1
|
Zhao Z, Meng Q, Sun TZ, Zhu B. Mannose modified targeted immersion vaccine delivery system improves protective immunity against Infectious spleen and kidney necrosis virus in mandarin fish (Siniperca chuatsi). Vaccine 2024; 42:2886-2894. [PMID: 38519342 DOI: 10.1016/j.vaccine.2024.03.047] [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: 08/12/2022] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 03/24/2024]
Abstract
Vaccination is an effective method to prevent viral diseases. However, the biological barrier prevents the immersion vaccine from achieving the best effect without adding adjuvants and carriers. Researches on the targeted presentation technology of vaccines with nanocarriers are helpful to develop immersion vaccines for fish that can break through biological barriers and play an effective role in fish defense. In our study, functionally modified single-walled carbon nanotubes (SWCNTs) were used as carriers to construct a targeted immersion vaccine (SWCNTs-M-MCP) with mannose modified major capsid protein (MCP) to target antigen-presenting cells (APCs), against iridovirus diseases. After bath immunization, our results showed that SWCNTs-M-MCP induced the presentation process and uptake of APCs, triggering a powerful immune response. Moreover, the highest relative percent survival (RPS) was 81.3% in SWCNTs-M-MCP group, which was only 41.5% in SWCNTs-MCP group. Altogether, this study indicates that the SWCNTs-based targeted immersion vaccine induces strong immune response and provided an effective protection against iridovirus diseases.
Collapse
Affiliation(s)
- Zhao Zhao
- College of Animal Science and Technology, Northwest A&F University, Xinong Road 22nd, Yangling, Shaanxi 712100, PR China
| | - Qiang Meng
- Shenzhen Vaccine Biotechnology Co., Ltd, B618, Virtual University Experimental Platform Building, Nanshan District, Shenzhen 518000, PR China
| | - Tian-Zi Sun
- College of Animal Science and Technology, Northwest A&F University, Xinong Road 22nd, Yangling, Shaanxi 712100, PR China
| | - Bin Zhu
- College of Animal Science and Technology, Northwest A&F University, Xinong Road 22nd, Yangling, Shaanxi 712100, PR China.
| |
Collapse
|
2
|
Freitas R, Peixoto A, Ferreira E, Miranda A, Santos LL, Ferreira JA. Immunomodulatory glycomedicine: Introducing next generation cancer glycovaccines. Biotechnol Adv 2023; 65:108144. [PMID: 37028466 DOI: 10.1016/j.biotechadv.2023.108144] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 03/17/2023] [Accepted: 03/30/2023] [Indexed: 04/09/2023]
Abstract
Cancer remains a leading cause of death worldwide due to the lack of safer and more effective therapies. Cancer vaccines developed from neoantigens are an emerging strategy to promote protective and therapeutic anti-cancer immune responses. Advances in glycomics and glycoproteomics have unveiled several cancer-specific glycosignatures, holding tremendous potential to foster effective cancer glycovaccines. However, the immunosuppressive nature of tumours poses a major obstacle to vaccine-based immunotherapy. Chemical modification of tumour associated glycans, conjugation with immunogenic carriers and administration in combination with potent immune adjuvants constitute emerging strategies to address this bottleneck. Moreover, novel vaccine vehicles have been optimized to enhance immune responses against otherwise poorly immunogenic cancer epitopes. Nanovehicles have shown increased affinity for antigen presenting cells (APCs) in lymph nodes and tumours, while reducing treatment toxicity. Designs exploiting glycans recognized by APCs have further enhanced the delivery of antigenic payloads, improving glycovaccine's capacity to elicit innate and acquired immune responses. These solutions show potential to reduce tumour burden, while generating immunological memory. Building on this rationale, we provide a comprehensive overview on emerging cancer glycovaccines, emphasizing the potential of nanotechnology in this context. A roadmap towards clinical implementation is also delivered foreseeing advances in glycan-based immunomodulatory cancer medicine.
