1
|
Wang L, Zhou M, Kong X, Wu S, Ding C, Hu X, Guo H, Yan J. Specific Targeting of STAT3 in B Cells Suppresses Progression of B Cell Lymphoma. Int J Mol Sci 2023; 24:13666. [PMID: 37686472 PMCID: PMC10563066 DOI: 10.3390/ijms241713666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 09/10/2023] Open
Abstract
The signal transducer and activator of transcription 3 (STAT3), which regulates multiple oncogenic processes, has been found to be constitutively activated in lymphoma, suggesting its potential as a therapeutic target. Here, we constructed an anti-CD19-N-(4-carboxycyclohexylmethyl) maleimide N-hydroxysuccinimide ester (SMCC)-protamine (CSP)-STAT3 small interfering RNA (siRNA) conjugate and demonstrated that the CSP-STAT3 siRNA conjugate could specifically bind to normal B cells and A20 lymphoma cells in vitro. It decreased the STAT3 expression in B cell lymphoma cell lines (A20, SU-DHL-2 and OCI-Ly3), resulting in reduced proliferation of lymphoma cells featured with lower S-phase and higher apoptosis. Using an A20 transplantable lymphoma model, we found that the CSP-STAT3 siRNA conjugate significantly inhibited tumor growth and weight. Ki-67, p-STAT3, STAT3, and serum IL-6 levels were all significantly reduced in A20-bearing mice treated with CSP-STAT3 siRNA. These findings indicate that specifically targeting STAT3 siRNA to B cell lymphoma cell lines can significantly decrease STAT3 activity and inhibit tumor progression in vitro and in vivo, suggesting its potential utilization for cancer treatment.
Collapse
Affiliation(s)
- Lipei Wang
- Division of Immunotherapy, The Hiram C. Polk, Jr. MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY 40202, USA; (M.Z.); (X.K.); (S.W.); (C.D.); (X.H.)
- School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou 310030, China
| | - Mingqian Zhou
- Division of Immunotherapy, The Hiram C. Polk, Jr. MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY 40202, USA; (M.Z.); (X.K.); (S.W.); (C.D.); (X.H.)
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xiangyu Kong
- Division of Immunotherapy, The Hiram C. Polk, Jr. MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY 40202, USA; (M.Z.); (X.K.); (S.W.); (C.D.); (X.H.)
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Shouzhen Wu
- Division of Immunotherapy, The Hiram C. Polk, Jr. MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY 40202, USA; (M.Z.); (X.K.); (S.W.); (C.D.); (X.H.)
| | - Chuanlin Ding
- Division of Immunotherapy, The Hiram C. Polk, Jr. MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY 40202, USA; (M.Z.); (X.K.); (S.W.); (C.D.); (X.H.)
| | - Xiaoling Hu
- Division of Immunotherapy, The Hiram C. Polk, Jr. MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY 40202, USA; (M.Z.); (X.K.); (S.W.); (C.D.); (X.H.)
| | - Haixun Guo
- Department of Radiology, University of Louisville School of Medicine, Louisville, KY 40202, USA;
| | - Jun Yan
- Division of Immunotherapy, The Hiram C. Polk, Jr. MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY 40202, USA; (M.Z.); (X.K.); (S.W.); (C.D.); (X.H.)
| |
Collapse
|
2
|
Ung T, Rutledge NS, Weiss AM, Esser-Kahn AP, Deak P. Cell-targeted vaccines: implications for adaptive immunity. Front Immunol 2023; 14:1221008. [PMID: 37662903 PMCID: PMC10468591 DOI: 10.3389/fimmu.2023.1221008] [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: 05/11/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023] Open
Abstract
Recent advancements in immunology and chemistry have facilitated advancements in targeted vaccine technology. Targeting specific cell types, tissue locations, or receptors can allow for modulation of the adaptive immune response to vaccines. This review provides an overview of cellular targets of vaccines, suggests methods of targeting and downstream effects on immune responses, and summarizes general trends in the literature. Understanding the relationships between vaccine targets and subsequent adaptive immune responses is critical for effective vaccine design. This knowledge could facilitate design of more effective, disease-specialized vaccines.
Collapse
Affiliation(s)
- Trevor Ung
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Nakisha S. Rutledge
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Adam M. Weiss
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Aaron P. Esser-Kahn
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Peter Deak
- Chemical and Biological Engineering Department, Drexel University, Philadelphia, PA, United States
| |
Collapse
|
3
|
Xiong J, Chi H, Yang G, Zhao S, Zhang J, Tran LJ, Xia Z, Yang F, Tian G. Revolutionizing anti-tumor therapy: unleashing the potential of B cell-derived exosomes. Front Immunol 2023; 14:1188760. [PMID: 37342327 PMCID: PMC10277631 DOI: 10.3389/fimmu.2023.1188760] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/22/2023] [Indexed: 06/22/2023] Open
Abstract
B cells occupy a vital role in the functioning of the immune system, working in tandem with T cells to either suppress or promote tumor growth within the tumor microenvironment(TME). In addition to direct cell-to-cell communication, B cells and other cells release exosomes, small membrane vesicles ranging in size from 30-150 nm, that facilitate intercellular signaling. Exosome research is an important development in cancer research, as they have been shown to carry various molecules such as major histocompatibility complex(MHC) molecules and integrins, which regulate the TME. Given the close association between TME and cancer development, targeting substances within the TME has emerged as a promising strategy for cancer therapy. This review aims to present a comprehensive overview of the contributions made by B cells and exosomes to the tumor microenvironment (TME). Additionally, we delve into the potential role of B cell-derived exosomes in the progression of cancer.
Collapse
Affiliation(s)
- Jingwen Xiong
- Department of Sports Rehabilitation, Southwest Medical University, Luzhou, China
| | - Hao Chi
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Guanhu Yang
- Department of Specialty Medicine, Ohio University, Athens, OH, United States
| | - Songyun Zhao
- Department of Neurosurgery, Wuxi People’s Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Jing Zhang
- Division of Basic Biomedical Sciences, The University of South Dakota Sanford School of Medicine, Vermillion, SD, United States
| | - Lisa Jia Tran
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Zhijia Xia
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Fang Yang
- Department of Ophthalmology, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Gang Tian
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
4
|
Shimizu T, Kawaguchi Y, Ando H, Ishima Y, Ishida T. Development of an Antigen Delivery System for a B Cell-Targeted Vaccine as an Alternative to Dendritic Cell-Targeted Vaccines. Chem Pharm Bull (Tokyo) 2022; 70:341-350. [DOI: 10.1248/cpb.c22-00047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Taro Shimizu
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University
| | - Yoshino Kawaguchi
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University
| | - Hidenori Ando
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University
| | - Yu Ishima
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University
| | - Tatsuhiro Ishida
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University
| |
Collapse
|
5
|
Wang J, Yang J, Kopeček J. Nanomedicines in B cell-targeting therapies. Acta Biomater 2022; 137:1-19. [PMID: 34687954 PMCID: PMC8678319 DOI: 10.1016/j.actbio.2021.10.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/29/2021] [Accepted: 10/14/2021] [Indexed: 02/08/2023]
Abstract
B cells play multiple roles in immune responses related to autoimmune diseases as well as different types of cancers. As such, strategies focused on B cell targeting attracted wide interest and developed intensively. There are several common mechanisms various B cell targeting therapies have relied on, including direct B cell depletion, modulation of B cell antigen receptor (BCR) signaling, targeting B cell survival factors, targeting the B cell and T cell costimulation, and immune checkpoint blockade. Nanocarriers, used as drug delivery vehicles, possess numerous advantages to low molecular weight drugs, reducing drug toxicity, enhancing blood circulation time, as well as augmenting targeting efficacy and improving therapeutic effect. Herein, we review the commonly used targets involved in B cell targeting approaches and the utilization of various nanocarriers as B cell-targeted delivery vehicles. STATEMENT OF SIGNIFICANCE: As B cells are engaged significantly in the development of many kinds of diseases, utilization of nanomedicines in B cell depletion therapies have been rapidly developed. Although numerous studies focused on B cell targeting have already been done, there are still various potential receptors awaiting further investigation. This review summarizes the most relevant studies that utilized nanotechnologies associated with different B cell depletion approaches, providing a useful tool for selection of receptors, agents and/or nanocarriers matching specific diseases. Along with uncovering new targets in the function map of B cells, there will be a growing number of candidates that can benefit from nanoscale drug delivery.
