapse
Affiliation(s)
- Lingqian Li
- Immunotherapy Center, Georgia Health Sciences University Augusta, GA, USA
| | | | | | | | | |
Collapse
|
38
|
Sioud M, Mobergslien A. Efficient siRNA Targeted Delivery into Cancer Cells by Gastrin-Releasing Peptides. Bioconjug Chem 2012; 23:1040-9. [DOI: 10.1021/bc300050j] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Mouldy Sioud
- Department of Immunology, Molecular
Medicine Group, Institute for Cancer Research, Oslo University Hospital, Montebello, N-0310, Oslo, Norway
| | - Anne Mobergslien
- Department of Immunology, Molecular
Medicine Group, Institute for Cancer Research, Oslo University Hospital, Montebello, N-0310, Oslo, Norway
| |
Collapse
|
39
|
Mobergslien A, Sioud M. Galectin-1 and -3 gene silencing in immature and mature dendritic cells enhances T cell activation and interferon-γ production. J Leukoc Biol 2011; 91:461-7. [PMID: 22167721 DOI: 10.1189/jlb.0711361] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
DCs are specialized APCs capable of inducing T cell activation as well as promoting tolerance. Although Gal, a family of β-galactoside-binding proteins, were found to affect immunity, little is known about the contribution of DC-expressed Gal on T cell activation. Here, we show that human imDCs and mDCs constitutively express Gal-1, Gal-3, Gal-8, and Gal-9 at mRNA and protein levels. Two of the most abundant Gal-Gal-1 and Gal-3-were highly expressed and detected on the cell surface of DCs. In contrast to Gal-8, knockdown of Gal-1 or Gal-3 in DCs enhanced allogeneic T cell responses. This was observed with imDCs and mDCs, but the effects were more pronounced with imDCs. Furthermore, allogeneic CD4(+) T cells incubated with Gal-1 or Gal-3 knockdown DCs produced more IFN-γ and less IL-10 than did control cells. The percentage of apoptotic T cells was significantly higher in cultures with control DCs than that with Gal-1 or Gal-3 knockdown DCs. Collectively, the data indicate that DC-expressed Gal-1 and Gal-3 are regulatory molecules that favor the inhibition of T cell activation. Furthermore, the data provide a new mechanism for the poor capacity of imDCs to stimulate T cells.
Collapse
Affiliation(s)
- Anne Mobergslien
- Department of Immunology, Institute for Cancer Research, Radiumhospitalet-Rikshospitalet University Hospital, Montebello, Oslo, Norway
| | | |
Collapse
|
40
|
Ezzelarab M, Thomson AW. Tolerogenic dendritic cells and their role in transplantation. Semin Immunol 2011; 23:252-63. [PMID: 21741270 DOI: 10.1016/j.smim.2011.06.007] [Citation(s) in RCA: 119] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 06/10/2011] [Indexed: 01/09/2023]
Abstract
The pursuit of clinical transplant tolerance has led to enhanced understanding of mechanisms underlying immune regulation, including the characterization of immune regulatory cells, in particular antigen-presenting cells (APC) and regulatory T cells (Treg), that may play key roles in promoting operational tolerance. Dendritic cells (DC) are highly efficient APC that have been studied extensively in rodents and humans, and more recently in non-human primates. Owing to their ability to regulate both innate and adaptive immune responses, DC are considered to play crucial roles in directing the alloimmune response towards transplant tolerance or rejection. Mechanisms via which they can promote central and peripheral tolerance include clonal deletion, the induction of Treg, and inhibition of memory T cell responses. These properties have led to the use of tolerogenic DC as a therapeutic strategy to promote organ transplant tolerance. In rodents, infusion of donor- or recipient-derived tolerogenic DC can extensively prolong donor-specific allograft survival, in association with regulation of the host T cell response. In clinical transplantation, progress has been made in monitoring DC in relation to graft outcome, including studies in operational liver transplant tolerance. Although clinical trials involving immunotherapeutic DC for patients with cancer are ongoing, implementation of human DC therapy in clinical transplantation will require assessment of various critical issues. These include cell isolation and purification techniques, source, route and timing of administration, and combination immunosuppressive therapy. With ongoing non-human primate studies focused on DC therapy, these logistics can be investigated seeking the optimal approaches. The scientific rationale for implementation of tolerogenic DC therapy to promote clinical transplant tolerance is strong. Evaluation of technical and therapeutic logistic issues is an important next step prior to the application of tolerogenic DC in clinical organ transplantation.
