1
|
Rahman T, Das A, Abir MH, Nafiz IH, Mahmud AR, Sarker MR, Emran TB, Hassan MM. Cytokines and their role as immunotherapeutics and vaccine Adjuvants: The emerging concepts. Cytokine 2023; 169:156268. [PMID: 37320965 DOI: 10.1016/j.cyto.2023.156268] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/02/2023] [Accepted: 06/06/2023] [Indexed: 06/17/2023]
Abstract
Cytokines are a protein family comprising interleukins, lymphokines, chemokines, monokines and interferons. They are significant constituents of the immune system, and they act in accordance with specific cytokine inhibiting compounds and receptors for the regulation of immune responses. Cytokine studies have resulted in the establishment of newer therapies which are being utilized for the treatment of several malignant diseases. The advancement of these therapies has occurred from two distinct strategies. The first strategy involves administrating the recombinant and purified cytokines, and the second strategy involves administrating the therapeutics which inhibits harmful effects of endogenous and overexpressed cytokines. Colony stimulating factors and interferons are two exemplary therapeutics of cytokines. An important effect of cytokine receptor antagonist is that they can serve as anti-inflammatory agents by altering the treatments of inflammation disorder, therefore inhibiting the effects of tumour necrosis factor. In this article, we have highlighted the research behind the establishment of cytokines as therapeutics and vaccine adjuvants, their role of immunotolerance, and their limitations.
Collapse
Affiliation(s)
- Tanjilur Rahman
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chattogram 4331, Bangladesh
| | - Ayan Das
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chattogram 4331, Bangladesh
| | - Mehedy Hasan Abir
- Faculty of Food Science and Technology, Chattogram Veterinary and Animal Sciences University, Chattogram 4225, Bangladesh
| | - Iqbal Hossain Nafiz
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chattogram 4331, Bangladesh
| | - Aar Rafi Mahmud
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail 1902, Bangladesh
| | - Md Rifat Sarker
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail 1902, Bangladesh
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chattogram 4381, Bangladesh; Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
| | - Mohammad Mahmudul Hassan
- Department of Physiology, Biochemistry and Pharmacology, Faculty of Veterinary Medicine, Chattogram Veterinary and Animal Sciences University, Chattogram 4225, Bangladesh; Queensland Alliance for One Health Sciences, School of Veterinary Science, The University of Queensland, Queensland 4343, Australia.
| |
Collapse
|
2
|
Abdelbaky SB, Ibrahim MT, Samy H, Mohamed M, Mohamed H, Mustafa M, Abdelaziz MM, Forrest ML, Khalil IA. Cancer immunotherapy from biology to nanomedicine. J Control Release 2021; 336:410-432. [PMID: 34171445 DOI: 10.1016/j.jconrel.2021.06.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/16/2021] [Accepted: 06/19/2021] [Indexed: 02/06/2023]
Abstract
With the significant drawbacks of conventional cancer chemotherapeutics, cancer immunotherapy has demonstrated the ability to eradicate cancer cells and circumvent multidrug resistance (MDR) with fewer side effects than traditional cytotoxic therapies. Various immunotherapeutic agents have been investigated for that purpose including checkpoint inhibitors, cytokines, monoclonal antibodies and cancer vaccines. All these agents aid immune cells to recognize and engage tumor cells by acting on tumor-specific pathways, antigens or cellular targets. However, immunotherapeutics are still associated with some concerns such as off-target side effects and poor pharmacokinetics. Nanomedicine may resolve some limitations of current immunotherapeutics such as localizing delivery, controlling release and enhancing the pharmacokinetic profile. Herein, we discuss recent advances of immunotherapeutic agents with respect to their development and biological mechanisms of action, along with the advantages that nanomedicine strategies lend to immunotherapeutics by possibly improving therapeutic outcomes and minimizing side effects.
Collapse
Affiliation(s)
- Salma B Abdelbaky
- University of Science and Technology, Zewail City, 6th of October City, Giza 12578, Egypt; Molecular, Cellular, and Developmental Biology, College of Arts and Sciences, The Ohio State University, Columbus, OH 43210, United States of America
| | - Mayar Tarek Ibrahim
- University of Science and Technology, Zewail City, 6th of October City, Giza 12578, Egypt; Department of Chemistry, Center for Scientific Computation, Center for Drug Discovery, Design, and Delivery (CD4), Southern Methodist University, Dallas, Texas 75275, United States of America
| | - Hebatallah Samy
- University of Science and Technology, Zewail City, 6th of October City, Giza 12578, Egypt; Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Québec H3C 3J7, Canada
| | - Menatalla Mohamed
- University of Science and Technology, Zewail City, 6th of October City, Giza 12578, Egypt; Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario L8S 4M1, Canada
| | - Hebatallah Mohamed
- University of Science and Technology, Zewail City, 6th of October City, Giza 12578, Egypt; Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario L8S 4M1, Canada
| | - Mahmoud Mustafa
- University of Science and Technology, Zewail City, 6th of October City, Giza 12578, Egypt
| | - Moustafa M Abdelaziz
- Department of Bioengineering, School of Engineering, The University of Kansas, Lawrence, KS 66045, USA
| | - M Laird Forrest
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, Lawrence, KS 66047, USA.
| | - Islam A Khalil
- Department of Pharmaceutics, College of Pharmacy and Drug Manufacturing, Misr University of Science and Technology (MUST), 6th of October, Giza 12582, Egypt.
| |
Collapse
|
3
|
Dobrovolskaia MA, Afonin KA. Use of human peripheral blood mononuclear cells to define immunological properties of nucleic acid nanoparticles. Nat Protoc 2020; 15:3678-3698. [PMID: 33097923 PMCID: PMC7875514 DOI: 10.1038/s41596-020-0393-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 07/31/2020] [Indexed: 12/21/2022]
Abstract
This protocol assesses proinflammatory properties of nucleic acid nanoparticles (NANPs) using a validated preclinical model, peripheral blood mononuclear cells (PBMCs), that is highly predictive of cytokine responses. The experimental procedure details the preparation of pyrogen-free NANPs, isolation of PBMCs from freshly collected human blood, and analysis of characteristic biomarkers (type I and III interferons) produced by PBMCs transfected with NANPs. Although representative NANPs with high and low immunostimulatory potential are used as standards throughout the procedure, this protocol can be adapted to any NANPs or therapeutic nucleic acids, irrespective of whether they are carrier based or carrier free; additional cytokine biomarkers can also be included. We test several commercial platforms and controls broadly accessible to the research community to quantify all biomarkers in either single- or multiplex format. The continuous execution of this protocol takes <48 h; when immediate analysis is not feasible, single-use aliquots of the supernatants can be frozen and stored (-20 °C; 12 months).