Collapse
Affiliation(s)
- Rui Freitas
- Experimental Pathology and Therapeutics Group, IPO Porto Research Center (CI-IPOP), RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute (IPO Porto), 4200-072 Porto, Portugal; Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal; Porto Comprehensive Cancer Center (P.ccc), 4200-072 Porto, Portugal; Abel Salazar Biomedical Sciences Institute - University of Porto (ICBAS), 4050-313 Porto, Portugal
| | - Andreia Peixoto
- Experimental Pathology and Therapeutics Group, IPO Porto Research Center (CI-IPOP), RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute (IPO Porto), 4200-072 Porto, Portugal; Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal; Porto Comprehensive Cancer Center (P.ccc), 4200-072 Porto, Portugal
| | - Eduardo Ferreira
- Experimental Pathology and Therapeutics Group, IPO Porto Research Center (CI-IPOP), RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute (IPO Porto), 4200-072 Porto, Portugal
| | - Andreia Miranda
- Experimental Pathology and Therapeutics Group, IPO Porto Research Center (CI-IPOP), RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute (IPO Porto), 4200-072 Porto, Portugal; Abel Salazar Biomedical Sciences Institute - University of Porto (ICBAS), 4050-313 Porto, Portugal
| | - Lúcio Lara Santos
- Experimental Pathology and Therapeutics Group, IPO Porto Research Center (CI-IPOP), RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute (IPO Porto), 4200-072 Porto, Portugal; Porto Comprehensive Cancer Center (P.ccc), 4200-072 Porto, Portugal; Abel Salazar Biomedical Sciences Institute - University of Porto (ICBAS), 4050-313 Porto, Portugal; Health School of University Fernando Pessoa, 4249-004 Porto, Portugal; GlycoMatters Biotech, 4500-162 Espinho, Portugal; Department of Surgical Oncology, Portuguese Oncology Institute (IPO Porto), 4200-072 Porto, Portugal
| | - José Alexandre Ferreira
- Experimental Pathology and Therapeutics Group, IPO Porto Research Center (CI-IPOP), RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute (IPO Porto), 4200-072 Porto, Portugal; Porto Comprehensive Cancer Center (P.ccc), 4200-072 Porto, Portugal; GlycoMatters Biotech, 4500-162 Espinho, Portugal.
| |
Collapse
|
3
|
Galati D, Zanotta S, Capitelli L, Bocchino M. A bird's eye view on the role of dendritic cells in SARS‐CoV‐2 infection: Perspectives for immune‐based vaccines. Allergy 2022. [DOI: 10.1111/all.15004
expr 869230256 + 930548950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Affiliation(s)
- Domenico Galati
- Hematology‐Oncology and Stem Cell Transplantation Unit Department of Hematology and Developmental Therapeutics Istituto Nazionale Tumori‐ IRCCS‐ Fondazione G. Pascale Napoli Italy
| | - Serena Zanotta
- Hematology‐Oncology and Stem Cell Transplantation Unit Department of Hematology and Developmental Therapeutics Istituto Nazionale Tumori‐ IRCCS‐ Fondazione G. Pascale Napoli Italy
| | - Ludovica Capitelli
- Department of Clinical Medicine and Surgery Università degli Studi di Napoli Federico II Napoli Italy
| | - Marialuisa Bocchino
- Department of Clinical Medicine and Surgery Università degli Studi di Napoli Federico II Napoli Italy
| |
Collapse
|
4
|
Galati D, Zanotta S, Capitelli L, Bocchino M. A bird's eye view on the role of dendritic cells in SARS-CoV-2 infection: Perspectives for immune-based vaccines. Allergy 2022; 77:100-110. [PMID: 34245591 PMCID: PMC8441836 DOI: 10.1111/all.15004] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 07/05/2021] [Indexed: 12/15/2022]
Abstract
Coronavirus disease-19 (COVID-19) is a complex disorder caused by the pandemic diffusion of a novel coronavirus named SARS-CoV-2. Clinical manifestations vary from silent infection to severe pneumonia, disseminated thrombosis, multi-organ failure, and death. COVID-19 pathogenesis is still not fully elucidated, while increasing evidence suggests that disease phenotypes are strongly related to the virus-induced immune system's dysregulation. Indeed, when the virus-host cross talk is out of control, the occurrence of an aberrant systemic inflammatory reaction, named "cytokine storm," leads to a detrimental impairment of the adaptive immune response. Dendritic cells (DCs) are the most potent antigen-presenting cells able to support innate immune and promote adaptive responses. Besides, DCs play a key role in the anti-viral defense. The aim of this review is to focus on DC involvement in SARS-CoV-2 infection to better understand pathogenesis and clinical behavior of COVID-19 and explore potential implications for immune-based therapy strategies.
