1
|
Poudel K, Vithiananthan T, Kim JO, Tsao H. Recent progress in cancer vaccines and nanovaccines. Biomaterials 2025; 314:122856. [PMID: 39366184 DOI: 10.1016/j.biomaterials.2024.122856] [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: 04/25/2024] [Revised: 09/03/2024] [Accepted: 09/26/2024] [Indexed: 10/06/2024]
Abstract
Vaccine science, nanotechnology, and immunotherapy are at the forefront of cancer treatment strategies, each offering significant potential for enhancing tumor-specific immunity and establishing long-lasting immune memory to prevent tumor recurrence. Despite the promise of these personalized and precision-based anti-cancer approaches, challenges such as immunosuppression, suboptimal immune activation, and T-cell exhaustion continue to hinder their effectiveness. The limited clinical success of cancer vaccines often stems from difficulties in identifying effective antigens, efficiently targeting immune cells, lymphoid organs, and the tumor microenvironment, overcoming immune evasion, enhancing immunogenicity, and avoiding lysosomal degradation. However, numerous studies have demonstrated that integrating nanotechnology with immunotherapeutic strategies in vaccine development can overcome these challenges, leading to potent antitumor immune responses and significant progress in the field. This review highlights the critical components of cancer vaccine and nanovaccine strategies for immunomodulatory antitumor therapy. It covers general vaccine strategies, types of vaccines, antigen forms, nanovaccine platforms, challenges faced, potential solutions, and key findings from preclinical and clinical studies, along with future perspectives. To fully unlock the potential of cancer vaccines and nanovaccines, precise immunological monitoring during early-phase trials is essential. This approach will help identify and address obstacles, ultimately expanding the available options for patients who are resistant to conventional cancer immunotherapies.
Collapse
Affiliation(s)
- Kishwor Poudel
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tulasi Vithiananthan
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Hensin Tsao
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Sun D, Yu L, Wang G, Xu Y, Wang P, Wang N, Wu Z, Zhang G, Zhang J, Zhang Y, Tian G, Wei P. Rationally designed catalytic nanoplatform for enhanced chemoimmunotherapy via deploying endogenous plus exogenous copper and remodeling tumor microenvironment. J Nanobiotechnology 2024; 22:551. [PMID: 39252079 PMCID: PMC11385821 DOI: 10.1186/s12951-024-02696-x] [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: 02/22/2024] [Accepted: 07/03/2024] [Indexed: 09/11/2024] Open
Abstract
Chemodynamic therapy represents a novel tumor therapeutic modality via triggering catalytic reactions in tumors to yield highly toxic reactive oxygen species (ROS). Nevertheless, low efficiency catalytic ability, potential systemic toxicity and inefficient tumor targeting, have hindered the efficacy of chemodynamic therapy. Herein, a rationally designed catalytic nanoplatform, composed of folate acid conjugated liposomes loaded with copper peroxide (CP) and chloroquine (CQ; a clinical drug) (denoted as CC@LPF), could power maximal tumor cytotoxicity, mechanistically via maneuvering endogenous and exogenous copper for a highly efficient catalytic reaction. Despite a massive autophagosome accumulation elicited by CP-powered autophagic initiation and CQ-induced autolysosomal blockage, the robust ROS, but not aberrant autophagy, underlies the synergistic tumor inhibition. Otherwise, this combined mode also elicits an early onset, above all, long-term high-level existence of immunogenic cell death markers, associated with ROS and aberrant autophagy -triggered endoplasmic reticulum stress. Besides, CC@LPF, with tumor targeting capability and selective tumor cytotoxicity, could elicit intratumor dendritic cells (mainly attributed to CQ) and tumor infiltrating CD8+ T cells, upon combining with PD-L1 therapeutic antibody, further induce significant anti-tumor effect. Collectively, the rationally designed nanoplatform, CC@LPF, could enhance tumor chemoimmunotherapy via deploying endogenous plus exogenous copper and remodeling tumor microenvironment.
Collapse
Affiliation(s)
- Daxi Sun
- School of Pharmacy, Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, 264003, China
| | - Liting Yu
- School of Pharmacy, Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, 264003, China
| | - Gang Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Yuxue Xu
- School of Pharmacy, Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, 264003, China
| | - Peng Wang
- School of Pharmacy, Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, 264003, China
| | - Ningning Wang
- School of Pharmacy, Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, 264003, China
| | - Zhengyan Wu
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, P.R. China.
- University of Science and Technology of China, Hefei, 230026, P.R. China.
| | - Guilong Zhang
- School of Pharmacy, Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, 264003, China
| | - Jia Zhang
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, P.R. China
- University of Science and Technology of China, Hefei, 230026, P.R. China
| | - Yunjiao Zhang
- The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Geng Tian
- School of Pharmacy, Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, 264003, China.
| | - Pengfei Wei
- School of Pharmacy, Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, 264003, China.
| |
Collapse
|
3
|
Borrajo ML, Lou G, Anthiya S, Lapuhs P, Álvarez DM, Tobío A, Loza MI, Vidal A, Alonso MJ. Nanoemulsions and nanocapsules as carriers for the development of intranasal mRNA vaccines. Drug Deliv Transl Res 2024; 14:2046-2061. [PMID: 38811465 PMCID: PMC11208213 DOI: 10.1007/s13346-024-01635-5] [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] [Accepted: 05/21/2024] [Indexed: 05/31/2024]
Abstract
The global emergency of coronavirus disease 2019 (COVID-19) has spurred extensive worldwide efforts to develop vaccines for protection against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Our contribution to this global endeavor involved the development of a diverse library of nanocarriers, as alternatives to lipid nanoparticles (LNPs), including nanoemulsions (NEs) and nanocapsules (NCs), with the aim of protecting and delivering messenger ribonucleic acid (mRNA) for nasal vaccination purposes. A wide range of prototypes underwent rigorous screening through a series of in vitro and in vivo experiments, encompassing assessments of cellular transfection, cytotoxicity, and intramuscular administration of a model mRNA for protein translation. As a result, two promising candidates were identified for nasal administration. One of them was a NE incorporating a combination of an ionizable lipid (C12-200) and cationic lipid (DOTAP), both intended to condense mRNA, along with DOPE, which is known to facilitate endosomal escape. This NE exhibited a size of 120 nm and a highly positive surface charge (+ 50 mV). Another candidate was an NC formulation comprising the same components and endowed with a dextran sulfate shell. This formulation showed a size of 130 nm and a moderate negative surface charge (-16 mV). Upon intranasal administration of mRNA encoding for ovalbumin (mOVA) associated with optimized versions of the said NE and NCs, a robust antigen-specific CD8 + T cell response was observed. These findings underscore the potential of NEs and polymeric NCs in advancing mRNA vaccine development for combating infectious diseases.
Collapse
Affiliation(s)
- Mireya L Borrajo
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University de Santiago de Compostela, Av. Barcelona s/n, Campus Vida, Santiago de Compostela, 15782, Spain
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
- IDIS Research Institute, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Gustavo Lou
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University de Santiago de Compostela, Av. Barcelona s/n, Campus Vida, Santiago de Compostela, 15782, Spain
- IDIS Research Institute, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Shubaash Anthiya
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University de Santiago de Compostela, Av. Barcelona s/n, Campus Vida, Santiago de Compostela, 15782, Spain
- IDIS Research Institute, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Philipp Lapuhs
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University de Santiago de Compostela, Av. Barcelona s/n, Campus Vida, Santiago de Compostela, 15782, Spain
- IDIS Research Institute, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - David Moreira Álvarez
- Biofarma Research Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Av. Barcelona s/n, Campus Vida, Santiago de Compostela, 15782, Spain
| | - Araceli Tobío
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University de Santiago de Compostela, Av. Barcelona s/n, Campus Vida, Santiago de Compostela, 15782, Spain
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
- IDIS Research Institute, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - María Isabel Loza
- Biofarma Research Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Av. Barcelona s/n, Campus Vida, Santiago de Compostela, 15782, Spain
| | - Anxo Vidal
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University de Santiago de Compostela, Av. Barcelona s/n, Campus Vida, Santiago de Compostela, 15782, Spain
- IDIS Research Institute, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - María José Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University de Santiago de Compostela, Av. Barcelona s/n, Campus Vida, Santiago de Compostela, 15782, Spain.
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain.
- IDIS Research Institute, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain.
| |
Collapse
|
4
|
Yang J, Rong SJ, Zhou HF, Yang C, Sun F, Li JY. Lysosomal control of dendritic cell function. J Leukoc Biol 2023; 114:518-531. [PMID: 37774493 DOI: 10.1093/jleuko/qiad117] [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: 04/26/2023] [Revised: 06/22/2023] [Accepted: 09/08/2023] [Indexed: 10/01/2023] Open
Abstract
Lysosomal compartments undergo extensive remodeling during dendritic cell (DC) activation to meet the dynamic functional requirements of DCs. Instead of being regarded as stationary and digestive organelles, recent studies have increasingly appreciated the versatile roles of lysosomes in regulating key aspects of DC biology. Lysosomes actively control DC motility by linking calcium efflux to the actomyosin contraction, while enhanced DC lysosomal membrane permeability contributes to the inflammasome activation. Besides, lysosomes provide a platform for the transduction of innate immune signaling and the intricate host-pathogen interplay. Lysosomes and lysosome-associated structures are also critically engaged in antigen presentation and cross-presentation processes, which are pivotal for the induction of antigen-specific adaptive immune response. Through the current review, we emphasize that lysosome targeting strategies serve as vital DC-based immunotherapies in fighting against tumor, infectious diseases, and autoinflammatory disorders.