Collapse
Affiliation(s)
- Marina A Dobrovolskaia
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, MD, USA.
| | - Kirill A Afonin
- Nanoscale Science Program, Department of Chemistry, University of North Carolina at Charlotte, Charlotte, NC, USA.
| |
Collapse
|
4
|
Afonin KA, Dobrovolskaia MA, Church G, Bathe M. Opportunities, Barriers, and a Strategy for Overcoming Translational Challenges to Therapeutic Nucleic Acid Nanotechnology. ACS NANO 2020; 14:9221-9227. [PMID: 32706238 PMCID: PMC7731581 DOI: 10.1021/acsnano.0c04753] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Recent clinical successes using therapeutic nucleic acids (TNAs) have accelerated the transition of nucleic acid nanotechnology toward therapeutic applications. Significant progress in the development, production, and characterization of nucleic acid nanomaterials and nucleic acid nanoparticles (NANPs), as well as abundant proof-of-concept data, are paving the way toward biomedical applications of these materials. This recent progress has catalyzed the development of new strategies for biosensing, imaging, drug delivery, and immunotherapies with previously unrecognized opportunities and identified some barriers that may impede the broader clinical translation of NANP technologies. A recent workshop sponsored by the Kavli Foundation and the Materials Research Society discussed the future directions and current challenges for the development of therapeutic nucleic acid nanotechnology. Herein, we communicate discussions on the opportunities, barriers, and strategies for realizing the clinical grand challenge of TNA nanotechnology, with a focus on ways to overcome barriers to advance NANPs to the clinic.
Collapse
Affiliation(s)
- Kirill A Afonin
- Nanoscale Science Program, Department of Chemistry, The University of North Carolina at Charlotte, Charlotte, North Carolina 28223, United States
| | - Marina A Dobrovolskaia
- Nanotechnology Characterization Lab, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, Maryland 21702, United States
| | - George Church
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, United States
- Wyss Institute for Biologically Inspired Engineering, Boston, Massachusetts 02115, United States
- Harvard Graduate Program in Biological and Biomedical Sciences, Boston, Massachusetts 02115, United States
| | - Mark Bathe
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
5
|
Genetically engineered mesenchymal stem cells: targeted delivery of immunomodulatory agents for tumor eradication. Cancer Gene Ther 2020; 27:854-868. [PMID: 32418986 DOI: 10.1038/s41417-020-0179-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/28/2020] [Accepted: 05/05/2020] [Indexed: 12/17/2022]
Abstract
Cancer immunotherapy emerged as a novel therapeutic option that employs enhanced or amended native immune system to create a robust response against malignant cells. The systemic therapies with immune-stimulating cytokines have resulted in substantial dose-limiting toxicities. Targeted cytokine immunotherapy is being explored to overcome the heterogeneity of malignant cells and tumor cell defense with a remarkable reduction of systemic side effects. Cell-based strategies, such as dendritic cells (DCs), fibroblasts or mesenchymal stem cells (MSCs) seek to minimize the numerous toxic side effects of systemic administration of cytokines for extended periods of time. The usual toxicities comprised of a vascular leak, hypotension, and respiratory insufficiency. Natural and strong tropism of MSCs toward malignant cells made them an ideal systemic delivery vehicle to direct the proposed therapeutic genes to the vicinity of a tumor where their expression could evoke an immune reaction against the tumor. Compared with other methods, the delivery of cytokines via engineered MSCs is safer and renders a more practical, and promising strategy. Large numbers of genes code for cytokines have been utilized to reengineer MSCs as therapeutic cells. This review highlights the recent findings on the cytokine gene therapy for human malignancies by focusing on MSCs application in cancer immunotherapy.
Collapse
|
6
|
Dobrovolskaia MA. Nucleic Acid Nanoparticles at a Crossroads of Vaccines and Immunotherapies. Molecules 2019; 24:molecules24244620. [PMID: 31861154 PMCID: PMC6943637 DOI: 10.3390/molecules24244620] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 12/13/2019] [Accepted: 12/13/2019] [Indexed: 02/06/2023] Open
Abstract
Vaccines and immunotherapies involve a variety of technologies and act through different mechanisms to achieve a common goal, which is to optimize the immune response against an antigen. The antigen could be a molecule expressed on a pathogen (e.g., a disease-causing bacterium, a virus or another microorganism), abnormal or damaged host cells (e.g., cancer cells), environmental agent (e.g., nicotine from a tobacco smoke), or an allergen (e.g., pollen or food protein). Immunogenic vaccines and therapies optimize the immune response to improve the eradication of the pathogen or damaged cells. In contrast, tolerogenic vaccines and therapies retrain or blunt the immune response to antigens, which are recognized by the immune system as harmful to the host. To optimize the immune response to either improve the immunogenicity or induce tolerance, researchers employ different routes of administration, antigen-delivery systems, and adjuvants. Nanocarriers and adjuvants are of particular interest to the fields of vaccines and immunotherapy as they allow for targeted delivery of the antigens and direct the immune response against these antigens in desirable direction (i.e., to either enhance immunogenicity or induce tolerance). Recently, nanoparticles gained particular attention as antigen carriers and adjuvants. This review focuses on a particular subclass of nanoparticles, which are made of nucleic acids, so-called nucleic acid nanoparticles or NANPs. Immunological properties of these novel materials and considerations for their clinical translation are discussed.