Collapse
Affiliation(s)
- Jiawei Wang
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT, United States; Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, United States
| | - Jiyuan Yang
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT, United States; Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, United States
| | - Jindřich Kopeček
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT, United States; Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, United States; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States.
| |
Collapse
|
6
|
Complement activation induced by PEG enhances humoral immune responses against antigens encapsulated in PEG-modified liposomes. J Control Release 2021; 329:1046-1053. [DOI: 10.1016/j.jconrel.2020.10.033] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 10/09/2020] [Accepted: 10/16/2020] [Indexed: 12/16/2022]
|
7
|
Lv Z, Zhang P, Li D, Qin M, Nie L, Wang X, Ai L, Feng Z, Odhiambo WO, Ma Y, Ji Y. CD19-targeting fusion protein combined with PD1 antibody enhances anti-tumor immunity in mouse models. Oncoimmunology 2020; 9:1747688. [PMID: 32363119 PMCID: PMC7185221 DOI: 10.1080/2162402x.2020.1747688] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 03/04/2020] [Accepted: 03/17/2020] [Indexed: 12/24/2022] Open
Abstract
In our previous studies, using a B cell vaccine (scFv-Her2), the targeting of tumor-associated antigen Her2 (human epidermal growth factor receptor-2) to B cells via the anti-CD19 single chain variable fragment (scFv) was shown to augment tumor-specific immunity, which enhanced tumor control in the prophylactic and therapeutic setting. However, the fusion protein displayed limited activity against established tumors, and local relapses often occurred following scFv-Her2 treatment, indicating that scFv-Her2-induced responses are inadequate to maintain anti-tumor immunity. In this study, targeting the IV region (D4) of the extracellular region of Her2 to B cells via CD19 molecules (scFv-Her2D4) was found to enhance IFN-γ-producing-CD8+ T cell infiltration in tumor tissues and reduced the number of tumor-infiltrating myeloid-derived suppressor cells (MDSCs). However, negative co-stimulatory molecules such as programmed cell death protein-1 (PD-1), CD160, and LAG-3 on T cells and programmed death protein ligand-1 (PD-L1) on tumor cells were upregulated in the tumor microenvironment after scFv-Her2D4 treatment. Further, anti-PD1 administration enhanced the efficacy of scFv-Her2D4 and anti-tumor immunity, as evidenced by the reversal of tumor-infiltrating CD8+ T cell exhaustion and the reduction of MDSCs and Treg cells, which suppress T cells and alter the tumor immune microenvironment. Moreover, combining this with anti-PD1 antibodies promoted complete tumor rejection. Our data provide evidence of a close interaction among tumor vaccines, T cells, and the PD-L1/PD-1 axis and establish a basis for the rational design of combination therapy with immune modulators and tumor vaccine therapy.
Collapse
Affiliation(s)
- Zhuangwei Lv
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Institute of Infection and Immunity, Xi'an Jiaotong University Translational Medicine Center, Xi'an, China
| | - Ping Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Institute of Infection and Immunity, Xi'an Jiaotong University Translational Medicine Center, Xi'an, China
| | - Dandan Li
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Institute of Infection and Immunity, Xi'an Jiaotong University Translational Medicine Center, Xi'an, China
| | - Mengting Qin
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Institute of Infection and Immunity, Xi'an Jiaotong University Translational Medicine Center, Xi'an, China
| | - Longzhu Nie
- School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Xiaoqian Wang
- The Clinical Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Li Ai
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Zhaozu Feng
- School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Woodvine Otieno Odhiambo
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Yunfeng Ma
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Institute of Infection and Immunity, Xi'an Jiaotong University Translational Medicine Center, Xi'an, China
| | - Yanhong Ji
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Institute of Infection and Immunity, Xi'an Jiaotong University Translational Medicine Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an Jiaotong University, Xi'an, P. R. China
| |
Collapse
|
8
|
Jin H, Kong Z, Mehboob A, Jiang B, Xu J, Cai Y, Liu W, Hong J, Li Y. Transcriptional Profiles Associated with Marek's Disease Virus in Bursa and Spleen Lymphocytes Reveal Contrasting Immune Responses during Early Cytolytic Infection. Viruses 2020; 12:v12030354. [PMID: 32210095 PMCID: PMC7150966 DOI: 10.3390/v12030354] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 03/20/2020] [Accepted: 03/20/2020] [Indexed: 01/02/2023] Open
Abstract
Marek's disease virus (MDV), an alpha herpes virus, causes a lymphoproliferative state in chickens known as Marek's disease (MD), resulting in severe monetary losses to the poultry industry. Because lymphocytes of bursa of Fabricius and spleen are prime targets of MDV replication during the early cytolytic phase of infection, the immune response in bursa and spleen should be the foundation of late immunity induced by MDV. However, the mechanism of the MDV-mediated host immune response in lymphocytes in the early stage is poorly understood. The present study is primarily aimed at identifying the crucial genes and significant pathways involved in the immune response of chickens infected with MDV CVI988 and the very virulent RB1B (vvRB1B) strains. Using the RNA sequencing approach, we analyzed the generated transcriptomes from lymphocytes isolated from chicken bursa and spleen. Our findings validated the expression of previously characterized genes; however, they also revealed the expression of novel genes during the MDV-mediated immune response. The results showed that after challenge with CVI988 or vvRB1B strains, 634 and 313 differentially expressed genes (DEGs) were identified in splenic lymphocytes, respectively. However, 58 and 47 DEGs were observed in bursal lymphocytes infected with CVI988 and vvRB1B strains, respectively. Following MDV CVI988 or vvRB1B challenge, the bursal lymphocytes displayed changes in IL-6 and IL-4 gene expression. Surprisingly, splenic lymphocytes exhibited an overwhelming alteration in the expression of cytokines and cytokine receptors involved in immune response signaling. On the other hand, there was no distinct trend between infection with CVI988 and vvRB1B and the expression of cytokines and chemokines, such as IL-10, IFN-γ, STAT1, IRF1, CCL19, and CCL26. However, the expression profiles of IL-1β, IL-6, IL8L1, CCL4 (GGCL1), and CCL5 were significantly upregulated in splenic lymphocytes from chickens infected with CVI988 compared with those of chickens infected with vvRB1B. Because these cytokines and chemokines are considered to be associated with B cell activation and antigenic signal transduction to T cells, they may indicate differences of immune responses initiated by vaccinal and virulent strains during the early phase of infection. Collectively, our study provides valuable data on the transcriptional landscape using high-throughput sequencing to understand the different mechanism between vaccine-mediated protection and pathogenesis of virulent MDV in vivo.