Collapse
Affiliation(s)
- Jia Yang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue No.1277, 430000, Wuhan, China
| | - Shan-Jie Rong
- Department of Respiratory and Critical Care Medicine, Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Jiefang Avenue No.1095, 430000, Wuhan, China
| | - Hai-Feng Zhou
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue No.1277, 430000, Wuhan, China
| | - Chao Yang
- Department of Gerontology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Ling Jiaohu Road No.11, 430000, Wuhan, China
| | - Fei Sun
- Department of Respiratory and Critical Care Medicine, Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Jiefang Avenue No.1095, 430000, Wuhan, China
| | - Jun-Yi Li
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue No.1277, 430000, Wuhan, China
| |
Collapse
|
5
|
Beier UH, Baker DJ, Baur JA. Thermogenic T cells: a cell therapy for obesity? Am J Physiol Cell Physiol 2022; 322:C1085-C1094. [PMID: 35476503 PMCID: PMC9169824 DOI: 10.1152/ajpcell.00034.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/20/2022] [Accepted: 04/22/2022] [Indexed: 11/22/2022]
Abstract
Obesity is a widespread public health problem with profound medical consequences and its burden is increasing worldwide. Obesity causes significant morbidity and mortality and is associated with conditions including cardiovascular disease and diabetes mellitus. Conventional treatment options are insufficient, or in the case of bariatric surgery, quite invasive. The etiology of obesity is complex, but at its core is often a caloric imbalance with an inability to burn off enough calories to exceed caloric intake, resulting in storage. Interventions such as dieting often lead to decreased resting energy expenditure (REE), with a rebound in weight ("yo-yo effect" or weight cycling). Strategies that increase REE are attractive treatment options. Brown fat tissue engages in nonshivering thermogenesis whereby mitochondrial respiration is uncoupled from ATP production, increasing REE. Medications that replicate brown fat metabolism by mitochondrial uncoupling (e.g., 2,4-dinitrophenol) effectively promote weight loss but are limited by toxicity to a narrow therapeutic range. This review explores the possibility of a new therapeutic approach to engineer autologous T cells into acquiring a thermogenic phenotype like brown fat. Engineered autologous T cells have been used successfully for years in the treatment of cancers (chimeric antigen receptor T cells), and the principle of engineering T cells ex vivo and transferring them back to the patient is established. Engineering T cells to acquire a brown fat-like metabolism could increase REE without the risks of pharmacological mitochondrial uncoupling. These thermogenic T cells may increase basal metabolic rate and are therefore a potentially novel therapeutic strategy for obesity.
Collapse
Affiliation(s)
- Ulf H Beier
- Janssen Research and Development, Spring House, Pennsylvania
| | - Daniel J Baker
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
- Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Medicine, Perelman School of Medicine, Cardiovascular Institute and Institute of Diabetes, Obesity, and Metabolism, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Joseph A Baur
- Department of Physiology, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
6
|
Qin L, Zhang H, Zhou Y, Umeshappa CS, Gao H. Nanovaccine-Based Strategies to Overcome Challenges in the Whole Vaccination Cascade for Tumor Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2006000. [PMID: 33768693 DOI: 10.1002/smll.202006000] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/26/2020] [Indexed: 06/12/2023]
Abstract
Nanovaccine-based immunotherapy (NBI) has received greater attention recently for its potential to prime tumor-specific immunity and establish a long-term immune memory that prevents tumor recurrence. Despite encouraging results in the recent studies, there are still numerous challenges to be tackled for eliciting potent antitumor immunity using NBI strategies. Based on the principles that govern immune response, here it is proposed that these challenges need to be addressed at the five critical cascading events: Loading tumor-specific antigens by nanoscale drug delivery systems (L); Draining tumor antigens to lymph nodes (D); Internalization by dendritic cells (DCs) (I); Maturation of DCs by costimulatory signaling (M); and Presenting tumor-peptide-major histocompatibility complexes to T cells (P) (LDIMP cascade in short). This review provides a detailed and objective overview of emerging NBI strategies to improve the efficacy of nanovaccines in each step of the LDIMP cascade. It is concluded that the balance between each step must be optimized by delicate designing and modification of nanovaccines and by combining with complementary approaches to provide a synergistic immunity in the fight against cancer.
Collapse
Affiliation(s)
- Lin Qin
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
- Chongqing Vocational College of Transportation, Chongqing, 400715, China
| | - Huilin Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yang Zhou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Channakeshava Sokke Umeshappa
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
7
|
Martinez GP, Zabaleta ME, Di Giulio C, Charris JE, Mijares MR. The Role of Chloroquine and Hydroxychloroquine in Immune Regulation and Diseases. Curr Pharm Des 2020; 26:4467-4485. [DOI: 10.2174/1381612826666200707132920] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/27/2020] [Indexed: 02/06/2023]
Abstract
Chloroquine (CQ) and hydroxychloroquine (HCQ) are derivatives of the heterocyclic aromatic compound
quinoline. These economical compounds have been used as antimalarial agents for many years. Currently,
they are used as monotherapy or in conjunction with other therapies for the treatment of autoimmune diseases
such as systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), Sjögren's syndrome (SS) and antiphospholipid
antibody syndrome (APS). Based on its effects on the modulation of the autophagy process, various
clinical studies suggest that CQ and HCQ could be used in combination with other chemotherapeutics for the
treatment of various types of cancer. Furthermore, the antiviral effects showed against Zika, Chikungunya, and
HIV are due to the annulation of endosomal/lysosomal acidification. Recently, CQ and HCQ were approved for
the U.S. Food and Drug Administration (FDA) for the treatment of infected patients with the coronavirus SARSCoV-
2, causing the disease originated in December 2019, namely COVID-2019. Several mechanisms have been
proposed to explain the pharmacological effects of these drugs: 1) disruption of lysosomal and endosomal pH, 2)
inhibition of protein secretion/expression, 3) inhibition of antigen presentation, 4) decrease of proinflammatory
cytokines, 5) inhibition of autophagy, 6) induction of apoptosis and 7) inhibition of ion channels activation. Thus,
evidence has shown that these structures are leading molecules that can be modified or combined with other
therapeutic agents. In this review, we will discuss the most recent findings in the mechanisms of action of CQ and
HCQ in the immune system, and the use of these antimalarial drugs on diseases.
Collapse
Affiliation(s)
- Gricelis P. Martinez
- Institute of Immunology, Faculty of Medicine, Central University of Venezuela, 50109, Los Chaguaramos 1050-A, Caracas, Venezuela
| | - Mercedes E. Zabaleta
- Institute of Immunology, Faculty of Medicine, Central University of Venezuela, 50109, Los Chaguaramos 1050-A, Caracas, Venezuela
| | - Camilo Di Giulio
- Institute of Immunology, Faculty of Medicine, Central University of Venezuela, 50109, Los Chaguaramos 1050-A, Caracas, Venezuela
| | - Jaime E. Charris
- Organic Synthesis Laboratory, Faculty of Pharmacy, Central University of Venezuela, 47206, Los Chaguaramos 1041-A, Caracas, Venezuela
| | - Michael R. Mijares
- Institute of Immunology, Faculty of Medicine, Central University of Venezuela, 50109, Los Chaguaramos 1050-A, Caracas, Venezuela
| |
Collapse
|
8
|
Engineering anti-cancer nanovaccine based on antigen cross-presentation. Biosci Rep 2020; 39:220729. [PMID: 31652460 PMCID: PMC6822533 DOI: 10.1042/bsr20193220] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 09/27/2019] [Accepted: 10/01/2019] [Indexed: 01/16/2023] Open
Abstract
Dendritic cells (DCs) present exogenous antigens on major histocompatibility complex (MHC) class I molecules, thereby activating CD8+ T cells, contributing to tumor elimination through a mechanism known as antigen cross-presentation. A variety of factors such as maturation state of DCs, co-stimulatory signals, T-cell microenvironment, antigen internalization routes and adjuvants regulate the process of DC-mediated antigen cross-presentation. Recently, the development of successful cancer immunotherapies may be attributed to the ability of DCs to cross-present tumor antigens. In this review article, we focus on the underlying mechanism of antigen cross-presentation and ways to improve antigen cross-presentation in different DC subsets. We have critically summarized the recent developments in the generation of novel nanovaccines for robust CD8+ T-cell response in cancer. In this context, we have reviewed nanocarriers that have been used for cancer immunotherapeutics based on antigen cross-presentation mechanism. Additionally, we have also expressed our views on the future applications of this mechanism in curing cancer.