Collapse
Affiliation(s)
- Marina A Dobrovolskaia
- Nanotechnology Characterization Lab, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, MD 21702, USA
| |
Collapse
|
7
|
Phung CD, Nguyen HT, Tran TH, Choi HG, Yong CS, Kim JO. Rational combination immunotherapeutic approaches for effective cancer treatment. J Control Release 2018; 294:114-130. [PMID: 30553850 DOI: 10.1016/j.jconrel.2018.12.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 12/12/2018] [Accepted: 12/12/2018] [Indexed: 12/18/2022]
Abstract
Immunotherapy is an important mode of cancer treatment. Over the past decades, immunotherapy has improved the clinical outcome for cancer patients. However, in many cases, mutations in cancer cells, lack of selectivity, insufficiency of tumor-reactive T cells, and host immunosuppression limit the clinical benefit of immunotherapy. Combination approaches in immunotherapy may overcome these obstacles. Accumulating evidence demonstrates that combination immunotherapy is the future of cancer treatment. However, designing safe and rational combinations of immunotherapy with other treatment modalities is critical. This review will discuss the optimal immunotherapy-based combinations mainly with respect to the mechanisms of action of individual therapeutic agents that target multiple steps in evasion and progression of tumor.
Collapse
Affiliation(s)
- Cao Dai Phung
- College of Pharmacy, Yeungnam University, 280 Deahak-ro, Gyeongsan 38541, Republic of Korea
| | - Hanh Thuy Nguyen
- College of Pharmacy, Yeungnam University, 280 Deahak-ro, Gyeongsan 38541, Republic of Korea
| | - Tuan Hiep Tran
- Department for Management of Science and Technology Development, Ton Duc Thang University, Ho Chi Minh City, Viet Nam; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, Viet Nam
| | - Han-Gon Choi
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, 55, Hanyangdaehak-ro, Sangnok-gu, Ansan 426-791, Republic of Korea
| | - Chul Soon Yong
- College of Pharmacy, Yeungnam University, 280 Deahak-ro, Gyeongsan 38541, Republic of Korea
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, 280 Deahak-ro, Gyeongsan 38541, Republic of Korea.
| |
Collapse
|
8
|
Zhao W, Zhao G, Zhang S, Wang X, Yu X, Wang B. Clearance of HBeAg and HBsAg of HBV in mice model by a recombinant HBV vaccine combined with GM-CSF and IFN-α as an effective therapeutic vaccine adjuvant. Oncotarget 2018; 9:34213-34228. [PMID: 30344938 PMCID: PMC6188151 DOI: 10.18632/oncotarget.25789] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 10/30/2017] [Indexed: 12/19/2022] Open
Abstract
Chronic hepatitis B virus (CHB) infection is a significant public threat. Current interferon-α (IFN-α) based therapies and anti-viral drugs have failed to clear the infection in the majority of CHB patients and animal models. In our previous study, we established a combined protocol that employed a 3-day pretreatment with granulocyte-macrophage colony stimulating factor (GM-CSF) prior to a standard HBV vaccine. It achieved a 90% reduction of HBsAg level in the HBsAg transgenic mouse model. This protocol, while effective, remains too complex for clinical use. In this study, we formulated a new regimen by combining GM-CSF, IFN-α and a recombinant HBV vaccine (GM-CSF/IFN-α/VACCINE) into a single preparation and tested its efficacy in a HBV infection model. After four vaccinations, both serum HBeAg and HBsAg were cleared, accompanied by a 95% reduction of HBV+ hepatocytes and the presence of a large number of infiltrating CD8+ T cells in the liver. Mechanistically these robust responses were initiated by a vaccine-induced conversion of CCR2-dependent CD11b+Ly6Chi monocytes into CD11b+CD11c+ DCs. This finding sheds light on the potential mechanism of action of the GM-CSF-based vaccine adjuvant and provides definable markers for clinical assessment during future testing of such highly potent vaccine protocols in HBV patients.
Collapse
Affiliation(s)
- Weidong Zhao
- Key Laboratory of Medical Molecular Virology of The Ministry of Health and Ministry of Education, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Gan Zhao
- Key Laboratory of Medical Molecular Virology of The Ministry of Health and Ministry of Education, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Shuren Zhang
- Key Laboratory of Medical Molecular Virology of The Ministry of Health and Ministry of Education, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xianzheng Wang
- Key Laboratory of Medical Molecular Virology of The Ministry of Health and Ministry of Education, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xueping Yu
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Bin Wang
- Key Laboratory of Medical Molecular Virology of The Ministry of Health and Ministry of Education, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
9
|
Zhao D, Plotnikoff N, Griffin N, Song T, Shan F. Methionine enkephalin, its role in immunoregulation and cancer therapy. Int Immunopharmacol 2016; 37:59-64. [DOI: 10.1016/j.intimp.2016.02.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 02/16/2016] [Accepted: 02/16/2016] [Indexed: 11/25/2022]
|
10
|
Zhu M, Xu W, Su H, Huang Q, Wang B. Addition of CpG ODN and Poly (I:C) to a standard maturation cocktail generates monocyte-derived dendritic cells and induces a potent Th1 polarization with migratory capacity. Hum Vaccin Immunother 2016; 11:1596-605. [PMID: 26039883 DOI: 10.1080/21645515.2015.1046659] [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] [Indexed: 12/23/2022] Open
Abstract
Monocyte-derived dendritic cells (DCs) are used as immunoadjuvant cells in cancer vaccines and have made great progress. However, an optimal DCs subset is vital for this treatment effect, the current 'gold standard' cytokine cocktail DCs have a shortcoming in their cytokines secretion, especially IL-12p70, mainly because of the existence of PGE2. Therefore, it is necessary to find an appropriate DCs-based immunotherapeutic protocol. In this study, we compared a novel 'improved' maturation cytokine cocktail with the current 'gold standard' maturation cytokine cocktail used for generating standard DCs. The 'improved' maturation cytokine cocktail DCs showed a higher levels surface markers expression (CD80, CD83, CD86 and HLA-DR), the chemokine receptors CXCR4 and CCR7 and chemokine CCL19, CCL21 and CXCL21, whereas CCR5 expression was reduced. Most importantly, in contrast to 'gold standard' DCs, which secrete little IL-12p70 and as a result induce mainly Th2 immunity, 'improved' cytokine cocktail DCs secreted higher levels IL-12p70 and also secreted similar concentration IL-10. To removal of PGE2 from the 'improved' DCs did increase the IL-12p70 production. In conclusion, we here present the 'improved' DCs, as an optimal maturation cocktail protocol, can induce high migratory potential, generate immunostimulatory DCs, produce higher levels IL-12p70 with superior capacity to induce Th1 immunity, when compared with the 'gold standard' DCs.