Collapse
Affiliation(s)
- Domenico Galati
- Hematology‐Oncology and Stem Cell Transplantation UnitDepartment of Hematology and Developmental TherapeuticsIstituto Nazionale Tumori‐ IRCCS‐ Fondazione G. PascaleNapoliItaly
| | - Serena Zanotta
- Hematology‐Oncology and Stem Cell Transplantation UnitDepartment of Hematology and Developmental TherapeuticsIstituto Nazionale Tumori‐ IRCCS‐ Fondazione G. PascaleNapoliItaly
| | - Ludovica Capitelli
- Department of Clinical Medicine and SurgeryUniversità degli Studi di Napoli Federico IINapoliItaly
| | - Marialuisa Bocchino
- Department of Clinical Medicine and SurgeryUniversità degli Studi di Napoli Federico IINapoliItaly
| |
Collapse
|
5
|
Dhodapkar MV, Dhodapkar KM. Tissue-resident memory-like T cells in tumor immunity: Clinical implications. Semin Immunol 2020; 49:101415. [PMID: 33011063 DOI: 10.1016/j.smim.2020.101415] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 09/06/2020] [Accepted: 09/21/2020] [Indexed: 12/13/2022]
Abstract
Tissue-resident memory (TRM) T cells are distinct population of non-circulating lymphocytes that play an important role in mediating regional immunity. TRM- like cells have now been identified as a component of tumor-infiltrating lymphocytes in several human tumors and correlate with outcome and response to immunotherapy. TRM cells have also been shown to mediate anti-tumor immunity in murine models. Biology of TRM cells has several implications for clinical cancer immunotherapy. Here we discuss newer insights into the biology of TRM T cells and discuss their implications for understanding the heterogeneity of immune microenvironment in tumors as well as improving the efficacy of cancer vaccines, immune-checkpoint blockade and adoptive cellular therapies in the clinic.
Collapse
Affiliation(s)
- Madhav V Dhodapkar
- Department of Hematology / Medical Oncology, Emory University, Atlanta, GA, United States; Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | - Kavita M Dhodapkar
- Winship Cancer Institute, Emory University, Atlanta, GA, United States; Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University, Atlanta, GA, United States.
| |
Collapse
|
6
|
Al-Hatamleh MAI, E.A.R. ENS, Boer JC, Ferji K, Six JL, Chen X, Elkord E, Plebanski M, Mohamud R. Synergistic Effects of Nanomedicine Targeting TNFR2 and DNA Demethylation Inhibitor-An Opportunity for Cancer Treatment. Cells 2019; 9:E33. [PMID: 31877663 PMCID: PMC7016661 DOI: 10.3390/cells9010033] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 11/17/2019] [Accepted: 11/20/2019] [Indexed: 12/31/2022] Open
Abstract
Tumor necrosis factor receptor 2 (TNFR2) is expressed on some tumor cells, such as myeloma, Hodgkin lymphoma, colon cancer and ovarian cancer, as well as immunosuppressive cells. There is increasingly evidence that TNFR2 expression in cancer microenvironment has significant implications in cancer progression, metastasis and immune evasion. Although nanomedicine has been extensively studied as a carrier of cancer immunotherapeutic agents, no study to date has investigated TNFR2-targeting nanomedicine in cancer treatment. From an epigenetic perspective, previous studies indicate that DNA demethylation might be responsible for high expressions of TNFR2 in cancer models. This perspective review discusses a novel therapeutic strategy based on nanomedicine that has the capacity to target TNFR2 along with inhibition of DNA demethylation. This approach may maximize the anti-cancer potential of nanomedicine-based immunotherapy and, consequently, markedly improve the outcomes of the management of patients with malignancy.