Collapse
|
9
|
Vabret N, Britton GJ, Gruber C, Hegde S, Kim J, Kuksin M, Levantovsky R, Malle L, Moreira A, Park MD, Pia L, Risson E, Saffern M, Salomé B, Esai Selvan M, Spindler MP, Tan J, van der Heide V, Gregory JK, Alexandropoulos K, Bhardwaj N, Brown BD, Greenbaum B, Gümüş ZH, Homann D, Horowitz A, Kamphorst AO, Curotto de Lafaille MA, Mehandru S, Merad M, Samstein RM. Immunology of COVID-19: Current State of the Science. Immunity 2020; 52:910-941. [PMID: 32505227 PMCID: PMC7200337 DOI: 10.1016/j.immuni.2020.05.002] [Citation(s) in RCA: 1171] [Impact Index Per Article: 234.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has affected millions of people worldwide, igniting an unprecedented effort from the scientific community to understand the biological underpinning of COVID19 pathophysiology. In this Review, we summarize the current state of knowledge of innate and adaptive immune responses elicited by SARS-CoV-2 infection and the immunological pathways that likely contribute to disease severity and death. We also discuss the rationale and clinical outcome of current therapeutic strategies as well as prospective clinical trials to prevent or treat SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Nicolas Vabret
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Graham J Britton
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Conor Gruber
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Samarth Hegde
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joel Kim
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Maria Kuksin
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rachel Levantovsky
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Louise Malle
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alvaro Moreira
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew D Park
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Luisanna Pia
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emma Risson
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miriam Saffern
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bérengère Salomé
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Myvizhi Esai Selvan
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew P Spindler
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jessica Tan
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Verena van der Heide
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jill K Gregory
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Nina Bhardwaj
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brian D Brown
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Benjamin Greenbaum
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zeynep H Gümüş
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dirk Homann
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Amir Horowitz
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alice O Kamphorst
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Saurabh Mehandru
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miriam Merad
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Robert M Samstein
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
10
|
Meyerowitz EA, Vannier AGL, Friesen MGN, Schoenfeld S, Gelfand JA, Callahan MV, Kim AY, Reeves PM, Poznansky MC. Rethinking the role of hydroxychloroquine in the treatment of COVID-19. FASEB J 2020; 34:6027-6037. [PMID: 32350928 PMCID: PMC7267640 DOI: 10.1096/fj.202000919] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 04/20/2020] [Indexed: 02/07/2023]
Abstract
There are currently no proven or approved treatments for coronavirus disease 2019 (COVID-19). Early anecdotal reports and limited in vitro data led to the significant uptake of hydroxychloroquine (HCQ), and to lesser extent chloroquine (CQ), for many patients with this disease. As an increasing number of patients with COVID-19 are treated with these agents and more evidence accumulates, there continues to be no high-quality clinical data showing a clear benefit of these agents for this disease. Moreover, these agents have the potential to cause harm, including a broad range of adverse events including serious cardiac side effects when combined with other agents. In addition, the known and potent immunomodulatory effects of these agents which support their use in the treatment of auto-immune conditions, and provided a component in the original rationale for their use in patients with COVID-19, may, in fact, undermine their utility in the context of the treatment of this respiratory viral infection. Specifically, the impact of HCQ on cytokine production and suppression of antigen presentation may have immunologic consequences that hamper innate and adaptive antiviral immune responses for patients with COVID-19. Similarly, the reported in vitro inhibition of viral proliferation is largely derived from the blockade of viral fusion that initiates infection rather than the direct inhibition of viral replication as seen with nucleoside/tide analogs in other viral infections. Given these facts and the growing uncertainty about these agents for the treatment of COVID-19, it is clear that at the very least thoughtful planning and data collection from randomized clinical trials are needed to understand what if any role these agents may have in this disease. In this article, we review the datasets that support or detract from the use of these agents for the treatment of COVID-19 and render a data informed opinion that they should only be used with caution and in the context of carefully thought out clinical trials, or on a case-by-case basis after rigorous consideration of the risks and benefits of this therapeutic approach.
Collapse
Affiliation(s)
- Eric A. Meyerowitz
- Division of Infectious DiseasesMassachusetts General Hospital (MGH) and Harvard Medical School (HMS)BostonMAUSA
| | - Augustin G. L. Vannier
- Division of Infectious DiseasesMassachusetts General Hospital (MGH) and Harvard Medical School (HMS)BostonMAUSA
- Vaccine and Immunotherapy Center (VIC)MGH and HMSBostonMAUSA
| | - Morgan G. N. Friesen
- Division of Infectious DiseasesMassachusetts General Hospital (MGH) and Harvard Medical School (HMS)BostonMAUSA
- Vaccine and Immunotherapy Center (VIC)MGH and HMSBostonMAUSA
| | - Sara Schoenfeld
- Division of Allergy, Immunology and RheumatologyMGH and HMSBostonMAUSA
| | - Jeffrey A. Gelfand
- Division of Infectious DiseasesMassachusetts General Hospital (MGH) and Harvard Medical School (HMS)BostonMAUSA
- Vaccine and Immunotherapy Center (VIC)MGH and HMSBostonMAUSA
| | - Michael V. Callahan
- Division of Infectious DiseasesMassachusetts General Hospital (MGH) and Harvard Medical School (HMS)BostonMAUSA
- Vaccine and Immunotherapy Center (VIC)MGH and HMSBostonMAUSA
- Special Advisor to the Assistant Secretary of Public Health Preparedness and Response U.S Dept of Health and Human ServicesWashingtonDCUSA
| | - Arthur Y. Kim
- Division of Infectious DiseasesMassachusetts General Hospital (MGH) and Harvard Medical School (HMS)BostonMAUSA
| | - Patrick M. Reeves
- Division of Infectious DiseasesMassachusetts General Hospital (MGH) and Harvard Medical School (HMS)BostonMAUSA
- Vaccine and Immunotherapy Center (VIC)MGH and HMSBostonMAUSA
| | - Mark C. Poznansky
- Division of Infectious DiseasesMassachusetts General Hospital (MGH) and Harvard Medical School (HMS)BostonMAUSA
- Vaccine and Immunotherapy Center (VIC)MGH and HMSBostonMAUSA
| |
Collapse
|
11
|
Fu Y, Lu X, Zhu F, Zhao Y, Ding Y, Ye L, Guo B, Liu T, Xu W. Improving the immunogenicity and protective efficacy of a whole-killed malaria blood-stage vaccine by chloroquine. Parasite Immunol 2020; 42:e12682. [PMID: 31644820 DOI: 10.1111/pim.12682] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 10/12/2019] [Accepted: 10/21/2019] [Indexed: 12/15/2022]
Abstract
A whole-killed malaria blood-stage vaccine (WKV) is promising in reducing the morbidity and mortality of malaria patients, but its efficacy needs to be improved. We found that the antimalarial drug chloroquine could augment the protective efficacy of the WKV of Plasmodium yoelii. The direct antimalarial effect of chloroquine on parasites during immunization could be excluded, as the administration of chloroquine or chloroquine plus alum every two weeks had a slight effect on parasitemia, and an immunization with NP-KLH (4-hydroxy-3-nitrophenylacetyl Keyhole Limpet Hemocyanin) plus chloroquine could significantly promote the generation of NP-specific antibodies. Additionally, alum was required for chloroquine to augment the immunogenicity of the pRBC lysate. Chloroquine did not promote the parasite-specific CD4+ T-cell responses, but significantly enhanced the WKV-induced germinal centre B cell reactions, class-switch recombination and secretion of functionally protective antibodies to plasmodium. The elevated parasite-specific antibodies were demonstrated to largely contribute to the chloroquine-enhanced protective immunity. Thus, we report that chloroquine could be used as an adjuvant to enhance the protective immunity of WKVs through promoting humoral responses.
Collapse
Affiliation(s)
- Yong Fu
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiao Lu
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Feng Zhu
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yunxiang Zhao
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yan Ding
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Lilin Ye
- Institute of Immunology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Bo Guo
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Taiping Liu
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Wenyue Xu
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
12
|
Cross-presentation of Exogenous Antigens. Transfus Clin Biol 2019; 26:346-351. [DOI: 10.1016/j.tracli.2019.01.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 01/25/2019] [Indexed: 01/07/2023]
|
13
|
Ma X, Zhang Y, Zhu D, Chen Z, Xu M, He L, Shi T, Huang L, Zou J. Low dosage chloroquine protects retinal ganglion cells against glutamate-induced cell death. Exp Eye Res 2019; 181:285-293. [PMID: 30831085 DOI: 10.1016/j.exer.2019.02.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 01/17/2019] [Accepted: 02/25/2019] [Indexed: 01/18/2023]
Affiliation(s)
- Xiaoyun Ma
- Department of Ophthalmology, Guanghua Integrative Medicine Hospital, Shanghai, China.
| | - Yun Zhang
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Dan Zhu
- Department of Ophthalmology, Guanghua Integrative Medicine Hospital, Shanghai, China
| | - Zufeng Chen
- Department of Ophthalmology, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Manshan Xu
- Department of Ophthalmology, Guanghua Integrative Medicine Hospital, Shanghai, China
| | - Linping He
- Department of Ophthalmology, Guanghua Integrative Medicine Hospital, Shanghai, China
| | - Tingli Shi
- Department of Ophthalmology, Guanghua Integrative Medicine Hospital, Shanghai, China
| | - Lvzhen Huang
- Department of Ophthalmology, Peking University People's Hospital, Eye Diseases and Optometry Instistute, Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases College of Optometry, Peking University Health Science Center, China.
| | - Jun Zou
- Department of Ophthalmology, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, China.