Collapse
Affiliation(s)
- Huan Jin
- Research Center for Infectious Disease in Livestock and Poultry, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, China; (H.J.); (Z.K.); (A.M.); (B.J.); (J.X.); (Y.C.); (W.L.); (J.H.)
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, China
| | - Zimeng Kong
- Research Center for Infectious Disease in Livestock and Poultry, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, China; (H.J.); (Z.K.); (A.M.); (B.J.); (J.X.); (Y.C.); (W.L.); (J.H.)
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, China
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China
| | - Arslan Mehboob
- Research Center for Infectious Disease in Livestock and Poultry, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, China; (H.J.); (Z.K.); (A.M.); (B.J.); (J.X.); (Y.C.); (W.L.); (J.H.)
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, China
| | - Bo Jiang
- Research Center for Infectious Disease in Livestock and Poultry, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, China; (H.J.); (Z.K.); (A.M.); (B.J.); (J.X.); (Y.C.); (W.L.); (J.H.)
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, China
| | - Jian Xu
- Research Center for Infectious Disease in Livestock and Poultry, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, China; (H.J.); (Z.K.); (A.M.); (B.J.); (J.X.); (Y.C.); (W.L.); (J.H.)
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, China
| | - Yunhong Cai
- Research Center for Infectious Disease in Livestock and Poultry, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, China; (H.J.); (Z.K.); (A.M.); (B.J.); (J.X.); (Y.C.); (W.L.); (J.H.)
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, China
| | - Wenxiao Liu
- Research Center for Infectious Disease in Livestock and Poultry, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, China; (H.J.); (Z.K.); (A.M.); (B.J.); (J.X.); (Y.C.); (W.L.); (J.H.)
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, China
| | - Jiabing Hong
- Research Center for Infectious Disease in Livestock and Poultry, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, China; (H.J.); (Z.K.); (A.M.); (B.J.); (J.X.); (Y.C.); (W.L.); (J.H.)
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, China
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China
| | - Yongqing Li
- Research Center for Infectious Disease in Livestock and Poultry, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, China; (H.J.); (Z.K.); (A.M.); (B.J.); (J.X.); (Y.C.); (W.L.); (J.H.)
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agricultural and Forestry Sciences, Beijing 100097, China
- Correspondence: ; Tel.: +86-010-51503195
| |
Collapse
|
9
|
Efficient Acquisition of Fully Human Antibody Genes against Self-Proteins by Sorting Single B Cells Stimulated with Vaccines Based on Nitrated T Helper Cell Epitopes. J Immunol Res 2020; 2019:7914326. [PMID: 32083142 PMCID: PMC7012236 DOI: 10.1155/2019/7914326] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 11/07/2019] [Indexed: 11/19/2022] Open
Abstract
Single B cell antibody technology is a method for isolating antigen-specific B cells from human peripheral blood and obtaining antibody genes in developing antibody drugs. However, owing to immune tolerance to autoantigen, human autoantigen-specific B cells are difficult to acquire by conventional single B cell technology. In this study, we constructed a nitrated T-cell epitope named NitraTh by incorporating p-nitrophenylalanine into a universal T helper epitope. NitraTh had enhanced ability to activate CD4+ T cells and can be recognized by CD4+ T cells with different HLA class II haplotypes. This NitraTh can also break immune tolerance to autoantigens, such as human epidermal growth factor receptor 2 (HER2) and cannabinoid receptor 1, and induce strong specific IgM+ B cell responses in vitro. HER2-NitraTh vaccine can also stimulate the generation of HER2-specific IgG+ B cells in human immune system mice, which was established by cotransplanting lymphocytes and autologous dendritic cells in immunodeficient mice. We obtained 30 fully human IgG antibody genes by sorting single B cells from the human immune system mice immunized with HER2-NitraTh vaccine. The analysis of antibody genes showed that sorted B cells underwent the extensive somatic mutation of the antibody genes. We randomly selected eight genes for cloning, six of which expressed antibodies that can bind to HER2. Hence, we provided a convenient and effective method in acquiring fully human antibody genes against self-proteins, which can be used in developing therapeutic antibody drugs.
Collapse
|
10
|
Briquez PS, Hauert S, de Titta A, Gray LT, Alpar AT, Swartz MA, Hubbell JA. Engineering Targeting Materials for Therapeutic Cancer Vaccines. Front Bioeng Biotechnol 2020; 8:19. [PMID: 32117911 PMCID: PMC7026271 DOI: 10.3389/fbioe.2020.00019] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 01/10/2020] [Indexed: 12/24/2022] Open
Abstract
Therapeutic cancer vaccines constitute a valuable tool to educate the immune system to fight tumors and prevent cancer relapse. Nevertheless, the number of cancer vaccines in the clinic remains very limited to date, highlighting the need for further technology development. Recently, cancer vaccines have been improved by the use of materials, which can strongly enhance their intrinsic properties and biodistribution profile. Moreover, vaccine efficacy and safety can be substantially modulated through selection of the site at which they are delivered, which fosters the engineering of materials capable of targeting cancer vaccines to specific relevant sites, such as within the tumor or within lymphoid organs, to further optimize their immunotherapeutic effects. In this review, we aim to give the reader an overview of principles and current strategies to engineer therapeutic cancer vaccines, with a particular focus on the use of site-specific targeting materials. We will first recall the goal of therapeutic cancer vaccination and the type of immune responses sought upon vaccination, before detailing key components of cancer vaccines. We will then present how materials can be engineered to enhance the vaccine's pharmacokinetic and pharmacodynamic properties. Finally, we will discuss the rationale for site-specific targeting of cancer vaccines and provide examples of current targeting technologies.
Collapse
Affiliation(s)
- Priscilla S. Briquez
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, United States
| | - Sylvie Hauert
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, United States
| | | | - Laura T. Gray
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, United States
| | - Aaron T. Alpar
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, United States
| | - Melody A. Swartz
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, United States
- Ben May Department of Cancer Research, The University of Chicago, Chicago, IL, United States
- Committee on Immunology, The University of Chicago, Chicago, IL, United States
| | - Jeffrey A. Hubbell
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, United States
- Committee on Immunology, The University of Chicago, Chicago, IL, United States
| |
Collapse
|
11
|
Shimizu T, Abu Lila AS, Kawaguchi Y, Shimazaki Y, Watanabe Y, Mima Y, Hashimoto Y, Okuhira K, Storm G, Ishima Y, Ishida T. A Novel Platform for Cancer Vaccines: Antigen-Selective Delivery to Splenic Marginal Zone B Cells via Repeated Injections of PEGylated Liposomes. THE JOURNAL OF IMMUNOLOGY 2018; 201:2969-2976. [DOI: 10.4049/jimmunol.1701351] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 09/12/2018] [Indexed: 12/14/2022]
|
12
|
Jie J, Zhang Y, Zhou H, Zhai X, Zhang N, Yuan H, Ni W, Tai G. CpG ODN1826 as a Promising Mucin1-Maltose-Binding Protein Vaccine Adjuvant Induced DC Maturation and Enhanced Antitumor Immunity. Int J Mol Sci 2018; 19:ijms19030920. [PMID: 29558459 PMCID: PMC5877781 DOI: 10.3390/ijms19030920] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 03/13/2018] [Accepted: 03/15/2018] [Indexed: 12/14/2022] Open
Abstract
Mucin 1 (MUC1), being an oncogene, is an attractive target in tumor immunotherapy. Maltose binding protein (MBP) is a potent built-in adjuvant to enhance protein immunogenicity. Thus, a recombinant MUC1 and MBP antitumor vaccine (M-M) was constructed in our laboratory. To enhance the antitumor immune activity of M-M, CpG oligodeoxynucleotides 1826 (CpG 1826), a toll-like receptor-9 agonist, was examined in this study as an adjuvant. The combination of M-M and CpG 1826 significantly inhibited MUC1-expressing B16 cell growth and prolonged the survival of tumor-bearing mice. It induced MUC1-specific antibodies and Th1 immune responses, as well as the Cytotoxic T Lymphocytes (CTL) cytotoxicity in vivo. Further studies showed that it promoted the maturation and activation of the dendritic cell (DC) and skewed towards Th1 phenotype in vitro. Thus, our study revealed that CpG 1826 is an efficient adjuvant, laying a foundation for further M-M clinical research.