Collapse
Affiliation(s)
- Mohamed Ezzelarab
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, 200 Lothrop Street, BST W1540, Pittsburgh, PA 15261, USA
| | | |
Collapse
|
41
|
Torabi-Rahvar M, Bozorgmehr M, Jeddi-Tehrani M, Zarnani AH. Potentiation strategies of dendritic cell-based antitumor vaccines: combinational therapy takes the front seat. Drug Discov Today 2011; 16:733-40. [PMID: 21558016 DOI: 10.1016/j.drudis.2011.04.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2010] [Revised: 03/01/2011] [Accepted: 04/27/2011] [Indexed: 12/12/2022]
Abstract
Despite recent attempts to take advantage of dendritic cell (DC)-based vaccines for cancer immunotherapy, the results of clinical studies have been disappointing. This is mainly as a result of the diverse immune escape mechanisms used by the tumor together with the insufficient ability of DCs to mount an effective immune response against these mechanisms. In this regard, several approaches have been devised to improve the efficacy of DC-based vaccines. However, the application of each individual approach per se might not be sufficient to overwhelm the diverse immune escape mechanisms. In this review, we focus on current strategies for the ex vivo potentiation of DC-based vaccines, with an emphasis on combinational therapy methods as a promising alternative for tumor immunotherapy.
Collapse
Affiliation(s)
- Monireh Torabi-Rahvar
- Department of Immunology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | |
Collapse
|
42
|
Bowles R, Patil S, Pincas H, Sealfon SC. Validation of efficient high-throughput plasmid and siRNA transfection of human monocyte-derived dendritic cells without cell maturation. J Immunol Methods 2010; 363:21-8. [PMID: 20875421 PMCID: PMC3964480 DOI: 10.1016/j.jim.2010.09.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Accepted: 09/17/2010] [Indexed: 12/25/2022]
Abstract
Transfection of primary immune cells is difficult to achieve at high efficiency and without cell activation and maturation. Dendritic cells (DCs) represent a key link between the innate and adaptive immune systems. Delineating the signaling pathways involved in the activation of human primary DCs and reverse engineering cellular inflammatory pathways have been challenging tasks. We optimized and validated an effective high-throughput transfection protocol, allowing us to transiently express DNA in naïve primary DCs, as well as investigate the effect of gene silencing by RNA interference. Using a high-throughput nucleofection system, monocyte-derived DCs were nucleoporated with a plasmid expressing green fluorescent protein (GFP), and transfection efficiency was determined by flow cytometry, based on GFP expression. To evaluate the effect of nucleoporation on DC maturation, the expression of cell surface markers CD86 and MHCII in GFP-positive cells was analyzed by flow cytometry. We established optimal assay conditions with a cell viability reaching 70%, a transfection efficiency of over 50%, and unchanged CD86 and MHCII expression. We examined the impact of small interfering RNA (siRNA)-mediated knockdown of RIG-I, a key viral recognition receptor, on the induction of the interferon (IFN) response in DCs infected with Newcastle disease virus. RIG-I protein was undetectable by Western blot in siRNA-treated cells. RIG-I knockdown caused a 75% reduction in the induction of IFNβ mRNA compared with the negative control siRNA. This protocol should be a valuable tool for probing the immune response pathways activated in human DCs.
Collapse
Affiliation(s)
| | | | - Hanna Pincas
- Center for Translational Systems Biology and Department of Neurology, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Stuart C. Sealfon
- Center for Translational Systems Biology and Department of Neurology, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029, USA
| |
Collapse
|
43
|
Sioud M, Mobergslien A, Boudabous A, Fløisand Y. Evidence for the involvement of galectin-3 in mesenchymal stem cell suppression of allogeneic T-cell proliferation. Scand J Immunol 2010; 71:267-74. [PMID: 20384870 DOI: 10.1111/j.1365-3083.2010.02378.x] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Human bone marrow-derived mesenchymal stem cells (MSC) are multipotent non-hematopoietic progenitors that have regulatory activity on immune cells. NOD- and Toll-like receptors (NLR, TLR) have several roles in immunity, including those relevant to pathogen recognition and shaping the course of immune responses by controlling gene expression. We have shown that these innate immune receptors are expressed by hematopoietic CD34+ progenitors and MSC. To uncover genes critical in MSC function, first we have used microarray to screen for potential transcripts whose levels are altered in response to NOD-1 and TLR-2 activation, and second we validated some candidate genes using real-time RT-PCR, Western blots and cellular assays. Amongst the altered genes, galectin-3 was upregulated at both mRNA and protein levels in response to TLR-2 activation. Interestingly, MSC secreted galectin-3, a protein known to modulate T-cell proliferation, gene expression, cell adhesion and migration. Knockdown of galectin-3 in MSC using small interfering RNA (siRNA) reduced the immunosuppressive effect of MSC on mixed lymphocyte cultures when compared to cells treated with an irrelevant siRNA (P < 0.05). Collectively, the data emphasize a new role of galectin-3 in the immunomodulatory function of MSC and indicate that NOD signalling pathway is also functional in these cells.