| |
Collapse
|
14
|
Arbogast F, Gros F. Lymphocyte Autophagy in Homeostasis, Activation, and Inflammatory Diseases. Front Immunol 2018; 9:1801. [PMID: 30127786 PMCID: PMC6087746 DOI: 10.3389/fimmu.2018.01801] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 07/23/2018] [Indexed: 12/12/2022] Open
Abstract
Autophagy is a catabolic mechanism, allowing the degradation of cytoplasmic content via lysosomal activity. Several forms of autophagy are described in mammals. Macroautophagy leads to integration of cytoplasmic portions into vesicles named autophagosomes that ultimately fuse with lysosomes. Chaperone-mediated autophagy is in contrast the direct translocation of protein in lysosomes. Macroautophagy is central to lymphocyte homeostasis. Although its role is controversial in lymphocyte development and in naive cell survival, it seems particularly involved in the maintenance of certain lymphocyte subtypes. Its importance in memory B and T cells biology has recently emerged. Moreover, some effector cells like plasma cells rely on autophagy for survival. Autophagy is central to glucose and lipid metabolism, and to the maintenance of organelles like mitochondria and endoplasmic reticulum. In addition macroautophagy, or individual components of its machinery, are also actors in antigen presentation by B cells, a crucial step to receive help from T cells, this crosstalk favoring their final differentiation into memory or plasma cells. Autophagy is deregulated in several autoimmune or autoinflammatory diseases like systemic lupus erythematosus, rheumatoid arthritis, multiple sclerosis, and Crohn’s disease. Some treatments used in these pathologies impact autophagic activity, even if the causal link between autophagy regulation and the efficiency of the treatments has not yet been clearly established. In this review, we will first discuss the mechanisms linking autophagy to lymphocyte subtype survival and the signaling pathways involved. Finally, potential impacts of autophagy modulation in lymphocytes on the course of these diseases will be approached.
Collapse
Affiliation(s)
- Florent Arbogast
- CNRS UPR3572, Immunology, Immunopathology and Therapeutic Chemistry/Laboratory of Excellence MEDALIS, Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France.,University of Strasbourg, Strasbourg, France
| | - Frédéric Gros
- CNRS UPR3572, Immunology, Immunopathology and Therapeutic Chemistry/Laboratory of Excellence MEDALIS, Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France.,University of Strasbourg, Strasbourg, France
| |
Collapse
|
15
|
Pathangey LB, McCurry DB, Gendler SJ, Dominguez AL, Gorman JE, Pathangey G, Mihalik LA, Dang Y, Disis ML, Cohen PA. Surrogate in vitro activation of innate immunity synergizes with interleukin-7 to unleash rapid antigen-driven outgrowth of CD4+ and CD8+ human peripheral blood T-cells naturally recognizing MUC1, HER2/neu and other tumor-associated antigens. Oncotarget 2017; 8:10785-10808. [PMID: 27974697 PMCID: PMC5355224 DOI: 10.18632/oncotarget.13911] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 11/23/2016] [Indexed: 01/21/2023] Open
Abstract
Effective adoptive immunotherapy has proved elusive for many types of human cancer, often due to difficulties achieving robust expansion of natural tumor-specific T-cells from peripheral blood. We hypothesized that antigen-driven T-cell expansion might best be triggered in vitro by acute activation of innate immunity to mimic a life-threatening infection. Unfractionated peripheral blood mononuclear cells (PBMC) were subjected to a two-step culture, first synchronizing their exposure to exogenous antigens with aggressive surrogate activation of innate immunity, followed by γ-chain cytokine-modulated T-cell hyperexpansion. Step 1 exposure to GM-CSF plus paired Toll-like receptor agonists (resiquimod and LPS), stimulated abundant IL-12 and IL-23 secretion, as well as upregulated co-stimulatory molecules and CD11c expression within the myeloid (CD33+) subpopulation. Added synthetic long peptides (>20aa) derived from widely expressed oncoproteins (MUC1, HER2/neu and CMVpp65), were reliably presented to CD4+ T-cells and cross-presented to CD8+ T-cells. Both presentation and cross-presentation demonstrated proteasomal and Sec61 dependence that could bypass the endoplasmic reticulum. Step 2 exposure to exogenous IL-7 or IL-7+IL-2 produced selective and sustained expansion of both CD4+ and CD8+ peptide-specific T-cells with a predominant interferon-γ-producing T1-type, as well as the antigen-specific ability to lyse tumor targets. Other γ-chain cytokines and/or combinations were initially proliferogenic, but followed by a contractile phase not observed with IL-7 or IL-7+IL-2. Regulatory T-cells were minimally propagated under these culture conditions. This mechanistically rational culture sequence, effective even for unvaccinated donors, enables rapid preparation of T-cells recognizing tumor-associated antigens expressed by the majority of human cancers, including pancreatic cancers, breast cancers and glioblastomas.
Collapse
Affiliation(s)
- Latha B Pathangey
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Scottsdale, AZ, USA
| | - Dustin B McCurry
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Scottsdale, AZ, USA
| | - Sandra J Gendler
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Scottsdale, AZ, USA.,Department of Immunology, Mayo Clinic, Scottsdale, AZ, USA.,Department of Hematology and Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Ana L Dominguez
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Scottsdale, AZ, USA
| | - Jessica E Gorman
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Scottsdale, AZ, USA
| | - Girish Pathangey
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Scottsdale, AZ, USA
| | - Laurie A Mihalik
- Department of Hematology and Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Yushe Dang
- Tumor Vaccine Group, Center for Translational Medicine in Women's Health, University of Washington, Seattle, WA, USA
| | - Mary L Disis
- Tumor Vaccine Group, Center for Translational Medicine in Women's Health, University of Washington, Seattle, WA, USA
| | - Peter A Cohen
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, USA.,Department of Hematology and Oncology, Mayo Clinic, Scottsdale, AZ, USA
| |
Collapse
|
16
|
Recruitment of bone marrow CD11b +Gr-1 + cells by polymeric nanoparticles for antigen cross-presentation. Sci Rep 2017; 7:44691. [PMID: 28317931 PMCID: PMC5357800 DOI: 10.1038/srep44691] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 02/13/2017] [Indexed: 12/31/2022] Open
Abstract
The objective of this study was to investigate the function of poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) on the activation of antigen-specific CD8+ T cell responses via the CD11b+Gr-1+ myeloid subpopulations in murine bone marrow (BM). PLGA NPs containing ovalbumin (OVA) were fabricated by the double-emulsion method. The CD11b+Gr-1lowLy-6Chigh and CD11b+Gr-1highLy-6Clow subsets from mice bone marrow were sorted and treated with the PLGA/OVA NPs, followed by co-culture with the carboxyfluorescein succinimidyl ester (CFSE)-labelled OT-I CD8+ cells. Co-culture of OT-I CD8+ T cells with PLGA/OVA NPs-primed CD11b+Gr-1+ subsets upregulated the expression of IL-2, TNF-α, INF-γ, granzyme B, and perforin, resulting in proliferation of CD8+ T cells and differentiation into effector cytotoxic T lymphocytes (CTLs). In vivo proliferation of CFSE-labelled OT-I CD8+ cells in response to OVA was also obtained in the animals immunized with PLGA/OVA NPs. The results presented in this study demonstrate the ability of polymeric NPs to recruit two CD11b+Gr-1+ myeloid subsets for effective presentation of exogenous antigen to OT-I CD8+ T cells in the context of major histocompatibility complex (MHC) class I, leading to an induction of antigen-specific cell proliferation and differentiation into effector cells.
Collapse
|
17
|
Chotprakaikiat W, Allen A, Bui-Minh D, Harden E, Jobsri J, Cavallo F, Gleba Y, Stevenson FK, Ottensmeier C, Klimyuk V, Savelyeva N. A plant-expressed conjugate vaccine breaks CD4(+) tolerance and induces potent immunity against metastatic Her2(+) breast cancer. Oncoimmunology 2016; 5:e1166323. [PMID: 27471642 PMCID: PMC4938312 DOI: 10.1080/2162402x.2016.1166323] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 02/29/2016] [Accepted: 03/11/2016] [Indexed: 02/06/2023] Open
Abstract
Passive antibody therapy for cancer is an effective but costly treatment modality. Induction of therapeutically potent anticancer antibodies by active vaccination is an attractive alternative but has proven challenging in cancer due to tolerogenic pressure in patients. Here, we used the clinically relevant cancer target Her2, known to be susceptible to targeting by antibody therapy, to demonstrate how potent antibody can be induced by vaccination. A novel 44kD Her2 protein fragment was generated and found to be highly effective at inducing anti-Her2 antibody including trastuzumab-like reactivities. In the tolerant and spontaneous BALB-neuT mouse model of metastatic breast cancer this Her2-targeting vaccine was only effective if the fragment was conjugated to a foreign immunogenic carrier; Fragment C of tetanus toxin. Only the conjugate vaccine induced high affinity anti-Her2 antibody of multiple isotypes and suppressed tumor development. The magnitude of CD4(+) T-cell help and breadth of cytokines secreted by the CD4(+) T helper (Th) cells induced to the foreign antigen was critical. We used a highly efficient plant-based bio-manufacturing process for protein antigens, magnICON, for vaccine expression, to underpin feasibility of future clinical testing. Hence, our novel Her2-targeting conjugate vaccine combines preclinical efficacy with clinical deliverability, thus setting the scene for therapeutic testing.