Collapse
Affiliation(s)
- Jing Jie
- Department of Immunology, College of Basic Medical Science, Jilin University, Xinjiang Street 125, Changchun 130021, China.
| | - Yixin Zhang
- Department of Immunology, College of Basic Medical Science, Jilin University, Xinjiang Street 125, Changchun 130021, China.
| | - Hongyue Zhou
- Department of Immunology, College of Basic Medical Science, Jilin University, Xinjiang Street 125, Changchun 130021, China.
| | - Xiaoyu Zhai
- Department of Immunology, College of Basic Medical Science, Jilin University, Xinjiang Street 125, Changchun 130021, China.
| | - Nannan Zhang
- Department of Immunology, College of Basic Medical Science, Jilin University, Xinjiang Street 125, Changchun 130021, China.
| | - Hongyan Yuan
- Department of Immunology, College of Basic Medical Science, Jilin University, Xinjiang Street 125, Changchun 130021, China.
| | - Weihua Ni
- Department of Immunology, College of Basic Medical Science, Jilin University, Xinjiang Street 125, Changchun 130021, China.
| | - Guixiang Tai
- Department of Immunology, College of Basic Medical Science, Jilin University, Xinjiang Street 125, Changchun 130021, China.
| |
Collapse
|
13
|
Stergiou N, Glaffig M, Jonuleit H, Schmitt E, Kunz H. Immunization with a Synthetic Human MUC1 Glycopeptide Vaccine against Tumor‐Associated MUC1 Breaks Tolerance in Human MUC1 Transgenic Mice. ChemMedChem 2017; 12:1424-1428. [DOI: 10.1002/cmdc.201700387] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Indexed: 11/09/2022]
Affiliation(s)
- Natascha Stergiou
- Johannes Gutenberg University MainzUniversity Medical Center – Institute of Immunology Langenbeckstraße 1, Building 708 55131 Mainz Germany
| | - Markus Glaffig
- Johannes Gutenberg University MainzInstitute of Organic Chemistry Duesbergweg 10–14 55128 Mainz Germany
| | - Helmut Jonuleit
- Johannes Gutenberg University MainzUniversity Medical Center – Dermatology Langenbeckstraße 1, Building 401 55116 Mainz Germany
| | - Edgar Schmitt
- Johannes Gutenberg University MainzUniversity Medical Center – Institute of Immunology Langenbeckstraße 1, Building 708 55131 Mainz Germany
| | - Horst Kunz
- Johannes Gutenberg University MainzInstitute of Organic Chemistry Duesbergweg 10–14 55128 Mainz Germany
| |
Collapse
|
14
|
Glaffig M, Stergiou N, Schmitt E, Kunz H. Immunogenicity of a Fully Synthetic MUC1 Glycopeptide Antitumor Vaccine Enhanced by Poly(I:C) as a TLR3-Activating Adjuvant. ChemMedChem 2017; 12:722-727. [PMID: 28440596 DOI: 10.1002/cmdc.201700254] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Indexed: 11/08/2022]
Abstract
Fully synthetic MUC1 glycopeptide antitumor vaccines have a precisely specified structure and induce a targeted immune response without suppression of the immune response when using an immunogenic carrier protein. However, tumor-associated aberrantly glycosylated MUC1 glycopeptides are endogenous structures, "self-antigens", that exhibit only low immunogenicity. To overcome this obstacle, a fully synthetic MUC1 glycopeptide antitumor vaccine was combined with poly(inosinic acid:cytidylic acid), poly(I:C), as a structurally defined Toll-like receptor 3 (TLR3)-activating adjuvant. This vaccine preparation elicited extraordinary titers of IgG antibodies which strongly bound human breast cancer cells expressing tumor-associated MUC1. Beside the humoral response, the poly(I:C) glycopeptide vaccine induced a pro-inflammatory environment, very important to overcome the immune-suppressive mechanisms, and elicited a strong cellular immune response crucial for tumor elimination.
Collapse
Affiliation(s)
- Markus Glaffig
- Johannes Gutenberg University Mainz, Institute of Organic Chemistry, Duesbergweg 10-14, 55128, Mainz, Germany
| | - Natascha Stergiou
- Johannes Gutenberg University Mainz, University Medical Center, Institute of Immunology, Langenbeckstrasse 1, Geb. 708, 55101, Mainz, Germany
| | - Edgar Schmitt
- Johannes Gutenberg University Mainz, University Medical Center, Institute of Immunology, Langenbeckstrasse 1, Geb. 708, 55101, Mainz, Germany
| | - Horst Kunz
- Johannes Gutenberg University Mainz, Institute of Organic Chemistry, Duesbergweg 10-14, 55128, Mainz, Germany
| |
Collapse
|
15
|
Shimabukuro-Vornhagen A, Schlößer HA, Gryschok L, Malcher J, Wennhold K, Garcia-Marquez M, Herbold T, Neuhaus LS, Becker HJ, Fiedler A, Scherwitz P, Koslowsky T, Hake R, Stippel DL, Hölscher AH, Eidt S, Hallek M, Theurich S, von Bergwelt-Baildon MS. Characterization of tumor-associated B-cell subsets in patients with colorectal cancer. Oncotarget 2015; 5:4651-64. [PMID: 25026291 PMCID: PMC4148088 DOI: 10.18632/oncotarget.1701] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Purpose: A precise understanding of the mechanisms by which human immune cell subsets affect tumor biology will be critical for successful treatment of cancer using immunotherapeutic approaches. Recent evidence suggests that B cells can both promote and inhibit the development and progression of tumors. The aim of this study was to characterize the composition of the B-cell infiltrates in colorectal cancers (CRC) in order to gain further insight into the role of B cells in CRC. Experimental Design: In this study we characterized B-cell subsets in primary tumors (n=38), metastases (n=6) and blood (n=46) of 51 patients with a diagnosis of CRC and blood of 10 healthy controls. B-cell subsets were analyzed by flow cytometry or immunohistochemistry. Results: Peripheral blood of CRC patients contained a higher percentage of memory B cells than that of age-matched healthy controls. Furthermore, the percentage of B cells within tumors was higher than that in the peripheral blood of CRC patients while metastases were typically devoid of tumor-infiltrating B cells. Tumor-associated B cells were enriched for activated and terminally differentiated B cells. Relevant proportions of regulatory B cells could only be detected in advanced cancer and metastases. Conclusion: B cells constitute a significant proportion of the immune infiltrate in CRC. The B-cell infiltrate of primary CRC is characterized by an accumulation of terminally differentiated memory B cells or plasma cells suggestive of a specific immune response against the tumor. However advanced tumors and metastases are also infiltrated by a considerable number of regulatory B cells.