Collapse
Affiliation(s)
- Mei Zhu
- a Department of Laboratory Medicine ; Affiliated Provincial Hospital of Anhui Medical University ; Hefei , Anhui , China
| | | | | | | | | |
Collapse
|
11
|
Liu R, Luo F, Liu X, Wang L, Yang J, Deng Y, Huang E, Qian J, Lu Z, Jiang X, Zhang D, Chu Y. Biological Response Modifier in Cancer Immunotherapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 909:69-138. [PMID: 27240457 DOI: 10.1007/978-94-017-7555-7_2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Biological response modifiers (BRMs) emerge as a lay of new compounds or approaches used in improving cancer immunotherapy. Evidences highlight that cytokines, Toll-like receptor (TLR) signaling, and noncoding RNAs are of crucial roles in modulating antitumor immune response and cancer-related chronic inflammation, and BRMs based on them have been explored. In particular, besides some cytokines like IFN-α and IL-2, several Toll-like receptor (TLR) agonists like BCG, MPL, and imiquimod are also licensed to be used in patients with several malignancies nowadays, and the first artificial small noncoding RNA (microRNA) mimic, MXR34, has entered phase I clinical study against liver cancer, implying their potential application in cancer therapy. According to amounts of original data, this chapter will review the regulatory roles of TLR signaling, some noncoding RNAs, and several key cytokines in cancer and cancer-related immune response, as well as the clinical cases in cancer therapy based on them.
Collapse
Affiliation(s)
- Ronghua Liu
- Department of Immunology, Key Laboratory of Medical Molecular Virology of MOE/MOH, School of Basic Medical Sciences, Fudan University, No.138, Yi Xue Yuan Rd., mail box 226, Shanghai, 200032, People's Republic of China.,Biotherapy Research Center, Fudan University, Shanghai, 200032, China
| | - Feifei Luo
- Biotherapy Research Center, Fudan University, Shanghai, 200032, China.,Department of Digestive Diseases of Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaoming Liu
- Department of Immunology, Key Laboratory of Medical Molecular Virology of MOE/MOH, School of Basic Medical Sciences, Fudan University, No.138, Yi Xue Yuan Rd., mail box 226, Shanghai, 200032, People's Republic of China.,Department of Dermatology, Shenzhen Hospital, Peking University, Shenzhen, Guangdong, 518036, China
| | - Luman Wang
- Department of Immunology, Key Laboratory of Medical Molecular Virology of MOE/MOH, School of Basic Medical Sciences, Fudan University, No.138, Yi Xue Yuan Rd., mail box 226, Shanghai, 200032, People's Republic of China.,Biotherapy Research Center, Fudan University, Shanghai, 200032, China
| | - Jiao Yang
- Department of Immunology, Key Laboratory of Medical Molecular Virology of MOE/MOH, School of Basic Medical Sciences, Fudan University, No.138, Yi Xue Yuan Rd., mail box 226, Shanghai, 200032, People's Republic of China.,Biotherapy Research Center, Fudan University, Shanghai, 200032, China
| | - Yuting Deng
- Department of Immunology, Key Laboratory of Medical Molecular Virology of MOE/MOH, School of Basic Medical Sciences, Fudan University, No.138, Yi Xue Yuan Rd., mail box 226, Shanghai, 200032, People's Republic of China.,Biotherapy Research Center, Fudan University, Shanghai, 200032, China
| | - Enyu Huang
- Department of Immunology, Key Laboratory of Medical Molecular Virology of MOE/MOH, School of Basic Medical Sciences, Fudan University, No.138, Yi Xue Yuan Rd., mail box 226, Shanghai, 200032, People's Republic of China.,Biotherapy Research Center, Fudan University, Shanghai, 200032, China
| | - Jiawen Qian
- Department of Immunology, Key Laboratory of Medical Molecular Virology of MOE/MOH, School of Basic Medical Sciences, Fudan University, No.138, Yi Xue Yuan Rd., mail box 226, Shanghai, 200032, People's Republic of China.,Biotherapy Research Center, Fudan University, Shanghai, 200032, China
| | - Zhou Lu
- Department of Immunology, Key Laboratory of Medical Molecular Virology of MOE/MOH, School of Basic Medical Sciences, Fudan University, No.138, Yi Xue Yuan Rd., mail box 226, Shanghai, 200032, People's Republic of China.,Biotherapy Research Center, Fudan University, Shanghai, 200032, China
| | - Xuechao Jiang
- Department of Immunology, Key Laboratory of Medical Molecular Virology of MOE/MOH, School of Basic Medical Sciences, Fudan University, No.138, Yi Xue Yuan Rd., mail box 226, Shanghai, 200032, People's Republic of China.,Biotherapy Research Center, Fudan University, Shanghai, 200032, China
| | - Dan Zhang
- Department of Immunology, Key Laboratory of Medical Molecular Virology of MOE/MOH, School of Basic Medical Sciences, Fudan University, No.138, Yi Xue Yuan Rd., mail box 226, Shanghai, 200032, People's Republic of China.,Biotherapy Research Center, Fudan University, Shanghai, 200032, China
| | - Yiwei Chu
- Department of Immunology, Key Laboratory of Medical Molecular Virology of MOE/MOH, School of Basic Medical Sciences, Fudan University, No.138, Yi Xue Yuan Rd., mail box 226, Shanghai, 200032, People's Republic of China. .,Biotherapy Research Center, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
12
|
Velcheti V, Schalper K. Basic Overview of Current Immunotherapy Approaches in Cancer. Am Soc Clin Oncol Educ Book 2016; 35:298-308. [PMID: 27249709 DOI: 10.1200/edbk_156572] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Recent success of immunotherapy strategies such as immune checkpoint blockade in several malignancies has established the role of immunotherapy in the treatment of cancer. Cancers use multiple mechanisms to co-opt the host-tumor immune interactions, leading to immune evasion. Our understanding of the host-tumor interactions has evolved over the past few years and led to various promising new therapeutic strategies. This article will focus on the basic principles of immunotherapy, novel pathways/agents, and combinatorial immunotherapies.