Collapse
Affiliation(s)
| | - Alex Allen
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| | | | - Elena Harden
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Jantipa Jobsri
- Oral Biology Department, Naresuan University, Phitsanulok, Thailand
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| | | | - Freda K. Stevenson
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Christian Ottensmeier
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| | | | - Natalia Savelyeva
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| |
Collapse
|
18
|
Silva-Barrios S, Smans M, Duerr C, Qureshi S, Fritz J, Descoteaux A, Stäger S. Innate Immune B Cell Activation by Leishmania donovani Exacerbates Disease and Mediates Hypergammaglobulinemia. Cell Rep 2016; 15:2427-37. [DOI: 10.1016/j.celrep.2016.05.028] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 03/31/2016] [Accepted: 05/04/2016] [Indexed: 01/27/2023] Open
|
19
|
Coria LM, Ibañez AE, Tkach M, Sabbione F, Bruno L, Carabajal MV, Berguer PM, Barrionuevo P, Schillaci R, Trevani AS, Giambartolomei GH, Pasquevich KA, Cassataro J. A Brucella spp. Protease Inhibitor Limits Antigen Lysosomal Proteolysis, Increases Cross-Presentation, and Enhances CD8+ T Cell Responses. THE JOURNAL OF IMMUNOLOGY 2016; 196:4014-29. [PMID: 27084100 DOI: 10.4049/jimmunol.1501188] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 03/15/2016] [Indexed: 01/18/2023]
Abstract
In this study, we demonstrate that the unlipidated (U) outer membrane protein (Omp) 19 from Brucella spp. is a competitive inhibitor of human cathepsin L. U-Omp19 inhibits lysosome cathepsins and APC-derived microsome activity in vitro and partially inhibits lysosomal cathepsin L activity within live APCs. Codelivery of U-Omp19 with the Ag can reduce intracellular Ag digestion and increases Ag half-life in dendritic cells (DCs). U-Omp19 retains the Ag in Lamp-2(+) compartments after its internalization and promotes a sustained expression of MHC class I/peptide complexes in the cell surface of DCs. Consequently, U-Omp19 enhances Ag cross-presentation by DCs to CD8(+) T cells. U-Omp19 s.c. delivery induces the recruitment of CD11c(+)CD8α(+) DCs and monocytes to lymph nodes whereas it partially limits in vivo Ag proteolysis inside DCs. Accordingly, this protein is able to induce CD8(+) T cell responses in vivo against codelivered Ag. Antitumor responses were elicited after U-Omp19 coadministration, increasing survival of mice in a murine melanoma challenge model. Collectively, these results indicate that a cysteine protease inhibitor from bacterial origin could be a suitable component of vaccine formulations against tumors.
Collapse
Affiliation(s)
- Lorena M Coria
- Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico de Chascomús, Universidad Nacional de San Martín, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, 1650 Buenos Aires, Argentina
| | - Andrés E Ibañez
- Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico de Chascomús, Universidad Nacional de San Martín, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, 1650 Buenos Aires, Argentina
| | - Mercedes Tkach
- Laboratorio de Mecanismos Moleculares de Carcinogénesis, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, 1428 Buenos Aires, Argentina
| | - Florencia Sabbione
- Instituto de Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Academia Nacional de Medicina, 1425 Buenos Aires, Argentina
| | - Laura Bruno
- Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico de Chascomús, Universidad Nacional de San Martín, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, 1650 Buenos Aires, Argentina
| | - Marianela V Carabajal
- Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico de Chascomús, Universidad Nacional de San Martín, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, 1650 Buenos Aires, Argentina
| | - Paula M Berguer
- Fundación Instituto Leloir, Instituto de Investigaciones Bioquímicas de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, 1405 Buenos Aires, Argentina; and
| | - Paula Barrionuevo
- Instituto de Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Academia Nacional de Medicina, 1425 Buenos Aires, Argentina
| | - Roxana Schillaci
- Laboratorio de Mecanismos Moleculares de Carcinogénesis, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, 1428 Buenos Aires, Argentina
| | - Analía S Trevani
- Instituto de Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Academia Nacional de Medicina, 1425 Buenos Aires, Argentina
| | - Guillermo H Giambartolomei
- Instituto de Inmunología, Genética y Metabolismo, Laboratorio de Inmunogenética, Hospital de Clínicas "José de San Martín," Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Universidad de Buenos Aires, 1120 Buenos Aires, Argentina
| | - Karina A Pasquevich
- Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico de Chascomús, Universidad Nacional de San Martín, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, 1650 Buenos Aires, Argentina
| | - Juliana Cassataro
- Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico de Chascomús, Universidad Nacional de San Martín, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, 1650 Buenos Aires, Argentina;
| |
Collapse
|
20
|
Alyaqoub FS, Aldhamen YA, Koestler BJ, Bruger EL, Seregin SS, Pereira-Hicks C, Godbehere S, Waters CM, Amalfitano A. In Vivo Synthesis of Cyclic-di-GMP Using a Recombinant Adenovirus Preferentially Improves Adaptive Immune Responses against Extracellular Antigens. THE JOURNAL OF IMMUNOLOGY 2016; 196:1741-52. [PMID: 26792800 DOI: 10.4049/jimmunol.1501272] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 12/17/2015] [Indexed: 12/17/2022]
Abstract
There is a compelling need for more effective vaccine adjuvants to augment induction of Ag-specific adaptive immune responses. Recent reports suggested the bacterial second messenger bis-(3'-5')-cyclic-dimeric-guanosine monophosphate (c-di-GMP) acts as an innate immune system modulator. We recently incorporated a Vibrio cholerae diguanylate cyclase into an adenovirus vaccine, fostering production of c-di-GMP as well as proinflammatory responses in mice. In this study, we recombined a more potent diguanylate cyclase gene, VCA0848, into a nonreplicating adenovirus serotype 5 (AdVCA0848) that produces elevated amounts of c-di-GMP when expressed in mammalian cells in vivo. This novel platform further improved induction of type I IFN-β and activation of innate and adaptive immune cells early after administration into mice as compared with control vectors. Coadministration of the extracellular protein OVA and the AdVCA0848 adjuvant significantly improved OVA-specific T cell responses as detected by IFN-γ and IL-2 ELISPOT, while also improving OVA-specific humoral B cell adaptive responses. In addition, we found that coadministration of AdVCA0848 with another adenovirus serotype 5 vector expressing the HIV-1-derived Gag Ag or the Clostridium difficile-derived toxin B resulted in significant inhibitory effects on the induction of Gag and toxin B-specific adaptive immune responses. As a proof of principle, these data confirm that in vivo synthesis of c-di-GMP stimulates strong innate immune responses that correlate with enhanced adaptive immune responses to concomitantly administered extracellular Ag, which can be used as an adjuvant to heighten effective immune responses for protein-based vaccine platforms against microbial infections and cancers.
Collapse
Affiliation(s)
- Fadel S Alyaqoub
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824
| | - Yasser A Aldhamen
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824
| | - Benjamin J Koestler
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824; BEACON Center for the Study of Evolution in Action, Michigan State University, East Lansing, MI 48824; and
| | - Eric L Bruger
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824; BEACON Center for the Study of Evolution in Action, Michigan State University, East Lansing, MI 48824; and
| | - Sergey S Seregin
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824
| | - Cristiane Pereira-Hicks
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824
| | - Sarah Godbehere
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824
| | - Christopher M Waters
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824; BEACON Center for the Study of Evolution in Action, Michigan State University, East Lansing, MI 48824; and
| | - Andrea Amalfitano
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824; Department of Pediatrics, Michigan State University, East Lansing, MI 48824
| |
Collapse
|
21
|
Time to use a dose of Chloroquine as an adjuvant to anti-cancer chemotherapies. Eur J Pharmacol 2015; 771:139-44. [PMID: 26687632 DOI: 10.1016/j.ejphar.2015.12.017] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 11/27/2015] [Accepted: 12/09/2015] [Indexed: 01/05/2023]
Abstract
Chloroquine, a drug used for over 80 years to treat and prevent malaria and, more recently, to treat autoimmune diseases, is very safe but has a plethora of dose-dependent effects. By increasing pH in acidic compartments it inhibits for example lysosomal enzymes. In the context of cancer, Chloroquine was found to have direct effects on different types of malignancies that could potentiate chemotherapies. For example, the anti-malaria drug may inhibit both the multidrug-resistance pump and autophagy (mechanisms that tumor cells may use to resist chemotherapies), intercalate in DNA and enhance the penetration of chemotherapeutic drugs in cells or solid cancer tissues. However, these activities were mostly demonstrated at high doses of Chloroquine (higher than 10mg/kg or 10mg/l i.e. ca. 31μM). Nevertheless, it was reported that daily uptake of clinically acceptable doses (less than 10mg/kg) of Chloroquine in addition to chemo-radio-therapy increases the survival of glioblastoma patients (Sotelo et al., 2006; Briceno et al., 2007). However, the optimal dose and schedule of this multi-active drug with respect to chemotherapy has never been experimentally determined. The present article reviews the several known direct and indirect effects of different doses of Chloroquine on cancer and how those effects may indicate that a fine tuning of the dose/schedule of Chloroquine administration versus chemotherapy may be critical to obtain an adjuvant effect of Chloroquine in anti-cancer treatments. We anticipate that the appropriate (time and dose) addition of Chloroquine to the standard of care may greatly and safely potentiate current anti-cancer treatments.