Collapse
Affiliation(s)
- Mohammad A. I. Al-Hatamleh
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kelantan, Malaysia;
| | - Engku Nur Syafirah E.A.R.
- Department of Medical Microbiology and Parasitology, School of Medical Sciences, Universiti Sains Malaysia, Kelantan 16150, Malaysia;
| | - Jennifer C. Boer
- Translational Immunology and Nanotechnology Unit, School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Australia (M.P.)
| | - Khalid Ferji
- Université de Lorraine, CNRS, LCPM, F-5400 Nancy, France; (K.F.); (J.-L.S.)
| | - Jean-Luc Six
- Université de Lorraine, CNRS, LCPM, F-5400 Nancy, France; (K.F.); (J.-L.S.)
| | - Xin Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences. University of Macau, Macao 999078, China
| | - Eyad Elkord
- Cancer Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, 34110 Doha, Qatar;
| | - Magdalena Plebanski
- Translational Immunology and Nanotechnology Unit, School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Australia (M.P.)
| | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kelantan, Malaysia;
- Hospital Universiti Sains Malaysia, Universiti Sains Malaysia, Kelantan 16150, Malaysia
| |
Collapse
|
7
|
Dhodapkar MV, Dhodapkar KM. Moving Immunoprevention Beyond Virally Mediated Malignancies: Do We Need to Link It to Early Detection? Front Immunol 2019; 10:2385. [PMID: 31649683 PMCID: PMC6795703 DOI: 10.3389/fimmu.2019.02385] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 09/23/2019] [Indexed: 12/23/2022] Open
Abstract
Vaccines can successfully prevent viral infections and have emerged as an effective strategy for preventing some virally mediated malignancies. They also represent our major hope for cost-effective reduction of the cancer burden. The concept that the immune system mediates surveillance and editing roles against tumors is now well-established in murine models. However, harnessing the immune system to prevent human cancers that do not have a known viral etiology has not yet been realized. Most human cancers originate in a premalignant phase that is more common than the cancer itself. Many of the genetic changes that underlie carcinogenesis originate at this stage when the malignant phenotype is not manifest. Studies evaluating host response in human premalignancy have documented that these lesions are immunogenic, setting the stage for immune-based approaches for targeted prevention of human cancer. However, recent studies suggest that the hierarchy of T cell exhaustion and immune-suppressive factors have already begun to emerge in many preneoplastic states. These considerations underscore the need to link immune prevention to earlier detection of such lesions and to personalize such approaches based on the status of the pre-existing immune response.