Collapse
Affiliation(s)
- Alexander Shimabukuro-Vornhagen
- Cologne Interventional Immunology, University of Cologne, Germany. Department I of Internal Medicine, University of Cologne, Cologne, Germany. This authors contributed equally to this work
| | - Hans A Schlößer
- Cologne Interventional Immunology, University of Cologne, Germany. Department of General, Visceral and Cancer Surgery, University of Cologne, Germany. This authors contributed equally to this work
| | - Luise Gryschok
- Cologne Interventional Immunology, University of Cologne, Germany
| | - Joke Malcher
- Cologne Interventional Immunology, University of Cologne, Germany
| | - Kerstin Wennhold
- Cologne Interventional Immunology, University of Cologne, Germany
| | | | - Till Herbold
- Cologne Interventional Immunology, University of Cologne, Germany. Department of General, Visceral and Cancer Surgery, University of Cologne, Germany
| | - Laura S Neuhaus
- Cologne Interventional Immunology, University of Cologne, Germany
| | - Hans J Becker
- Cologne Interventional Immunology, University of Cologne, Germany
| | - Anne Fiedler
- Cologne Interventional Immunology, University of Cologne, Germany
| | | | | | - Roland Hake
- Institute of Pathology, St. Elisabeth Hospital, Cologne, Germany
| | - Dirk L Stippel
- Department of General, Visceral and Cancer Surgery, University of Cologne, Germany
| | - Arnulf H Hölscher
- Department of General, Visceral and Cancer Surgery, University of Cologne, Germany
| | - Sebastian Eidt
- Institute of Pathology, St. Elisabeth Hospital, Cologne, Germany
| | - Michael Hallek
- Department I of Internal Medicine, University of Cologne, Cologne, Germany
| | - Sebastian Theurich
- Cologne Interventional Immunology, University of Cologne, Germany. Department I of Internal Medicine, University of Cologne, Cologne, Germany
| | - Michael S von Bergwelt-Baildon
- Cologne Interventional Immunology, University of Cologne, Germany. Department I of Internal Medicine, University of Cologne, Cologne, Germany
| |
Collapse
|
16
|
Ding J, Feng T, Ning Y, Li W, Wu Q, Qian K, Wang Y, Qi C. β-Glucan enhances cytotoxic T lymphocyte responses by activation of human monocyte-derived dendritic cells via the PI3K/AKT pathway. Hum Immunol 2015; 76:146-54. [DOI: 10.1016/j.humimm.2015.01.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 10/10/2014] [Accepted: 01/14/2015] [Indexed: 12/11/2022]
|
17
|
Abdel-Aal ABM, Lakshminarayanan V, Thompson P, Supekar N, Bradley JM, Wolfert MA, Cohen PA, Gendler SJ, Boons GJ. Immune and anticancer responses elicited by fully synthetic aberrantly glycosylated MUC1 tripartite vaccines modified by a TLR2 or TLR9 agonist. Chembiochem 2014; 15:1508-13. [PMID: 24890740 DOI: 10.1002/cbic.201402077] [Citation(s) in RCA: 245] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Indexed: 11/12/2022]
Abstract
The mucin MUC1 is overexpressed and aberrantly glycosylated by many epithelial cancer cells manifested by truncated O-linked saccharides. Although tumor-associated MUC1 has generated considerable attention because of its potential for the development of a therapeutic cancer vaccine, it has been difficult to design constructs that consistently induce cytotoxic T-lymphocytes (CTLs) and ADCC-mediating antibodies specific for the tumor form of MUC1. We have designed, chemically synthesized, and immunologically examined vaccine candidates each composed of a glycopeptide derived from MUC1, a promiscuous Thelper peptide, and a TLR2 (Pam3 CysSK4 ) or TLR9 (CpG-ODN 1826) agonist. It was found that the Pam3 CysSK4 -containing compound elicits more potent antigenic and cellular immune responses, resulting in a therapeutic effect in a mouse model of mammary cancer. It is thus shown, for the first time, that the nature of an inbuilt adjuvant of a tripartite vaccine can significantly impact the quality of immune responses elicited against a tumor-associated glycopeptide. The unique adjuvant properties of Pam3 CysSK4 , which can reduce the suppressive function of regulatory T cells and enhance the cytotoxicity of tumor-specific CTLs, are likely responsible for the superior properties of the vaccine candidate 1.
Collapse
Affiliation(s)
- Abu-Baker M Abdel-Aal
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, GA 30602 (USA)
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Gehrmann U, Näslund TI, Hiltbrunner S, Larssen P, Gabrielsson S. Harnessing the exosome-induced immune response for cancer immunotherapy. Semin Cancer Biol 2014; 28:58-67. [PMID: 24859748 DOI: 10.1016/j.semcancer.2014.05.003] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 05/08/2014] [Indexed: 12/14/2022]
Abstract
In recent years exosomes have emerged as potent stimulators of immune responses and as agents for cancer therapy. Exosomes can carry a broad variety of immunostimulatory molecules depending on the cell of origin and in vitro culture conditions. Dendritic cell-derived exosomes (dexosomes) have been shown to carry NK cell activating ligands and can be loaded with antigen to activate invariant NKT cells and to induce antigen-specific T and B cell responses. Dexosomes have been investigated as therapeutic agents against cancer in two phase I clinical trials, with a phase II clinical trial currently ongoing. Dexosomes were well tolerated but therapeutic success and immune activation were limited. Several reports suggest that multiple factors need to be considered in order to improve exosomal immunogenicity for cancer immunotherapy. These include antigen-loading strategies, exosome composition and exosomal trafficking in vivo. Hence, a better understanding of how to engineer and deliver exosomes to specific cells is crucial to generate strong immune responses and to improve the immunotherapeutic potential of exosomes.