Collapse
|
22
|
Becker HJ, Kondo E, Shimabukuro-Vornhagen A, Theurich S, von Bergwelt-Baildon MS. Processing and MHC class II presentation of exogenous soluble antigen involving a proteasome-dependent cytosolic pathway in CD40-activated B cells. Eur J Haematol 2015; 97:166-74. [DOI: 10.1111/ejh.12699] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2015] [Indexed: 01/08/2023]
Affiliation(s)
- Hans Jiro Becker
- Cologne Interventional Immunology; Cologne University Hospital; Cologne Germany
| | - Eisei Kondo
- Department of General Medicine; Okayama University Graduate School of Medicine; Dentistry and Pharmaceutical Sciences; Okayama Japan
| | | | - Sebastian Theurich
- Cologne Interventional Immunology; Cologne University Hospital; Cologne Germany
- Max Planck Institute for Metabolism Research; Cologne Germany
| | | |
Collapse
|
23
|
Zachova K, Krupka M, Raska M. Antigen Cross-Presentation and Heat Shock Protein-Based Vaccines. Arch Immunol Ther Exp (Warsz) 2015; 64:1-18. [DOI: 10.1007/s00005-015-0370-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 08/31/2015] [Indexed: 12/15/2022]
|
24
|
Baleeiro RB, Rietscher R, Diedrich A, Czaplewska JA, Lehr CM, Scherließ R, Hanefeld A, Gottschaldt M, Walden P. Spatial separation of the processing and MHC class I loading compartments for cross-presentation of the tumor-associated antigen HER2/ neu by human dendritic cells. Oncoimmunology 2015; 4:e1047585. [PMID: 26985398 DOI: 10.1080/2162402x.2015.1047585] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 04/20/2015] [Accepted: 04/27/2015] [Indexed: 10/23/2022] Open
Abstract
Cross-presentation is the process by which professional antigen presenting cells (APCs) (B cells, dendritic cells (DCs) and macrophages) present endocytosed antigens (Ags) via MHC-I to CD8+ T cells. This process is crucial for induction of adaptive immune responses against tumors and infected cells. The pathways and cellular compartments involved in cross-presentation are unresolved and controversial. Among the cells with cross-presenting capacity, DCs are the most efficient, which was proposed to depend on prevention of endosomal acidification to block degradation of the epitopes. Contrary to this view, we show in this report that some cargoes induce strong endosomal acidification following uptake by human DCs, while others not. Moreover, processing of the tumor-associated antigen HER2/neu delivered in nanoparticles (NP) for cross-presentation of the epitope HER2/neu369-377 on HLA-A2 depended on endosomal acidification and cathepsin activity as well as proteasomes, and newly synthesized HLA class I. However, the HLA-A*0201/HER2/neu369-377 complexes were not found in the endoplasmic reticulum (ER) nor in endolysosomes but in hitherto not described vesicles. The data thus indicate spatial separation of antigen processing and loading of MHC-I for cross-presentation: antigen processing occurs in the uptake compartment and the cytosol whereas MHC-I loading with peptide takes place in a distinct subcellular compartment. The findings further elucidate the cellular pathways involved in the cross-presentation of a full-length, clinically relevant tumor-associated antigen by human DCs, and the impact of the vaccine formulation on antigen processing and CD8+ T cell induction.
Collapse
Affiliation(s)
- Renato B Baleeiro
- Department of Dermatology; Venerology and Allergology; Charité - Universitätsmedizin Berlin ; Berlin, Germany
| | - René Rietscher
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS); Helmholtz Centre for Infection Research; Saarland University ; Saarbrücken, Germany
| | - Andrea Diedrich
- Department of Pharmaceutics and Biopharmaceutics; Kiel University ; Kiel, Germany
| | - Justyna A Czaplewska
- Laboratory of Organic and Macromolecular Chemistry (IOMC); Friedrich Schiller University Jena; Jena, Germany; Jena Center for Soft Matter (JCSM); Friedrich Schiller University Jena; Jena, Germany
| | - Claus-Michael Lehr
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS); Helmholtz Centre for Infection Research; Saarland University; Saarbrücken, Germany; Biopharmaceutics and Pharmaceutical Technology; Saarland University; Saarbrücken, Germany
| | - Regina Scherließ
- Department of Pharmaceutics and Biopharmaceutics; Kiel University ; Kiel, Germany
| | | | - Michael Gottschaldt
- Laboratory of Organic and Macromolecular Chemistry (IOMC); Friedrich Schiller University Jena; Jena, Germany; Jena Center for Soft Matter (JCSM); Friedrich Schiller University Jena; Jena, Germany
| | - Peter Walden
- Department of Dermatology; Venerology and Allergology; Charité - Universitätsmedizin Berlin ; Berlin, Germany
| |
Collapse
|
25
|
Bijker EM, Nganou-Makamdop K, van Gemert GJ, Zavala F, Cockburn I, Sauerwein RW. Studying the effect of chloroquine on sporozoite-induced protection and immune responses in Plasmodium berghei malaria. Malar J 2015; 14:130. [PMID: 25889324 PMCID: PMC4389414 DOI: 10.1186/s12936-015-0626-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 02/20/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Sporozoite immunization of animals and humans under a chemo-prophylactic cover of chloroquine (CPS-CQ) efficiently induces sterile protection against malaria. In humans, CPS-CQ is strikingly more efficient than immunization with radiation attenuated sporozoites (RAS), raising the hypothesis that this might be partially due to CQ. Chloroquine, an established anti-malarial drug, is also well known for its immune modulating properties including improvement of cross-presentation. The aim of this study was to investigate whether co-administration of CQ during sporozoite immunization improves cellular responses and protective efficacy in Plasmodium berghei models. METHODS A number of experiments in selected complimentary P. berghei murine models in Balb/cByJ and C57BL/6j mice was performed. First, the effect of CQ administration on the induction of protection and immune responses by RAS immunization was studied. Next, the effect of CQ on the induction of circumsporozoite (CS) protein-specific CD8(+) T cells by immunization with P. berghei parasites expressing a mutant CS protein was investigated. Finally, a direct comparison of CPS-CQ to CPS with mefloquine (MQ), an anti-malarial with little known immune modulating effects, was performed. RESULTS When CQ was co-administered during immunization with graded numbers of RAS, this did not lead to an increase in frequencies of total memory CD8(+) T cells or CS protein-specific CD8(+) T cells. Also parasite-specific cytokine production and protection remained unaltered. Replacement of CQ by MQ for CPS immunization resulted in significantly reduced percentages of IFNγ producing memory T cells in the liver (p = 0.01), but similar protection. CONCLUSIONS This study does not provide evidence for a direct beneficial effect of CQ on the induction of sporozoite-induced immune responses and protection in P. berghei malaria models. Alternatively, the higher efficiency of CPS compared to RAS might be explained by an indirect effect of CQ through limiting blood-stage exposure after immunization or to increased antigen exposure and, therefore, improved breadth of the immune response.
Collapse
Affiliation(s)
- Else M Bijker
- Department of Medical Microbiology, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
| | - Krystelle Nganou-Makamdop
- Department of Medical Microbiology, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands. .,Current address: Vaccine Research Centre; National Institutes of Health, 40 Convent drive, Bethesda, MD, 20892, USA.
| | - Geert-Jan van Gemert
- Department of Medical Microbiology, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
| | - Fidel Zavala
- Johns Hopkins Malaria Research Institute and Department of Molecular Microbiology and Immunology, John Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA.
| | - Ian Cockburn
- Johns Hopkins Malaria Research Institute and Department of Molecular Microbiology and Immunology, John Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA. .,Current address: John Curtin School of Medical Research, Australian National University, GPO Box 334, Canberra City, ACT 2600, Australia.
| | - Robert W Sauerwein
- Department of Medical Microbiology, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
26
|
Combinatorial contextualization of peptidic epitopes for enhanced cellular immunity. PLoS One 2014; 9:e110425. [PMID: 25343355 PMCID: PMC4208766 DOI: 10.1371/journal.pone.0110425] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 09/13/2014] [Indexed: 11/25/2022] Open
Abstract
Invocation of cellular immunity by epitopic peptides remains largely dependent on empirically developed protocols, such as interfusion of aluminum salts or emulsification using terpenoids and surfactants. To explore novel vaccine formulation, epitopic peptide motifs were co-programmed with structural motifs to produce artificial antigens using our “motif-programming” approach. As a proof of concept, we used an ovalbumin (OVA) system and prepared an artificial protein library by combinatorially polymerizing MHC class I and II sequences from OVA along with a sequence that tends to form secondary structures. The purified endotoxin-free proteins were then examined for their ability to activate OVA-specific T-cell hybridoma cells after being processed within dendritic cells. One clone, F37A (containing three MHC I and two MHC II OVA epitopes), possessed a greater ability to evoke cellular immunity than the native OVA or the other artificial antigens. The sensitivity profiles of drugs that interfered with the F37A uptake differed from those of the other artificial proteins and OVA, suggesting that alteration of the cross-presentation pathway is responsible for the enhanced immunogenicity. Moreover, F37A, but not an epitopic peptide, invoked cellular immunity when injected together with monophosphoryl lipid A (MPL), and retarded tumor growth in mice. Thus, an artificially synthesized protein antigen induced cellular immunity in vivo in the absence of incomplete Freund's adjuvant or aluminum salts. The method described here could be potentially used for developing vaccines for such intractable ailments as AIDS, malaria and cancer, ailments in which cellular immunity likely play a crucial role in prevention and treatment.