Collapse
Affiliation(s)
- Madhav V. Dhodapkar
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, United States
- Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | - Kavita M. Dhodapkar
- Winship Cancer Institute, Emory University, Atlanta, GA, United States
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center of Children's Healthcare of Atlanta, School of Medicine, Emory University, Atlanta, GA, United States
| |
Collapse
|
8
|
Li Y, Ayala-Orozco C, Rauta PR, Krishnan S. The application of nanotechnology in enhancing immunotherapy for cancer treatment: current effects and perspective. NANOSCALE 2019; 11:17157-17178. [PMID: 31531445 PMCID: PMC6778734 DOI: 10.1039/c9nr05371a] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Cancer immunotherapy is emerging as a promising treatment modality that suppresses and eliminates tumors by re-activating and maintaining the tumor-immune cycle, and further enhancing the body's anti-tumor immune response. Despite the impressive therapeutic potential of immunotherapy approaches such as immune checkpoint inhibitors and tumor vaccines in pre-clinical and clinical applications, the effective response is limited by insufficient accumulation in tumor tissues and severe side-effects. Recent years have witnessed the rise of nanotechnology as a solution to improve these technical weaknesses due to its inherent biophysical properties and multifunctional modifying potential. In this review, we summarized and discussed the current status of nanoparticle-enhanced cancer immunotherapy strategies, including intensified delivery of tumor vaccines and immune adjuvants, immune checkpoint inhibitor vehicles, targeting capacity to tumor-draining lymph nodes and immune cells, triggered releasing and regulating specific tumor microenvironments, and adoptive cell therapy enhancement effects.
Collapse
Affiliation(s)
- Yongjiang Li
- Department of Medical Oncology, Cancer Center, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China. and Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA.
| | - Ciceron Ayala-Orozco
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA.
| | - Pradipta Ranjan Rauta
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA.
| | - Sunil Krishnan
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA. and Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
9
|
Savitsky K, Yu X. Combined strategies for tumor immunotherapy with nanoparticles. Clin Transl Oncol 2019; 21:1441-1449. [PMID: 31055713 DOI: 10.1007/s12094-019-02081-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 03/05/2019] [Indexed: 12/20/2022]
Abstract
A brief review of tumor immunotherapies shows significant advancements in academic research and preclinical studies. Analysis of different immune cell pathways, including macrophage activation, natural killer cells, and dendritic cell presentation show promising clinical results when targeted with different nanoparticle polymer and gold materials. Following a brief discussion on immuno-oncology successes, detailed results are discussed in macrophage activation, dendritic cell presentation, and lysis of tumor cells with natural killer cells. Common targets include tumor-associated macrophages and induction of the proinflammatory phenotype, dual targeting of cell and humoral immunity with dendritic cells, and creating chimeric antigen receptors on natural killer cells. An analysis of the results shows a variety of nanoparticle synthesis methods are required depending on drug type and tissue type affected by tumors. Future research is discussed in conjunction with a brief analysis of completed clinical trial data on cancer therapies using nanoparticles to date. Although paclitaxel-loaded albumin nanoparticles are most frequently studied, academic research shows there may be additional mechanisms of action to elicit anti-tumor activity.
Collapse
Affiliation(s)
- K Savitsky
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, USA.
| | - X Yu
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, USA.
| |
Collapse
|
10
|
Pati R, Shevtsov M, Sonawane A. Nanoparticle Vaccines Against Infectious Diseases. Front Immunol 2018; 9:2224. [PMID: 30337923 PMCID: PMC6180194 DOI: 10.3389/fimmu.2018.02224] [Citation(s) in RCA: 283] [Impact Index Per Article: 47.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 09/07/2018] [Indexed: 12/13/2022] Open
Abstract
Due to emergence of new variants of pathogenic micro-organisms the treatment and immunization of infectious diseases have become a great challenge in the past few years. In the context of vaccine development remarkable efforts have been made to develop new vaccines and also to improve the efficacy of existing vaccines against specific diseases. To date, some vaccines are developed from protein subunits or killed pathogens, whilst several vaccines are based on live-attenuated organisms, which carry the risk of regaining their pathogenicity under certain immunocompromised conditions. To avoid this, the development of risk-free effective vaccines in conjunction with adequate delivery systems are considered as an imperative need to obtain desired humoral and cell-mediated immunity against infectious diseases. In the last several years, the use of nanoparticle-based vaccines has received a great attention to improve vaccine efficacy, immunization strategies, and targeted delivery to achieve desired immune responses at the cellular level. To improve vaccine efficacy, these nanocarriers should protect the antigens from premature proteolytic degradation, facilitate antigen uptake and processing by antigen presenting cells, control release, and should be safe for human use. Nanocarriers composed of lipids, proteins, metals or polymers have already been used to attain some of these attributes. In this context, several physico-chemical properties of nanoparticles play an important role in the determination of vaccine efficacy. This review article focuses on the applications of nanocarrier-based vaccine formulations and the strategies used for the functionalization of nanoparticles to accomplish efficient delivery of vaccines in order to induce desired host immunity against infectious diseases.