Collapse
Affiliation(s)
- Vamsidhar Velcheti
- From the Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH; Departments of Pathology and Medicine (Medical Oncology), Yale School of Medicine, New Haven, CT
| | - Kurt Schalper
- From the Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH; Departments of Pathology and Medicine (Medical Oncology), Yale School of Medicine, New Haven, CT
| |
Collapse
|
13
|
Boghozian R, Saei A, Mirzaei R, Jamali A, Vaziri B, Razavi A, Hadjati J. Identification of Toxoplasma gondii protein fractions induce immune response against melanoma in mice. APMIS 2015; 123:800-9. [PMID: 26152792 DOI: 10.1111/apm.12420] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 06/04/2015] [Indexed: 12/11/2022]
Abstract
Dendritic cells (DCs) play a crucial role in the initiation of adaptive immune responses against tumor cells. We recently found that protein components of Toxoplasma gondii (T. gondii) could mature DCs efficiently. Therefore, in this study, we aimed to find the most effective protein components of T. gondii which are able to mature DCs and consequently instruct immune responses in tumor-bearing mice. Soluble tachyzoite antigens (STAgs) were fractionated by ammonium sulfate precipitation and subsequently by anion-exchange HPLC. Immature DCs (iDCs) were treated by these protein fractions and were monitored for IL-12p70 and IL-10 production. Moreover, the capacity of mature DCs (mDCs) to induce lymphocyte proliferation was investigated. Ultimately, we analyzed the ability of mDCs in instructing immune responses in tumor-bearing mice. We found that ammonium sulfate fraction one (A1) matured-DCs produced higher IL-12 level and IL-12/IL-10 ratio; therefore, this fraction was selected for further fractionation by anion-exchange HPLC. The results showed that anion-exchange HPLC fraction 14 (C14) matured-DCs secrete higher levels of IL-12p70 and IL-12p70/IL-10 ratio. Survival of the mice matured by A1 fraction increased significantly compared to other groups. Moreover, SDS-PAGE electrophoresis showed that different obtained fractions have distinct proteins based on their size. These results demonstrate that two protein fractions of T. gondii are able to mature DCs more efficient.
Collapse
Affiliation(s)
- Roobina Boghozian
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Azad Saei
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Mirzaei
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Arezoo Jamali
- Department of Laboratory Sciences, School of Paramedicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Behrooz Vaziri
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Alireza Razavi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Jamshid Hadjati
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Type I interferons as regulators of human antigen presenting cell functions. Toxins (Basel) 2014; 6:1696-723. [PMID: 24866026 PMCID: PMC4073125 DOI: 10.3390/toxins6061696] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 05/15/2014] [Accepted: 05/16/2014] [Indexed: 01/08/2023] Open
Abstract
Type I interferons (IFNs) are pleiotropic cytokines, initially described for their antiviral activity. These cytokines exhibit a long record of clinical use in patients with some types of cancer, viral infections and chronic inflammatory diseases. It is now well established that IFN action mostly relies on their ability to modulate host innate and adaptive immune responses. Work in recent years has begun to elucidate the mechanisms by which type I IFNs modify the immune response, and this is now recognized to be due to effects on multiple cell types, including monocytes, dendritic cells (DCs), NK cells, T and B lymphocytes. An ensemble of results from both animal models and in vitro studies emphasized the key role of type I IFNs in the development and function of DCs, suggesting the existence of a natural alliance between these cytokines and DCs in linking innate to adaptive immunity. The identification of IFN signatures in DCs and their dysregulation under pathological conditions will therefore be pivotal to decipher the complexity of this DC-IFN interaction and to better exploit the therapeutic potential of these cells.
Collapse
|
15
|
Human metapneumovirus SH and G glycoproteins inhibit macropinocytosis-mediated entry into human dendritic cells and reduce CD4+ T cell activation. J Virol 2014; 88:6453-69. [PMID: 24672038 DOI: 10.1128/jvi.03261-13] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Human metapneumovirus (HMPV) is a major etiologic agent of respiratory disease worldwide. HMPV reinfections are common in healthy adults and children, suggesting that the protective immune response to HMPV is incomplete and short-lived. We used gene-deletion viruses to evaluate the role of the attachment G and small hydrophobic SH glycoproteins on virus uptake by primary human monocyte-derived dendritic cells (MDDC) in vitro and on subsequent MDDC maturation and activation of autologous T cells. HMPV with deletion of G and SH (ΔSHG) exhibited increased infectivity but had little effect on MDDC maturation. However, MDDC stimulated with ΔSHG induced increased proliferation of autologous Th1-polarized CD4(+) T cells. This effect was independent of virus replication. Increased T cell proliferation was strictly dependent on contact between virus-stimulated MDDC and CD4(+) T cells. Confocal microscopy revealed that deletion of SH and G was associated with an increased number of immunological synapses between memory CD4(+) T cells and virus-stimulated MDDC. Uptake of HMPV by MDDC was found to be primarily by macropinocytosis. Uptake of wild-type (WT) virus was reduced compared to that of ΔSHG, indicative of inhibition by the SH and G glycoproteins. In addition, DC-SIGN-mediated endocytosis provided a minor alternative pathway that depended on SH and/or G and thus operated only for WT. Altogether, our results show that SH and G glycoproteins reduce the ability of HMPV to be internalized by MDDC, resulting in a reduced ability of the HMPV-stimulated MDDC to activate CD4(+) T cells. This study describes a previously unknown mechanism of virus immune evasion. IMPORTANCE Human metapneumovirus (HMPV) is a major etiologic agent of respiratory disease worldwide. HMPV reinfections are common in healthy adults and children, suggesting that the protective immune response to HMPV is incomplete and short-lived. We found that HMPV attachment G and small hydrophobic SH glycoproteins reduce the ability of HMPV to be internalized by macropinocytosis into human dendritic cells (DC). This results in a reduced ability of the HMPV-stimulated DC to activate Th1-polarized CD4(+) T cells. These results contribute to a better understanding of the nature of incomplete protection against this important human respiratory virus, provide new information on the entry of HMPV into human cells, and describe a new mechanism of virus immune evasion.