Collapse
|
27
|
Leroux-Roels G, Bourguignon P, Willekens J, Janssens M, Clement F, Didierlaurent AM, Fissette L, Roman F, Boutriau D. Immunogenicity and safety of a booster dose of an investigational adjuvanted polyprotein HIV-1 vaccine in healthy adults and effect of administration of chloroquine. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2014; 21:302-11. [PMID: 24391139 PMCID: PMC3957681 DOI: 10.1128/cvi.00617-13] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 12/21/2013] [Indexed: 11/20/2022]
Abstract
This phase II study evaluated the effect of chloroquine on the specific CD8(+) T-cell responses to and the safety of a booster dose of investigational human immunodeficiency virus type 1 (HIV-1) F4/AS01(B) vaccine containing 10 μg of recombinant fusion protein (F4) adjuvanted with the AS01(B) adjuvant system. Healthy adults aged 21 to 41 years, primed 3 years before with two F4/AS01(B) doses containing 10 or 30 μg of F4 (ClinicalTrials.gov registration number NCT00434512), were randomized (1:1) to receive the F4/AS01(B) booster administered alone or 2 days after chloroquine (300 mg). F4-specific CD8(+)/CD4(+) T-cell responses were characterized by intracellular cytokine staining and lymphoproliferation assays and anti-F4 antibodies by enzyme-linked immunosorbent assays (ELISAs). No effect of chloroquine on CD4(+)/CD8(+) T-cell and antibody responses and no vaccine effect on CD8(+) T-cell responses (cytokine secretion or proliferation) were detected following F4/AS01(B) booster administration. In vitro, chloroquine had a direct inhibitory effect on AS01(B) adjuvant properties; AS01-induced cytokine production decreased upon coincubation of cells with chloroquine. In the pooled group of participants primed with F4/AS01(B) containing 10 μg of F4, CD4(+) T-cell and antibody responses induced by primary vaccination persisted for at least 3 years. The F4/AS01(B) booster induced strong F4-specific CD4(+) T-cell responses, which persisted for at least 6 months with similar frequencies and polyfunctional phenotypes as following primary vaccination, and high anti-F4 antibody concentrations, reaching higher levels than those following primary vaccination. The F4/AS01(B) booster had a clinically acceptable safety and reactogenicity profile. An F4/AS01(B) booster dose, administered alone or after chloroquine, induced robust antibody and F4-specific CD4(+) T-cell responses but no significant CD8(+) T-cell responses (cytokine secretion or proliferation) in healthy adults. (This study has been registered at ClinicalTrials.gov under registration number NCT00972725).
Collapse
|
28
|
Dendritic cells treated with chloroquine modulate experimental autoimmune encephalomyelitis. Immunol Cell Biol 2013; 92:124-32. [PMID: 24217811 DOI: 10.1038/icb.2013.73] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Revised: 10/03/2013] [Accepted: 10/11/2013] [Indexed: 12/22/2022]
Abstract
Chloroquine (CQ), an antimalarial drug, has been shown to modulate the immune system and reduce the severity of experimental autoimmune encephalomyelitis (EAE). The mechanisms of disease suppression are dependent on regulatory T cell induction, although Tregs-independent mechanisms exist. We aimed to evaluate whether CQ is capable to modulate bone marrow-derived dendritic cells (DCs) both phenotypically and functionally as well as whether transfer of CQ-modulated DCs reduces EAE course. Our results show that CQ-treated DCs presented altered ultrastructure morphology and lower expression of molecules involved in antigen presentation. Consequently, T cell proliferation was diminished in coculture experiments. When transferred into EAE mice, DC-CQ was able to reduce the clinical manifestation of the disease through the modulation of the immune response against neuroantigens. The data presented herein indicate that chloroquine-mediated modulation of the immune system is achieved by a direct effect on DCs and that DC-CQ adoptive transfer may be a promising approach for avoiding drug toxicity.
Collapse
|
29
|
Chloroquine: modes of action of an undervalued drug. Immunol Lett 2013; 153:50-7. [PMID: 23891850 DOI: 10.1016/j.imlet.2013.07.004] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Revised: 07/10/2013] [Accepted: 07/15/2013] [Indexed: 12/30/2022]
Abstract
For more than two decades, chloroquine (CQ) was largely and deliberately used as first choice drug for malaria treatment. However, worldwide increasing cases of resistant strains of Plasmodium have hampered its use. Nevertheless, CQ has recently been tested as adjunct therapy in several inflammatory situations, such as rheumatoid arthritis and transplantation procedures, presenting intriguing and promising results. In this review, we discuss recent findings and CQ mechanisms of action vis-à-vis its use as a broad adjunct therapy.
Collapse
|
30
|
Enhancement of T cell-mediated immune responses to whole inactivated influenza virus by chloroquine treatment in vivo. Vaccine 2013; 31:1717-24. [PMID: 23380456 DOI: 10.1016/j.vaccine.2013.01.037] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Revised: 12/16/2012] [Accepted: 01/21/2013] [Indexed: 11/21/2022]
Abstract
Current influenza vaccines induce poor cross-reactive CD8+ T cell responses. Cellular immunity is generally specific for epitopes that are remarkably conserved among different subtypes, suggesting that strategies to improve the cross-presentation of viral antigens by dendritic cells (DC) could elicit a broadly protective immune response. Previous studies have shown that limited proteolysis within the endocytic pathway can favorably influence antigen processing and thus immune responses. Herein, we demonstrate that chloroquine improves the cross-presentation of non-replicating influenza virus in vitro and T cell responses in mice following a single administration of inactivated HI-X31 virus. CD8+ T cells were also recruited to lymph nodes draining the site of infection and able to reduce viral load following pulmonary challenge with the heterologous PR8 virus. These findings may have implications for vaccination strategies aimed at improving the cross-presentation capacity of DCs and thus the size of effector and memory CD8+ T cells against influenza vaccines.
Collapse
|
31
|
Hogenesch H. Mechanism of immunopotentiation and safety of aluminum adjuvants. Front Immunol 2013; 3:406. [PMID: 23335921 PMCID: PMC3541479 DOI: 10.3389/fimmu.2012.00406] [Citation(s) in RCA: 234] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Accepted: 12/16/2012] [Indexed: 12/24/2022] Open
Abstract
Aluminum-containing adjuvants are widely used in preventive vaccines against infectious diseases and in preparations for allergy immunotherapy. The mechanism by which they enhance the immune response remains poorly understood. Aluminum adjuvants selectively stimulate a Th2 immune response upon injection of mice and a mixed response in human beings. They support activation of CD8 T cells, but these cells do not undergo terminal differentiation to cytotoxic T cells. Adsorption of antigens to aluminum adjuvants enhances the immune response by facilitating phagocytosis and slowing the diffusion of antigens from the injection site which allows time for inflammatory cells to accumulate. The adsorptive strength is important as high affinity interactions interfere with the immune response. Adsorption can also affect the physical and chemical stability of antigens. Aluminum adjuvants activate dendritic cells via direct and indirect mechanisms. Phagocytosis of aluminum adjuvants followed by disruption of the phagolysosome activates NLRP3-inflammasomes resulting in the release of active IL-1β and IL-18. Aluminum adjuvants also activate dendritic cells by binding to membrane lipid rafts. Injection of aluminum-adjuvanted vaccines causes the release of uric acid, DNA, and ATP from damaged cells which in turn activate dendritic cells. The use of aluminum adjuvant is limited by weak stimulation of cell-mediated immunity. This can be enhanced by addition of other immunomodulatory molecules. Adsorption of these molecules is determined by the same mechanisms that control adsorption of antigens and can affect the efficacy of such combination adjuvants. The widespread use of aluminum adjuvants can be attributed in part to the excellent safety record based on a 70-year history of use. They cause local inflammation at the injection site, but also reduce the severity of systemic and local reactions by binding biologically active molecules in vaccines.
Collapse
Affiliation(s)
- Harm Hogenesch
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University West Lafayette, IN, USA
| |
Collapse
|
32
|
|
33
|
Borba EF, Saad CGS, Pasoto SG, Calich ALG, Aikawa NE, Ribeiro ACM, Moraes JCB, Leon EP, Costa LP, Guedes LKN, Silva CAA, Goncalves CR, Fuller R, Oliveira SA, Ishida MA, Precioso AR, Bonfa E. Influenza A/H1N1 vaccination of patients with SLE: can antimalarial drugs restore diminished response under immunosuppressive therapy? Rheumatology (Oxford) 2012; 51:1061-9. [PMID: 22298793 DOI: 10.1093/rheumatology/ker427] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE To assess the efficacy and safety of pandemic 2009 influenza A (H1N1) in SLE under different therapeutic regimens. METHODS A total of 555 SLE patients and 170 healthy controls were vaccinated with a single dose of a non-adjuvanted preparation. According to current therapy, patients were initially classified as SLE No Therapy (n = 75) and SLE with Therapy (n = 480). Subsequent evaluations included groups under monotherapy: chloroquine (CQ) (n = 105), prednisone (PRED) ≥20 mg (n = 76), immunosuppressor (IS) (n = 95) and those with a combination of these drugs. Anti-H1N1 titres and seroconversion (SC) rate were evaluated at entry and 21 days post-vaccination. RESULTS The SLE with Therapy group had lower SC compared with healthy controls (59.0 vs 80.0%; P < 0.0001), whereas the SLE No Therapy group had equivalent SC (72 vs 80.0%; P = 0.18) compared with healthy controls. Further comparison revealed that the SC of SLE No Therapy (72%) was similar to the CQ group (69.5%; P = 0.75), but it was significantly reduced in PRED ≥20 mg (53.9%; P = 0.028), IS (55.7%; P = 0.035) and PRED ≥20 mg + IS (45.4%; P = 0.038). The concomitant use of CQ in each of these later regimens was associated with SC responses comparable with SLE No Therapy group (72%): PRED ≥20 mg + CQ (71.4%; P = 1.00), IS + CQ (65.2%; P = 0.54) and PRED ≥20 mg + IS + CQ (57.4%; P = 0.09). CONCLUSION Pandemic influenza A H1N1/2009 vaccine response is diminished in SLE under immunosuppressive therapy and antimalarials seems to restore this immunogenicity. Trial registration. www.clinicaltrials.gov, NCT01151644.