Collapse
Affiliation(s)
| | - Maxim Shevtsov
- Institute of Cytology of the Russian Academy of Sciences (RAS), St. Petersburg, Russia
- Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
- First Pavlov State Medical University of St.Petersburg, St. Petersburg, Russia
| | - Avinash Sonawane
- School of Biotechnology, KIIT University, Bhubaneswar, India
- Discipline of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India
| |
Collapse
|
11
|
Harrison EB, Azam SH, Pecot CV. Targeting Accessories to the Crime: Nanoparticle Nucleic Acid Delivery to the Tumor Microenvironment. Front Pharmacol 2018; 9:307. [PMID: 29670528 PMCID: PMC5893903 DOI: 10.3389/fphar.2018.00307] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 03/16/2018] [Indexed: 12/18/2022] Open
Abstract
Nucleic acid delivery for cancer holds extraordinary promise. Increasing expression of tumor suppressor genes or inhibition of oncogenes in cancer cells has important therapeutic potential. However, several barriers impair progress in cancer gene delivery. These include effective delivery to cancer cells and relevant intracellular compartments. Although viral gene delivery can be effective, it has the disadvantages of being immuno-stimulatory, potentially mutagenic and lacking temporal control. Various nanoparticle (NP) platforms have been developed to overcome nucleic acid delivery hurdles, but several challenges still exist. One such challenge has been the accumulation of NPs in non-cancer cells within the tumor microenvironment (TME) as well as the circulation. While uptake by these cancer-associated cells is considered to be an off-target effect in some contexts, several strategies have now emerged to utilize NP-mediated gene delivery to intentionally alter the TME. For example, the similarity of NPs in shape and size to pathogens promotes uptake by antigen presenting cells, which can be used to increase immune stimulation and promote tumor killing by T-lymphocytes. In the era of immunotherapy, boosting the ability of the immune system to eliminate cancer cells has proven to be an exciting new area in cancer nanotechnology. Given the importance of cancer-associated cells in tumor growth and metastasis, targeting these cells in the TME opens up new therapeutic applications for NPs. This review will cover evidence for non-cancer cell accumulation of NPs in animal models and patients, summarize characteristics that promote NP delivery to different cell types, and describe several therapeutic strategies for gene modification within the TME.
Collapse
Affiliation(s)
- Emily B. Harrison
- Center for Nanotechnology in Drug Delivery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Salma H. Azam
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Chad V. Pecot
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Division of Hematology/Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
12
|
Snapper CM. Distinct Immunologic Properties of Soluble Versus Particulate Antigens. Front Immunol 2018; 9:598. [PMID: 29619034 PMCID: PMC5871672 DOI: 10.3389/fimmu.2018.00598] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 03/09/2018] [Indexed: 12/14/2022] Open
Abstract
Antigens in particulate form have distinct immunologic properties relative to soluble antigens. An understanding of the mechanisms and functional consequences of the distinct immunologic pathways engaged by these different forms of antigen is particularly relevant to the design of vaccines. It is also relevant regarding the use of therapeutic human proteins in clinical medicine that have been shown to aggregate, and perhaps as a result, elicit autoantibodies.