Collapse
Affiliation(s)
- Ulf Gehrmann
- Translational Immunology Unit, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Tanja I Näslund
- Translational Immunology Unit, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Stefanie Hiltbrunner
- Translational Immunology Unit, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Pia Larssen
- Translational Immunology Unit, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Susanne Gabrielsson
- Translational Immunology Unit, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
19
|
Chen X, Chang CH, Goldenberg DM. Novel strategies for improved cancer vaccines. Expert Rev Vaccines 2014; 8:567-76. [DOI: 10.1586/erv.09.11] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
20
|
Rodriguez PC, Sanchez B. Challenges and opportunities for cancer vaccines in the current NSCLC clinical scenario. Curr Top Med Chem 2013; 13:2551-61. [PMID: 24066886 PMCID: PMC4104452 DOI: 10.2174/15680266113136660182] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 01/25/2013] [Accepted: 08/02/2013] [Indexed: 12/20/2022]
Abstract
This review is aimed to focus on NSCLC as an emerging and promising model for active immunotherapy and the challenges for its inclusion in the current clinical scenario. Cancer vaccines for NSCLC have been focused as a therapeutic option based on the identification of a tumor hallmark and the active immunization with the related molecules that triggers cellular and/or humoral responses that consequently destroy or delay the rate of malignant progression. This therapeutic intervention in an established disease state has been aimed to impact into prolonging patient´s survival with ethically accepted quality of life. Understanding of relationship between structure and function in cancer vaccines is essential to interpret their opportunities to impact into prolonging survival and increasing quality of life in cancer patients. It is widely accepted that the failure of the cancer vaccines in the NSCLC scenario is related with its introduction in the advanced disease stages and poor performance status of the patients due to the combination of the tumor induced immunosuppression with the immune senescence. Despite first, second and emerging third line of onco-specific treatments the life expectancy for NSCLC patients diagnosed at advanced stages is surrounding the 12 months of median survival and in facts the today real circumstances are extremely demanding for the success inclusion of cancer vaccines as therapeutic choice in the clinical scenario. The kinetics of the active immunizations encompasses a sequential cascade of clinical endpoints: starting by the activation of the immune system, followed by the antitumor response and finalizing with the consequential impact on patients’ overall survival. Today this cascade of clinical endpoints is the backbone for active immunization assessment and moreover the concept of cancer vaccines, applied in the NSCLC setting, is just evolving as a complex therapeutic strategy, in which the opportunities for cancer vaccines start from the selection of the target cancer hallmark, followed by the vaccine formulation and its platforms for immune potentiating, also cover the successful insertion in the standard of care, the chronic administration beyond progression disease, the personalization based on predictors of response and the potential combination with other targeted therapies.
Collapse
Affiliation(s)
- Pedro C Rodriguez
- Center of Molecular Immunology, Clinical Research Direction, 216 and 15, Playa, P.O.Box: 16040, Havana 11600, Cuba.
| | | |
Collapse
|
21
|
Ma Y, Xiang D, Sun J, Ding C, Liu M, Hu X, Li G, Kloecker G, Zhang HG, Yan J. Targeting of antigens to B lymphocytes via CD19 as a means for tumor vaccine development. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2013; 190:5588-99. [PMID: 23630363 PMCID: PMC3660458 DOI: 10.4049/jimmunol.1203216] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Ab therapy against surface Ags on tumor cells has demonstrated significant efficacy for some cancers. However, it is costly and patients frequently develop acquired resistance over time. In cases of Ab therapy resistance, T cell responses have been shown to be essential in controlling disease progression. Thus, vaccination that generates a sustained Ab response as well as a T cell response may be more effective and economical. In this article, we have developed a vaccination strategy by targeting protein Ags to B cells via a CD19 single-chain variable fragment miniAb. Using the tumor-associated Ag her-2/neu extracellular domain, we showed that the coengagement of CD19 and BCR induced full B cell activation to produce a high titer of Abs and enhanced CD4 Th2 response and CD8 T cell activation and differentiation. These Abs competitively inhibited humanized her-2/neu Ab binding and were capable of activating the complement and inhibiting human breast cancer growth in vitro. Therapeutic efficacy was demonstrated in vivo using murine mammary carcinoma models. Furthermore, four different extracellular domains of her-2/neu could be targeted to B cells to generate Abs against particular domains with different antitumor properties. This approach may offer a new avenue for vaccine development with significantly lower cost, which may be of use not only for cancer therapy but also for infectious agents.
Collapse
Affiliation(s)
- Yunfeng Ma
- Tumor Immunobiology Program, James Graham Brown Cancer Center, University of Louisville School of Medicine, U.S.A
- Division of Hematology/Oncology, Department of Medicine, University of Louisville School of Medicine, U.S.A
| | - Dong Xiang
- Tumor Immunobiology Program, James Graham Brown Cancer Center, University of Louisville School of Medicine, U.S.A
- Division of Hematology/Oncology, Department of Medicine, University of Louisville School of Medicine, U.S.A
- Division of Hematology and Medical Oncology, Ellis Fischel Cancer Center, University of Missouri School of Medicine, U.S.A
| | - Jinwen Sun
- Tumor Immunobiology Program, James Graham Brown Cancer Center, University of Louisville School of Medicine, U.S.A
- Department of General Surgery & Surgical Oncology, Beijing Meitan General Hospital, P.R. China
| | - Chuanlin Ding
- Tumor Immunobiology Program, James Graham Brown Cancer Center, University of Louisville School of Medicine, U.S.A
- Division of Hematology/Oncology, Department of Medicine, University of Louisville School of Medicine, U.S.A
| | - Min Liu
- Tumor Immunobiology Program, James Graham Brown Cancer Center, University of Louisville School of Medicine, U.S.A
- Division of Hematology/Oncology, Department of Medicine, University of Louisville School of Medicine, U.S.A
| | - Xiaoling Hu
- Tumor Immunobiology Program, James Graham Brown Cancer Center, University of Louisville School of Medicine, U.S.A
| | - Guoxin Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Goetz Kloecker
- Division of Hematology/Oncology, Department of Medicine, University of Louisville School of Medicine, U.S.A
| | - Huang-ge Zhang
- Tumor Immunobiology Program, James Graham Brown Cancer Center, University of Louisville School of Medicine, U.S.A
- Department of Microbiology and Immunology, University of Louisville School of Medicine, U.S.A
| | - Jun Yan
- Tumor Immunobiology Program, James Graham Brown Cancer Center, University of Louisville School of Medicine, U.S.A
- Division of Hematology/Oncology, Department of Medicine, University of Louisville School of Medicine, U.S.A
- Department of Microbiology and Immunology, University of Louisville School of Medicine, U.S.A
| |
Collapse
|
22
|
A vaccine directed to B cells and produced by cell-free protein synthesis generates potent antilymphoma immunity. Proc Natl Acad Sci U S A 2012; 109:14526-31. [PMID: 22875703 DOI: 10.1073/pnas.1211018109] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Clinical studies of idiotype (Id) vaccination in patients with lymphoma have established a correlation between the induced anti-Id antibody responses and favorable clinical outcomes. To streamline the production of an Id vaccine, we engineered a small diabody (Db) molecule containing both a B-cell-targeting moiety (anti-CD19) and a lymphoma Id. This molecule (αCD19-Id) was designed to penetrate lymph nodes and bind to noncognate B cells to form an antigen presentation array. Indeed, the αCD19-Id molecule accumulated on B cells in vivo after s.c. administration. These noncognate B cells, decorated with the diabody, could then stimulate the more rare Id-specific B cells. Peptide epitopes present in the diabody linker augmented the response by activating CD4(+) helper T cells. Consequently, the αCD19-Id molecule induced a robust Id-specific antibody response and protected animals from tumor challenge. Such diabodies are produced in a cell-free protein expression system within hours of amplification of the specific Ig genes from the B-cell tumor. This customized product can now be available to vaccinate patients before they receive other, potentially immunosuppressive, therapies.