Collapse
Affiliation(s)
- Eduardo F Borba
- Faculdade de Medicina da Universidade de São Paulo, Reumatologia Av. Dr Arnaldo, n 455, 3 andar, sala 3190, São Paulo 05403-010, Brazil.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Rettig L, Seidenberg S, Parvanova I, Samaras P, Knuth A, Pascolo S. Gemcitabine depletes regulatory T-cells in human and mice and enhances triggering of vaccine-specific cytotoxic T-cells. Int J Cancer 2011; 129:832-8. [DOI: 10.1002/ijc.25756] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2010] [Accepted: 10/11/2010] [Indexed: 01/08/2023]
|
35
|
Chen J, Li Z, Huang H, Yang Y, Ding Q, Mai J, Guo W, Xu Y. Improved antigen cross-presentation by polyethyleneimine-based nanoparticles. Int J Nanomedicine 2011; 6:77-84. [PMID: 21289984 PMCID: PMC3025594 DOI: 10.2147/ijn.s15457] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Purpose In the development of therapeutic vaccines against cancer, it is important to design strategies for antigen cross-presentation to stimulate cell-mediated immune responses against tumor antigens. Methods We developed a polyethyleneimine (PEI)-based protein antigen delivery system to promote cross-presentation through the major histocompatibility complex (MHC) I pathway using ovalbumin (OVA) as a model antigen. PEIs formed nanoparticles with OVA by electrostatic interactions, as demonstrated by electrophoresis analysis, scanning electron microscopy, and photon correlation spectroscopy analysis. Results The nanoparticles were used to stimulate mouse bone marrow-derived dendritic cells in vitro and resulted in significantly more OVA257–264/MHC I complex presentation on dendritic cell surfaces. The activated dendritic cells interacted specifically with RF33.70 to stimulate interleukin-2 secretion. The cross-presentation promoting effect was more prominent in dendritic cells that had been cultured for longer periods of time (13 days). Further studies comparing the antigen presentation efficacies by other polyanionic agents, such as PLL or lysosomotropic agents, suggested that the unique “proton sponge effect” of PEI facilitated antigen escape from the endosome toward the MHC I pathway. Conclusion Such a PEI-based nanoparticle system may have the potential to be developed into an effective therapeutic vaccine delivery system.
Collapse
Affiliation(s)
- Jian Chen
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
36
|
IFN-{gamma} produced by CD8 T cells induces T-bet-dependent and -independent class switching in B cells in responses to alum-precipitated protein vaccine. Proc Natl Acad Sci U S A 2010; 107:17292-7. [PMID: 20855629 DOI: 10.1073/pnas.1004879107] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Alum-precipitated protein (alum protein) vaccines elicit long-lasting neutralizing antibody responses that prevent bacterial exotoxins and viruses from entering cells. Typically, these vaccines induce CD4 T cells to become T helper 2 (Th2) cells that induce Ig class switching to IgG1. We now report that CD8 T cells also respond to alum proteins, proliferating extensively and producing IFN-γ, a key Th1 cytokine. These findings led us to question whether adoptive transfer of antigen-specific CD8 T cells alters the characteristic CD4 Th2 response to alum proteins and the switching pattern in responding B cells. To this end, WT mice given transgenic ovalbumin (OVA)-specific CD4 (OTII) or CD8 (OTI) T cells, or both, were immunized with alum-precipitated OVA. Cotransfer of antigen-specific CD8 T cells skewed switching patterns in responding B cells from IgG1 to IgG2a and IgG2b. Blocking with anti-IFN-γ antibody largely inhibited this altered B-cell switching pattern. The transcription factor T-bet is required in B cells for IFN-γ-dependent switching to IgG2a. By contrast, we show that this transcription factor is dispensable in B cells both for IFN-γ-induced switching to IgG2b and for inhibition of switching to IgG1. Thus, T-bet dependence identifies distinct transcriptional pathways in B cells that regulate IFN-γ-induced switching to different IgG isotypes.
Collapse
|
37
|
Empowering malaria vaccination by drug administration. Curr Opin Immunol 2010; 22:367-73. [DOI: 10.1016/j.coi.2010.04.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2010] [Revised: 03/23/2010] [Accepted: 04/06/2010] [Indexed: 11/18/2022]
|
38
|
Yewdell JW. Designing CD8+ T cell vaccines: it's not rocket science (yet). Curr Opin Immunol 2010; 22:402-10. [PMID: 20447814 PMCID: PMC2908899 DOI: 10.1016/j.coi.2010.04.002] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Accepted: 04/12/2010] [Indexed: 01/09/2023]
Abstract
CD8+ T cells play important roles in clearing viral infections and eradicating tumors. Designing vaccines that elicit effective CD8+ T cell responses requires a thorough knowledge of the pathways of antigen presentation in vivo. Here, I review recent progress in understanding the activation of naïve CD8+ T cells in vivo, with particular emphasis on cross-priming, the presentation of protein antigens acquired by dendritic cells from their environment. With the rapid advances in this area of research, the dawn of rational vaccine design is at hand.
Collapse
|
39
|
IL-4 directs both CD4 and CD8 T cells to produce Th2 cytokines in vitro, but only CD4 T cells produce these cytokines in response to alum-precipitated protein in vivo. Mol Immunol 2010; 47:1914-22. [PMID: 20392496 DOI: 10.1016/j.molimm.2010.03.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2010] [Revised: 03/15/2010] [Accepted: 03/16/2010] [Indexed: 11/19/2022]
Abstract
While IL-4 directs CD4 T cells to produce Th2 cytokines (including IL-4, IL-13, IL-5) in vitro it has been shown that production of these cytokines can be induced in vivo in the absence of IL-4/IL-13/STAT-6 signaling. The present report shows that CD8 as well as CD4 T cells activated through their TCR, in vitro upregulate the Th2-features - IL-4, IL-13, IL-5, and GATA-3. However, in vivo while alum-precipitated antigen strongly and selectively induces these Th2-features in CD4 T cells, CD8 T cells mount a markedly different response to this antigen. This CD8 response is associated with strong proliferation and production of IFN-gamma, but no Th2-features are induced. Alum-protein formulations are widely used in human vaccines and typically induce strong antibody responses characterized by the differentiation of IL-4-producing CD4 T cells and immunoglobulin class switching to IgG1. Nevertheless, the mechanism responsible for CD4 Th2 and follicular helper T cell commitment triggered by these alum-protein vaccines is still poorly understood. Analysis of the in vivo response to alum-precipitated protein shows that while subsets of CD4 T cells strongly upregulate Th2 and follicular helper T cell features including the surface markers OX40, CXCR5, PD-1, IL-17RB and the transcription factor c-Maf, CD8 T cells do not. These discrete differences between responding CD4 and CD8 T cells provide further insight into the differences between Th2 polarization of CD4 T cells directed by IL-4 in vitro and the induction of IL-4 production by CD4 T cells in vivo in response to alum-precipitated protein.
Collapse
|
40
|
Bozzacco L, Trumpfheller C, Huang Y, Longhi MP, Shimeliovich I, Schauer JD, Park CG, Steinman RM. HIV gag protein is efficiently cross-presented when targeted with an antibody towards the DEC-205 receptor in Flt3 ligand-mobilized murine DC. Eur J Immunol 2010; 40:36-46. [PMID: 19830741 DOI: 10.1002/eji.200939748] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
DC present exogenous proteins to MHC class I-restricted CD8+ T cells. This function does not require endogenous antigen synthesis within DC, providing the potential to elicit CD8+ T-cell responses to immune complexes, inactivated microbes, dying cells, and proteins such as OVA. In mice, the CD8+ or DEC-205+ DC are specialized for cross-presentation, and this subset can be increased 10-fold in numbers following Fms-like tyrosine kinase 3 ligand (Flt3L) treatment in vivo. Therefore, we studied cross-presentation by abundant Flt3L DC using HIV gag protein. When enriched by positive selection with anti-CD11c beads, cells from Flt3L mice are not only more abundant but are also more highly enriched in CD11chigh DC, particularly the DEC-205+ subset. DC cross-present HIV gag to primed CD8+ T cells, but when the antigen is delivered within an antibody to DEC-205 receptor, cross-presentation becomes 100-fold more efficient than non-targeted antigen. This finding requires gag to be engineered into anti-DEC antibody, not just mixed with antibody. Flt3L DC are a valuable tool to study cross-presentation, since their use overcomes the obstacle posed by the low number of cross-presenting DC in the steady state. These findings support future experiments to use Flt3L to enhance presentation of DC-targeted vaccines.
Collapse
Affiliation(s)
- Leonia Bozzacco
- Laboratory of Cellular Physiology and Immunology and Chris Browne Center, The Rockefeller University, New York, NY 10065-6399, USA
| | | | | | | | | | | | | | | |
Collapse
|