Collapse
|
16
|
Meng Y, Wang Q, Zhang Z, Wang E, Plotnikoff NP, Shan F. Synergistic effect of methionine encephalin (MENK) combined with pidotimod(PTD) on the maturation of murine dendritic cells (DCs). Hum Vaccin Immunother 2013; 9:773-83. [PMID: 23470544 PMCID: PMC3903895 DOI: 10.4161/hv.23137] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Revised: 11/26/2012] [Accepted: 12/06/2012] [Indexed: 12/18/2022] Open
Abstract
To gain new insight into the functional interaction between dendritic cells and methionine encephalin (MENK) combined with pidotimod (PTD), we have analyzed the effect of MENK plus PTD on the morphology, phenotype and functions of murine bone-marrow derived dendritic cells (BMDCs) in vitro. The maturation of BMDCs cultured in the presence of either MENK or PTD alone, or MENK in combination with PTD, was detected. The cell proliferation was measured by 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxy-methoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt/phenazinemethosulphate (MTS/PMS). The changes of BMDCs morphology were confirmed with light microscopy, transmission electron microscopy (TEM) and scanning electron microscopy (SEM). The BMDCs treated with MENK combined with PTD displayed a higher expression of typical maturation markers of CD40, CD80, CD83, CD86 and MHC-IIidentified by fluorescence activated cell sorting (FACS), and stronger ability to drive T cells. The decrease of the endocytic ability was assayed by DAB kit, FITC-dextran and cellular immunohistochemistry. Finally upregulation of cytokines production of IL-12 and TNF-α was determined by ELISA. These data indicate that MENK combined with PTD could exert synergistic action on BMDC maturation.
Collapse
Affiliation(s)
- Yiming Meng
- Department of immunology; School of Basic Medical Science; China Medical University; Shenyang, P.R. China
| | - Qiushi Wang
- Central Blood Bank; Shengjing Hospital; China Medical University; Shenyang, P.R. China
| | - Zhenjie Zhang
- Department of immunology; School of Basic Medical Science; China Medical University; Shenyang, P.R. China
| | - Enhua Wang
- Institute of pathology and pathophysiology; School of Basic Medical Science; China Medical University; Shenyang, P.R. China
| | | | - Fengping Shan
- Department of immunology; School of Basic Medical Science; China Medical University; Shenyang, P.R. China
| |
Collapse
|
17
|
Pavón EJ, Zumaquero E, Rosal-Vela A, Khoo KM, Cerezo-Wallis D, García-Rodríguez S, Carrascal M, Abian J, Graeff R, Callejas-Rubio JL, Ortego-Centeno N, Malavasi F, Zubiaur M, Sancho J. Increased CD38 expression in T cells and circulating anti-CD38 IgG autoantibodies differentially correlate with distinct cytokine profiles and disease activity in systemic lupus erythematosus patients. Cytokine 2013; 62:232-43. [PMID: 23538292 DOI: 10.1016/j.cyto.2013.02.023] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Revised: 02/14/2013] [Accepted: 02/16/2013] [Indexed: 01/05/2023]
Abstract
CD38 is a multifunctional protein possessing ADP-ribosyl cyclase activity responsible for both the synthesis and the degradation of several Ca(2+)-mobilizing second messengers. In mammals, CD38 also functions as a receptor. In this study CD38 expression in CD4(+), CD8(+), or CD25(+) T cells was significantly higher in systemic lupus erythematosus (SLE) patients than in Normal controls. Increased CD38 expression in SLE T cells correlated with plasma levels of Th2 (IL-4, IL-10, IL-13) and Th1 (IL-1β, IL-12, IFN-γ, TNF-α) cytokines, and was more prevalent in clinically active SLE patients than in Normal controls. In contrast, elevated anti-CD38 IgG autoantibodies were more frequent in clinically quiescent SLE patients (SLEDAI=0) than in Normal controls, and correlated with moderate increased plasma levels of IL-10 and IFN-γ. However, clinically active SLE patients were mainly discriminated from quiescent SLE patients by increased levels of IL-10 and anti-dsDNA antibodies, with odds ratios (ORs) of 3.7 and 4.8, respectively. Increased frequency of anti-CD38 autoantibodies showed an inverse relationship with clinical activity (OR=0.43), and in particular with the frequency of anti-dsDNA autoantibodies (OR=0.21). Increased cell death occurred in CD38(+) Jurkat T cells treated with anti-CD38(+) SLE plasmas, and not in these cells treated with anti-CD38(-) SLE plasmas, or Normal plasmas. This effect did not occur in CD38-negative Jurkat T cells, suggesting that it could be attributed to anti-CD38 autoantibodies. These results support the hypothesis that anti-CD38 IgG autoantibodies or their associated plasma factors may dampen immune activation by affecting the viability of CD38(+) effector T cells and may provide protection from certain clinical SLE features.
Collapse
Affiliation(s)
- Esther J Pavón
- Instituto de Parasitología y Biomedicina López-Neyra (IPBLN), Consejo Superior de Investigaciones Científicas (CSIC), Parque Tecnológico de Ciencias de la Salud (PTS), Avenida del Conocimiento s/n, 18016 Armilla, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Kameshima H, Tsuruma T, Kutomi G, Shima H, Iwayama Y, Kimura Y, Imamura M, Torigoe T, Takahashi A, Hirohashi Y, Tamura Y, Tsukahara T, Kanaseki T, Sato N, Hirata K. Immunotherapeutic benefit of α-interferon (IFNα) in survivin2B-derived peptide vaccination for advanced pancreatic cancer patients. Cancer Sci 2012; 104:124-9. [PMID: 23078230 DOI: 10.1111/cas.12046] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Revised: 10/04/2012] [Accepted: 10/10/2012] [Indexed: 12/14/2022] Open
Abstract
Survivin, a member of the inhibitor of apoptosis protein (IAP) family containing a single baculovirus IAP repeat domain, is highly expressed in cancerous tissues but not in normal counterparts. Our group identified an HLA-A24-restricted antigenic peptide, survivin-2B80-88 (AYACNTSTL), that is recognized by CD8 + CTLs and functions as an immunogenic molecule in patients with cancers of various histological origins such as colon, breast, lung, oral, and urogenital malignancies. Subsequent clinical trials with this epitope peptide alone resulted in clinical and immunological responses. However, these were not strong enough for routine clinical use as a therapeutic cancer vaccine, and our previous study of colon cancer patients indicated that treatment with a vaccination protocol of survivin-2B80-88 plus incomplete Freund's adjuvant (IFA) and α-interferon (IFNα) conferred overt clinical improvement and enhanced the immunological responses of patients. In the current study, we further investigated whether this vaccination protocol could efficiently provide not only improved immune responses but also better clinical outcomes for advanced pancreatic cancers. Tetramer and enzyme-linked immunosorbent spot analysis data indicated that more than 50% of the patients had positive clinical and immunological responses. In contrast, assessment of treatment with IFNα only to another group of cancer patients resulted in no obvious increase in the frequency of survivin-2B80-88 peptide-specific CTLs. Taken together, our data clearly indicate that a vaccination protocol of survivin-2B80-88 plus IFA and IFNα is very effective and useful in immunotherapy for this type of poor-prognosis neoplasm. This trial was registered with the UMIN Clinical Trials Registry, no. UMIN000000905.