Collapse
|
23
|
Pichinuk E, Benhar I, Jacobi O, Chalik M, Weiss L, Ziv R, Sympson C, Karwa A, Smorodinsky NI, Rubinstein DB, Wreschner DH. Antibody targeting of cell-bound MUC1 SEA domain kills tumor cells. Cancer Res 2012; 72:3324-36. [PMID: 22507854 DOI: 10.1158/0008-5472.can-12-0067] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The cell-surface glycoprotein MUC1 is a particularly appealing target for antibody targeting, being selectively overexpressed in many types of cancers and a high proportion of cancer stem-like cells. However the occurrence of MUC1 cleavage, which leads to the release of the extracellular α subunit into the circulation where it can sequester many anti-MUC1 antibodies, renders the target problematic to some degree. To address this issue, we generated a set of unique MUC1 monoclonal antibodies that target a region termed the SEA domain that remains tethered to the cell surface after MUC1 cleavage. In breast cancer cell populations, these antibodies bound the cancer cells with high picomolar affinity. Starting with a partially humanized antibody, DMB5F3, we created a recombinant chimeric antibody that bound a panel of MUC1+ cancer cells with higher affinities relative to cetuximab (anti-EGFR1) or tratuzumab (anti-erbB2) control antibodies. DMB5F3 internalization from the cell surface occurred in an efficient temperature-dependent manner. Linkage to toxin rendered these DMB5F3 antibodies to be cytotoxic against MUC1+ cancer cells at low picomolar concentrations. Our findings show that high-affinity antibodies to cell-bound MUC1 SEA domain exert specific cytotoxicity against cancer cells, and they point to the SEA domain as a potential immunogen to generate MUC1 vaccines.
Collapse
Affiliation(s)
- Edward Pichinuk
- Department of Cell Research and Immunology, Tel Aviv University, Ramat Aviv, Israel
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Speetjens FM, Zeestraten ECM, Kuppen PJK, Melief CJM, van der Burg SH. Colorectal cancer vaccines in clinical trials. Expert Rev Vaccines 2011; 10:899-921. [PMID: 21692708 DOI: 10.1586/erv.11.63] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
This article elucidates current strategies of active immunotherapy for colorectal cancer patients with a focus on T-cell mediated immunotherapy. Poor prognosis of especially stage III and IV colorectal cancer patients emphasizes the need for advanced therapeutic intervention. Here, we refer to clinical trials using either tumor cell-derived vaccines or tumor antigen vaccines with a special interest on safety, induced immune responses, clinical benefit and efforts to improve the clinical impact of these vaccines in the context of colorectal cancer treatment.
Collapse
Affiliation(s)
- Frank M Speetjens
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | |
Collapse
|
25
|
Differential pathways regulating innate and adaptive antitumor immune responses by particulate and soluble yeast-derived β-glucans. Blood 2011; 117:6825-36. [PMID: 21531981 DOI: 10.1182/blood-2011-02-339812] [Citation(s) in RCA: 183] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
β-glucans have been reported to function as a potent adjuvant to stimulate innate and adaptive immune responses. However, β-glucans from different sources are differential in their structure, conformation, and thus biologic activity. Different preparations of β-glucans, soluble versus particulate, further complicate their mechanism of action. Here we show that yeast-derived particulate β-glucan activated dendritic cells (DCs) and macrophages via a C-type lectin receptor dectin-1 pathway. Activated DCs by particulate β-glucan promoted Th1 and cytotoxic T-lymphocyte priming and differentiation in vitro. Treatment of orally administered yeast-derived particulate β-glucan elicited potent antitumor immune responses and drastically down-regulated immunosuppressive cells, leading to the delayed tumor progression. Deficiency of the dectin-1 receptor completely abrogated particulate β-glucan-mediated antitumor effects. In contrast, yeast-derived soluble β-glucan bound to DCs and macrophages independent of the dectin-1 receptor and did not activate DCs. Soluble β-glucan alone had no therapeutic effect but significantly augmented antitumor monoclonal antibody-mediated therapeutic efficacy via a complement activation pathway but independent of dectin-1 receptor. These findings reveal the importance of different preparations of β-glucans in the adjuvant therapy and allow for the rational design of immunotherapeutic protocols usable in clinical trials.
Collapse
|
26
|
Vallhov H, Gutzeit C, Johansson SM, Nagy N, Paul M, Li Q, Friend S, George TC, Klein E, Scheynius A, Gabrielsson S. Exosomes Containing Glycoprotein 350 Released by EBV-Transformed B Cells Selectively Target B Cells through CD21 and Block EBV Infection In Vitro. THE JOURNAL OF IMMUNOLOGY 2010; 186:73-82. [DOI: 10.4049/jimmunol.1001145] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
27
|
Abstract
The overexpression and aberrant glycosylation of MUC1 is associated with a wide variety of cancers, making it an ideal target for immunotherapeutic strategies. This review highlights the main avenues of research in this field, focusing on adenocarcinomas, from the preclinical to clinical; the problems and possible solutions associated with each approach; and speculates on the direction of MUC1 immunotherapeutic research over the next 5-10 years.
Collapse
Affiliation(s)
- Richard E Beatson
- Breast Cancer Biology Group, King's College London, Guy's Hospital, London SE1 9RT, UK
| | | | | |
Collapse
|
28
|
Liu Y, He J, Xie X, Su G, Teitz-Tennenbaum S, Sabel MS, Lubman DM. Serum autoantibody profiling using a natural glycoprotein microarray for the prognosis of early melanoma. J Proteome Res 2010; 9:6044-51. [PMID: 20879797 DOI: 10.1021/pr100856k] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The poor prognosis of melanoma and the high cost of lymph node biopsy for melanoma patients have led to an urgent need for the discovery of convenient and accurate prognostic indicators. Here, we have developed a natural glycoprotein microarray to discover serum autoantibodies to distinguish between patients with node negative melanoma and node positive melanoma. Dual-lectin affinity chromatography was used to extract glycoproteins from a melanoma cell line. Liquid-based reverse phase separation and microarray platforms were then applied to separate and spot these natural proteins on nitrocellulose slides. The serum autoantibodies were investigated by exposing these proteins to sera from 43 patients that have already been diagnosed to have different stages of early melanoma. The combination of 9 fractions provides a 55% sensitivity with 100% specificity for the detection of node positive against node negative and a 62% sensitivity with 100% specificity for the detection of node negative against node positive. Recombinant proteins were used to confirm the results using a sample set with 79 patients with diagnosed melanoma. The response of sera against recombinant 94 kD glucose-regulated protein (GRP94), acid ceramidase (ASAH1), cathepsin D (CTSD), and lactate dehydrogenase B (LDHB) shared a similar pattern to the fractions where they were identified. The glycoarray platform provides a convenient and highly reproducible method to profile autoantibodies that could be used as serum biomarkers for prognosis of melanoma.
Collapse
Affiliation(s)
- Yashu Liu
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Li B, Cai Y, Qi C, Hansen R, Ding C, Mitchell TC, Yan J. Orally administered particulate beta-glucan modulates tumor-capturing dendritic cells and improves antitumor T-cell responses in cancer. Clin Cancer Res 2010; 16:5153-64. [PMID: 20855461 DOI: 10.1158/1078-0432.ccr-10-0820] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The beneficial properties of β-glucans have been recognized for centuries. Their proposed mechanisms of action in cancer therapy occur via stimulation of macrophages and priming of innate neutrophil complement receptor 3 for eliciting complement receptor 3-dependent cellular cytotoxicity of iC3b-opsonized tumor cells. The current study is to investigate whether β-glucan therapy has any effect on antitumor adaptive T-cell responses. EXPERIMENTAL DESIGN We first examined the trafficking of orally administered particulate yeast-derived β-glucan and its interaction with dendritic cells (DC) that captured tumor materials. Antigen-specific T cells were adoptively transferred into recipient mice to determine whether oral β-glucan therapy induces augmented T-cell responses. Lewis lung carcinoma and RAM-S lymphoma models were used to test oral β-glucan therapeutic effect. Further mechanistic studies including tumor-infiltrating T cells and cytokine profiles within the tumor milieu were determined. RESULTS Orally administered particulate β-glucan trafficked into spleen and lymph nodes and activated DCs that captured dying tumor cells in vivo, leading to the expansion and activation of antigen-specific CD4 and CD8 T cells. In addition, IFN-γ production of tumor-infiltrating T cells and CTL responses were significantly enhanced on β-glucan treatment, which ultimately resulted in significantly reduced tumor burden. Moreover, β-glucan-treated tumors had significantly more DC infiltration with the activated phenotype and significant levels of Th1-biased cytokines within the tumor microenvironment. CONCLUSIONS These data highlight the ability of yeast-derived β-glucan to bridge innate and adaptive antitumor immunity and suggest that it can be used as an adjuvant for tumor immunotherapy.