Collapse
Affiliation(s)
- M Sioud
- Department of Immunology, The Institute for Cancer Research, Rikshospitalet-Radiumhospitalet Medical Center, Oslo, Norway.
| | | | | | | |
Collapse
|
44
|
Abstract
Since its discovery in the late 1990s by Fire and Mello, RNA interference (RNAi) has proven a useful tool for scientists working in the fields of functional genomics, biotechnology, and therapeutic development. However, one of the obstacles of making small interfering RNAs (siRNAs), the main effector of RNAi, a therapeutic agent includes the activation of the immune system, off-target effects, and competition with endogenous microRNAs (miRNAs) for cellular miRNA-processing machinery. Therefore, the translation of RNAi technology into the clinic depends on the development of new strategies to surmount siRNA unwanted effects and identify siRNA sensing receptors as well as to understand the extend of the competition between exogenous and endogenous miRNAs. This minireview summarizes our current knowledge of siRNA sensing by the immune receptors and how to separate siRNA unwanted effects from gene silencing.
Collapse
Affiliation(s)
- Mouldy Sioud
- Institute for Cancer Research, Department of Immunology, Molecular Medicine Group, Montebello, Oslo, Norway.
| |
Collapse
|
45
|
Mobergslien A, Sioud M. Optimized protocols for siRNA delivery into monocytes and dendritic cells. Methods Mol Biol 2010; 629:71-85. [PMID: 20387143 DOI: 10.1007/978-1-60761-657-3_5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Over the past decade, immunotherapy has emerged as a promising alternative form of cancer treatment with the potential to eradicate tumour metastasis. Unfortunately, its curative potential is in general limited by immunosuppressive proteins that negatively regulate dendritic (DC) and/or T-cell function. The recent discovery of RNA interference (RNAi) has facilitated the study of gene function in immune cells and recent data indicate that DC maturation, function, and survival can be modulated by small interfering RNAs (siRNAs) targeting genes involved in immune suppression. This chapter describes detailed protocols for introducing siRNAs into human monocytes and dendritic cells using standard electroporation techniques.
Collapse
Affiliation(s)
- Anne Mobergslien
- Departments of Immunology, Institute for Cancer Research, Radiumhospitalet-Rikshopitalet University Hospital, Oslo, Norway
| | | |
Collapse
|
46
|
Sioud M. Advances in RNA sensing by the immune system: separation of siRNA unwanted effects from RNA interference. Methods Mol Biol 2010; 629:33-52. [PMID: 20387141 DOI: 10.1007/978-1-60761-657-3_3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Small interfering RNAs (siRNAs) are routinely used as a genetic tool and hold promise for a range of therapeutic applications. However, one of the hurdles of making these agents a real therapeutic modality includes the activation of innate immunity and off-target effects. Therefore, the use of siRNAs in functional genomics and therapies depends on the development of new strategies to overcome these unwanted effects. It appears that the major innate immune response to chemically synthesized siRNAs is mediated by TLR7 and/or TLR8 in immune cells. Importantly, it has also been shown that the replacement of uridines with their 2'-modified counterparts can prevent immune activation. Similarly, 2'-modifications, particularly at the seed sequence reduced the number of unwanted off-target genes without interfering with siRNA silencing potency of the anticipated target gene. This chapter describes how to separate gene silencing from immunostimulation. Also, it discusses the impact of these findings on the design of effective cancer vaccines.
Collapse
Affiliation(s)
- Mouldy Sioud
- Department of Immunology, Institute for Cancer Research, Radiumhospitalet-Rikshopitalet University Hospital, Oslo, Norway
| |
Collapse
|
47
|
Sioud M. Does our current understanding of immune tolerance, autoimmunity, and immunosuppressive mechanisms facilitate the design of efficient cancer vaccines? Scand J Immunol 2009; 70:516-25. [PMID: 19906192 DOI: 10.1111/j.1365-3083.2009.02326.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The therapeutic use of the immune system to attack cancer cells has been a longstanding vision among tumour immunologists. However, most human tumours are poorly immunogenic and are able to invade the host immune system. Although these obstacles are clearly critical to cancer vaccine development, the induction of a strong anti-tumour immune response may rely on the activation of high affinity T cells through a molecular mimicry mechanism which involves cross-reactive recognition of foreign antigens mimicking the structure of tumour proteins. Taking into account the disparity in HLA molecules needed to present shared antigens; in late 1990s Stauss et al. described the possibility of generating allorestricted high affinity cytotoxic T cells against synthetic self-peptides bound to non-self-MHC molecules. In addition to the strategies indicated above, the inhibition of the immunosuppressive mechanisms associated with tumour invasion of the immune system using RNA interference also offers a new approach to vaccine design. This review highlights the problem of immune tolerance, the induction of autoreactive T cells, and describes strategies to enhance tumour immunity.
Collapse
Affiliation(s)
- M Sioud
- Department of Immunology, The Norwegian Radium Hospital, Institute for Cancer Research, Oslo, Norway.
| |
Collapse
|