Collapse
Affiliation(s)
- Hidekazu Kameshima
- Department of Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Frasca L, Lande R. Overlapping, additive and counterregulatory effects of type II and I interferons on myeloid dendritic cell functions. ScientificWorldJournal 2011; 11:2071-90. [PMID: 22125457 PMCID: PMC3221594 DOI: 10.1100/2011/873895] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Accepted: 09/27/2011] [Indexed: 12/19/2022] Open
Abstract
Dendritic cells (DCs) are central player in immunity by bridging the innate and adaptive arms of the immune system (IS). Interferons (IFNs) are one of the most important factors that regulate both innate and adaptive immunity too. Thus, the understanding of how type II and I IFNs modulate the immune-regulatory properties of DCs is a central issue in immunology. In this paper, we will address this point in the light of the most recent literature, also highlighting the controversial data reported in the field. According to the wide literature available, type II as well as type I IFNs appear, at the same time, to collaborate, to induce additive effects or overlapping functions, as well as to counterregulate each one's effects on DC biology and, in general, the immune response. The knowledge of these effects has important therapeutic implications in the treatment of infectious/autoimmune diseases and cancer and indicates strategies for using IFNs as vaccine adjuvants and in DC-based immune therapeutic approaches.
Collapse
Affiliation(s)
- Loredana Frasca
- Department of Infectious, Parasitic and Immune-mediated Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy.
| | | |
Collapse
|
20
|
Lee S, Margolin K. Cytokines in cancer immunotherapy. Cancers (Basel) 2011; 3:3856-93. [PMID: 24213115 PMCID: PMC3763400 DOI: 10.3390/cancers3043856] [Citation(s) in RCA: 461] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Revised: 09/24/2011] [Accepted: 09/27/2011] [Indexed: 02/06/2023] Open
Abstract
Cytokines are molecular messengers that allow the cells of the immune system to communicate with one another to generate a coordinated, robust, but self-limited response to a target antigen. The growing interest over the past two decades in harnessing the immune system to eradicate cancer has been accompanied by heightened efforts to characterize cytokines and exploit their vast signaling networks to develop cancer treatments. The goal of this paper is to review the major cytokines involved in cancer immunotherapy and discuss their basic biology and clinical applications. The paper will also describe new cytokines in pre-clinical development, combinations of biological agents, novel delivery mechanisms, and potential directions for future investigation using cytokines.
Collapse
Affiliation(s)
- Sylvia Lee
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA 98195, USA; E-Mail:
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Kim Margolin
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA 98195, USA; E-Mail:
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| |
Collapse
|
21
|
Bacher N, Graulich E, Jonuleit H, Grabbe S, Steinbrink K. Interferon-α abrogates tolerance induction by human tolerogenic dendritic cells. PLoS One 2011; 6:e22763. [PMID: 21818385 PMCID: PMC3144929 DOI: 10.1371/journal.pone.0022763] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Accepted: 06/29/2011] [Indexed: 02/08/2023] Open
Abstract
Background Administration of interferon-α (IFN-α) represents an approved adjuvant therapy as reported for malignancies like melanoma and several viral infections. In malignant diseases, tolerance processes are critically involved in tumor progression. In this study, the effect of IFN-α on tolerance induction by human tolerogenic dendritic cells (DC) was analyzed. We focussed on tolerogenic IL-10-modulated DC (IL-10 DC) that are known to induce anergic regulatory T cells (iTregs). Methodology/Principal Findings IFN-α promoted an enhanced maturation of IL-10 DC as demonstrated by upregulation of the differentiation marker CD83 as well as costimulatory molecules. IFN-α treatment resulted in an increased capacity of DC to stimulate T cell activation compared to control tolerogenic DC. We observed a strengthened T cell proliferation and increased IFN-γ production of CD4+ and CD8+ T cells stimulated by IFN-α-DC, demonstrating a restoration of the immunogenic capacity of tolerogenic DC in the presence of IFN-α. Notably, restimulation experiments revealed that IFN-α treatment of tolerogenic DC abolished the induction of T cell anergy and suppressor function of iTregs. In contrast, IFN-α neither affected the priming of iTregs nor converted iTregs into effector T cells. Conclusions/Significance IFN-α inhibits the induction of T cell tolerance by reversing the tolerogenic function of human DC.
Collapse
Affiliation(s)
- Nicole Bacher
- Department of Dermatology, University Medical Center Mainz, University of Mainz, Mainz, Germany
- * E-mail: (KS); (NB)
| | - Edith Graulich
- Department of Dermatology, University Medical Center Mainz, University of Mainz, Mainz, Germany
| | - Helmut Jonuleit
- Department of Dermatology, University Medical Center Mainz, University of Mainz, Mainz, Germany
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center Mainz, University of Mainz, Mainz, Germany
| | - Kerstin Steinbrink
- Department of Dermatology, University Medical Center Mainz, University of Mainz, Mainz, Germany
- * E-mail: (KS); (NB)
| |
Collapse
|
22
|
Farkas A, Kemény L. Interferon-α in the generation of monocyte-derived dendritic cells: recent advances and implications for dermatology. Br J Dermatol 2011; 165:247-54. [PMID: 21410666 DOI: 10.1111/j.1365-2133.2011.10301.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Dendritic cells (DCs) have a critical role in antiviral responses, in autoimmune disease pathogenesis and in initiating and maintaining inflammatory skin disorders, and are candidates for cell-based immunotherapeutic approaches for tumours. Recent studies have shown the important role of type I interferons (IFNs) in DC differentiation and activation. In the presence of IFN-α and granulocyte/macrophage colony-stimulating factor monocytes differentiate into DCs referred to as IFN-DCs. In vitro generated IFN-DCs show a partially mature phenotype, are effective in taking up antigens, share features of myeloid DCs, plasmacytoid DCs and natural killer cells, exhibit an enhanced chemotactic response and are capable of migrating to the lymph nodes. IFN-DCs produce several chemokines and cytokines, including T-helper 1 (Th1) mediators belonging to the interleukin-12 family. IFN-DCs stimulate T- and B-cell responses and the production of IFN-γ in mixed lymphocyte reactions and have a capacity to produce IFN-γ themselves. IFN-DCs express several toll-like receptor (TLR) subtypes and TLR ligand stimulation improves their costimulatory molecule expression, increases their Th1 cytokine production and enhances their capacity to stimulate naive T-cell proliferation. Here we review the interaction of IFN-α and monocytes and the role of IFN-DCs in infections, in autoimmunity, in inflammation and in cancer immunotherapy focusing on dermatological conditions.