Collapse
Affiliation(s)
- Bing Li
- Tumor Immunobiology Program of the James Graham Brown Cancer Center, Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky 40202, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
Toll-like receptors (TLRs) are part of the innate immune system, and they belong to the pattern recognition receptors (PRR) family. The PRR family is designed to recognize and bind conserved pathogen-associated molecular patterns, which are not generated by the host and are restricted and essential to micro-organisms. TLR9, which recognizes unmethylated CpG (cytosine guanosine dinucleotide), is a very promising target for therapeutic activation. Stimulation of TLR9 activates human plasmacytoid dendritic cells and B cells, and results in potent T helper-1 (T(h)1)-type immune responses and antitumor responses in mouse tumor models and in patients. Several pharmaceutical companies, such as Pfizer, Idera, and Dynavax, are developing CpG oligodeoxynucleotides (ODNs) for the treatment of cancer, along with other conditions, such as infections and allergy. CpG ODNs have shown promising results as vaccine adjuvants and in combination with cancer immunotherapy. Several TLR9 agonists are being developed and have entered clinical trials to evaluate their safety and efficacy for the treatment of several hematopoietic and solid tumors. In this review, we discuss the use of CpG ODNs in several phase I and II clinical trials for the treatment of NHL, renal cell carcinoma, melanoma, and non-small cell lung cancer, either alone or in combination with other agents.
Collapse
Affiliation(s)
- Yanal M Murad
- Duke University Medical Center, Department of Surgery, Program in Molecular Therapeutics, Comprehensive Cancer Center, 401 MSRB, Research Drive, Durham, NC 27710, USA
| | | |
Collapse
|
31
|
Ryan SO, Turner MS, Gariépy J, Finn OJ. Tumor antigen epitopes interpreted by the immune system as self or abnormal-self differentially affect cancer vaccine responses. Cancer Res 2010; 70:5788-96. [PMID: 20587526 DOI: 10.1158/0008-5472.can-09-4519] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Epitope selection is an important consideration in the design of cancer vaccines, but factors affecting selection are not fully understood. We compared the immune responses to peptides and glycopeptides from the common human tumor antigen MUC1, a mucin that is coated with O-linked carbohydrates in its variable number of tandem repeats (VNTR) region. MUC1 expressed on tumor cells is characteristically underglycosylated, creating peptide and glycopeptide neoepitopes that are recognized by the immune system. The response to VNTR peptides is weaker in MUC1-transgenic mice (MUC1-Tg mice) than in wild-type (WT) mice, whereas the response to VNTR glycopeptides is equally strong in the two strains. Thus, glycopeptides seem to be recognized as foreign, whereas peptides, although immunogenic, are perceived as self. To explore this further, we generated MUC1 peptide- and glycopeptide-specific T-cell receptor transgenic mice and studied the function of their CD4 T cells when adoptively transferred into MUC1-Tg or WT mice. Peptide-specific T-cell precursors were not centrally deleted in MUC1-Tg mice and did not acquire a T regulatory phenotype. However, their response to the cognate peptide was reduced in MUC1-Tg mice compared with WT mice. In contrast, glycopeptide-specific CD4 T cells responded equally well in the two hosts and, when simultaneously activated, also enhanced the peptide-specific T-cell responses. Our data show that the immune system differentially recognizes various epitopes of tumor-associated antigens either as self or as foreign, and this controls the strength of antitumor immunity. This represents an important consideration for designing safe and effective cancer vaccines.
Collapse
Affiliation(s)
- Sean O Ryan
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15262, USA
| | | | | | | |
Collapse
|
32
|
Bridging innate and adaptive antitumor immunity targeting glycans. J Biomed Biotechnol 2010; 2010:354068. [PMID: 20617150 PMCID: PMC2896669 DOI: 10.1155/2010/354068] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2010] [Accepted: 04/22/2010] [Indexed: 01/12/2023] Open
Abstract
Effective immunotherapy for cancer depends on cellular responses to tumor antigens. The role of major histocompatibility complex (MHC) in T-cell recognition and T-cell receptor repertoire selection has become a central tenet in immunology. Structurally, this does not contradict earlier findings that T-cells can differentiate between small hapten structures like simple glycans. Understanding T-cell recognition of antigens as defined genetically by MHC and combinatorially by T cell receptors led to the “altered self” hypothesis. This notion reflects a more fundamental principle underlying immune surveillance and integrating evolutionarily and mechanistically diverse elements of the immune system. Danger associated molecular patterns, including those generated by glycan remodeling, represent an instance of altered self. A prominent example is the modification of the tumor-associated antigen MUC1. Similar examples emphasize glycan reactivity patterns of antigen receptors as a phenomenon bridging innate and adaptive but also humoral and cellular immunity and providing templates for immunotherapies.
Collapse
|
33
|
Ryan SO, Vlad AM, Islam K, Gariépy J, Finn OJ. Tumor-associated MUC1 glycopeptide epitopes are not subject to self-tolerance and improve responses to MUC1 peptide epitopes in MUC1 transgenic mice. Biol Chem 2009; 390:611-8. [PMID: 19426130 DOI: 10.1515/bc.2009.070] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Human adenocarcinomas overexpress a hypoglycosylated, tumor-associated form of the mucin-like glycoprotein MUC1 containing abnormal mono- and disaccharide antigens, such as Tn, sialyl-Tn, and TF, as well as stretches of unglycosylated protein backbone in the variable number of tandem repeats (VNTR) region. Both peptide and glycopeptide epitopes generated from the VNTR are candidates for cancer vaccines and we performed experiments to evaluate their relative potential to elicit tumor-MUC1-specific immunity. We show here that immunization with the 100 amino acid-long VNTR peptide (MUC1p) elicits weaker responses in MUC1 transgenic mice compared to wild type mice suggesting self-tolerance. In contrast, when glycosylated with tumor-associated Tn antigen (GalNAc-O-S/T), TnMUC1 induces glycopeptide-specific T cell and antibody responses in both strains of mice and helps enhance responses to MUC1p in MUC1 transgenic mice. Using newly derived MUC1-specific mouse T cell hybridomas we show that the only antigen-presenting cells able to cross-present TnMUC1 glycopeptide are dendritic cells (DCs). This is likely due to their exclusive expression of receptors capable of binding TnMUC1. We conclude that MUC1 glycopeptides induce stronger immunity in MUC1-Tg mice because they are recognized as 'foreign' rather than ;self' and because they are cross-presented preferentially by DCs.
Collapse
Affiliation(s)
- Sean O Ryan
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15232, USA
| | | | | | | | | |
Collapse
|