Collapse
Affiliation(s)
- A Farkas
- Department of Dermatology and Allergology, University of Szeged, 6720 Szeged, Hungary.
| | | |
Collapse
|
23
|
Balas C, Kennel A, Deauvieau F, Sodoyer R, Arnaud-Barbe N, Lang J, Guy B. Different innate signatures induced in human monocyte-derived dendritic cells by wild-type dengue 3 virus, attenuated but reactogenic dengue 3 vaccine virus, or attenuated nonreactogenic dengue 1-4 vaccine virus strains. J Infect Dis 2011; 203:103-8. [PMID: 21148502 PMCID: PMC3086443 DOI: 10.1093/infdis/jiq022] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2010] [Accepted: 10/01/2010] [Indexed: 11/15/2022] Open
Abstract
DNA microarrays were used to assess the innate gene signature in human myeloid dendritic cells infected with chimeric dengue 1-4 vaccines, a wild-type dengue 3 virus, or a classically attenuated serotype 3 vaccine shown to be reactogenic in humans. We observed a very reproducible signature for each of the 4 chimeric dengue vaccines, involving stimulation of type I interferon and associated genes, together with genes encoding chemokines and other mediators involved in the initiation of adaptive responses. In contrast, wild-typeDEN3 virus induced a predominantly inflammatory profile, while the reactogenic attenuated serotype 3 vaccine appeared to induce a blunted response.
Collapse
Affiliation(s)
- Claire Balas
- Research and Development, Sanofi Pasteur, 69280, Marcy l'Étoile, France
| | - Audrey Kennel
- Research and Development, Sanofi Pasteur, 69280, Marcy l'Étoile, France
| | | | - Regis Sodoyer
- Research and Development, Sanofi Pasteur, 69280, Marcy l'Étoile, France
| | | | - Jean Lang
- Research and Development, Sanofi Pasteur, 69280, Marcy l'Étoile, France
| | - Bruno Guy
- Research and Development, Sanofi Pasteur, 69280, Marcy l'Étoile, France
| |
Collapse
|
24
|
Jung TY, Pham TNN, Umeyama A, Shoji N, Hashimoto T, Lee JJ, Takei M. Ursolic acid isolated from Uncaria rhynchophylla activates human dendritic cells via TLR2 and/or TLR4 and induces the production of IFN-gamma by CD4+ naïve T cells. Eur J Pharmacol 2010; 643:297-303. [PMID: 20599915 DOI: 10.1016/j.ejphar.2010.06.030] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2009] [Revised: 06/09/2010] [Accepted: 06/16/2010] [Indexed: 11/16/2022]
Abstract
Ursolic acid is triterpene isolated from Uncaria rhynchophylla and is a pharmacologically active substance. The induction of dendritic cell maturation is critical for the induction of Ag-specific T-lymphocyte response and may be essential for the development of human vaccine relying on T cell immunity. In this study, we investigated that the effect of Ursolic acid on the phenotypic and functional maturation of human monocyte-derived dendritic cells in vitro. Dendritic cells harvested on day 8 were examined using functional assay. The expression levels of CD1a, CD80, CD83, CD86, HLA-DR and CCR7 on Ursolic acid-primed dendritic cells was slightly enhanced. Ursolic acid dose-dependently enhanced the T cell stimulatory capacity in an allogeneic mixed lymphocyte reaction, as measured by T cell proliferation. The production of IL-12p70 induced by Ursolic acid-primed dendritic cells was inhibited by the anti-Toll-like receptor-2 (TLR2) mAb and anti-TLR4 mAb. Moreover, Ursolic acid-primed dendritic cells expressed levels of mRNA coding for both TLR2 and TLR4. The majority of cells produced considerable interferon-gamma (IFN-gamma), but also small amounts of interleukin (IL-4)-4. Ursolic acid-primed dendritic cells have an intermediate migratory capacity towards CCL19 and CCL21. These results suggest that Ursolic acid modulates human dendritic cells function in a fashion that favors Th1 polarization via the activation of IL-12p70 dependent on TLR2 and/or TLR4, and may be used on dendritic cells-based vaccines for cancer immunotherapy.
Collapse
Affiliation(s)
- Tae-Young Jung
- Department of Neurosurgery, Chonnam National University Medical School, Gwangiu, South Korea
| | | | | | | | | | | | | |
Collapse
|
25
|
Kaehler KC, Sondak VK, Schadendorf D, Hauschild A. Pegylated interferons: prospects for the use in the adjuvant and palliative therapy of metastatic melanoma. Eur J Cancer 2010; 46:41-6. [PMID: 19857957 DOI: 10.1016/j.ejca.2009.10.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2009] [Revised: 09/23/2009] [Accepted: 10/10/2009] [Indexed: 11/18/2022]
Abstract
Classic interferon-alpha formulations have antitumour activity in a variety of neoplastic diseases, including the adjuvant and palliative setting of metastatic melanoma, as single agents or in combination with chemotherapy and/or interleukin-2. Pegylated interferon, widely used for the treatment of hepatitis, seems to be at least equally efficacious as standard recombinant interferon in the treatment of metastatic melanoma, and the available evidence suggests that equi-efficacious doses have somewhat lower acute toxicity. Moreover, the favourable pharmacokinetic properties of pegylated interferon allow the administration on a weekly basis, with sustained exposure to interferon during that entire period. Several clinical trials have been conducted testing adjuvant and palliative treatment with pegylated interferon-alpha in high-risk melanoma patients with promising results. The role of pegylated interferons in the setting of advanced metastatic melanoma will need further investigation in clinical trials, potentially in combination with targeted or cytotoxic agents with regard to synergistic antiangiogenic and cytotoxic effects. The use of pegylated interferons in earlier stage melanomas will be investigated in upcoming trials.
Collapse
|