Collapse
Affiliation(s)
- Clifford M Snapper
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
13
|
van Dinther D, Stolk DA, van de Ven R, van Kooyk Y, de Gruijl TD, den Haan JMM. Targeting C-type lectin receptors: a high-carbohydrate diet for dendritic cells to improve cancer vaccines. J Leukoc Biol 2017; 102:1017-1034. [PMID: 28729358 PMCID: PMC5597514 DOI: 10.1189/jlb.5mr0217-059rr] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 06/13/2017] [Accepted: 06/16/2017] [Indexed: 12/23/2022] Open
Abstract
There is a growing understanding of why certain patients do or do not respond to checkpoint inhibition therapy. This opens new opportunities to reconsider and redevelop vaccine strategies to prime an anticancer immune response. Combination of such vaccines with checkpoint inhibitors will both provide the fuel and release the brake for an efficient anticancer response. Here, we discuss vaccine strategies that use C-type lectin receptor (CLR) targeting of APCs, such as dendritic cells and macrophages. APCs are a necessity for the priming of antigen-specific cytotoxic and helper T cells. Because CLRs are natural carbohydrate-recognition receptors highly expressed by multiple subsets of APCs and involved in uptake and processing of Ags for presentation, these receptors seem particularly interesting for targeting purposes.
Collapse
Affiliation(s)
- Dieke van Dinther
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands; and
| | - Dorian A Stolk
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands; and
| | - Rieneke van de Ven
- Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Yvette van Kooyk
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands; and
| | - Tanja D de Gruijl
- Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Joke M M den Haan
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands; and
| |
Collapse
|
14
|
Askarizadeh A, Jaafari MR, Khamesipour A, Badiee A. Liposomal adjuvant development for leishmaniasis vaccines. THERAPEUTIC ADVANCES IN VACCINES 2017; 5:85-101. [PMID: 29201374 PMCID: PMC5697592 DOI: 10.1177/2051013617741578] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 10/18/2017] [Indexed: 08/29/2023]
Abstract
Leishmaniasis is a parasitic disease that ranges in severity from skin lesions to fatality. Since long-lasting protection is induced upon recovery from cutaneous leishmaniasis, development of an effective vaccine is promising. However, there is no vaccine for use in humans yet. It seems limited efficacy in leishmaniasis vaccines is due to lack of an appropriate adjuvant or delivery system. Hence, the use of particulate adjuvants such as liposomes for effective delivery to the antigen presenting cells (APCs) is a valuable strategy to enhance leishmaniasis vaccine efficacy. The extraordinary versatility of liposomes because of their unique amphiphilic and biphasic nature allows for using antigens or immunostimulators within the core, on the surface or within the bilayer, and modulates both the magnitude and the T-helper bias of the immune response. In this review article, we attempt to summarize the role of liposomal adjuvants in the development of Leishmania vaccines and describe the main physicochemical properties of liposomes like phospholipid composition, surface charge, and particle size during formulation design. We also suggest potentially useful formulation strategies in order for future experiments to have a chance to succeed as liposomal vaccines against leishmaniasis.
Collapse
Affiliation(s)
- Anis Askarizadeh
- Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran Department of Pharmaceutical Nanotechnology, School of Pharmacy Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Khamesipour
- Center for Research and Training in Skin Diseases and Leprosy, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Badiee
- Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
15
|
Abstract
Background Immunotherapy consists of activating the patient’s immune system to fight cancer and has the great potential of preventing future relapses thanks to immunological memory. A great variety of strategies have emerged to harness the immune system against tumors, from the administration of immunomodulatory agents that activate immune cells, to therapeutic vaccines or infusion of previously activated cancer-specific T cells. However, despite great recent progress many difficulties still remain, which prevent the widespread use of immunotherapy. Some of these limitations include: systemic toxicity, weak immune cellular responses or persistence over time and most ultimately costly and time-consuming procedures. Main body Synthetic and natural biomaterials hold great potential to address these hurdles providing biocompatible systems capable of targeted local delivery, co-delivery, and controlled and/or sustained release. In this review we discuss some of the bioengineered solutions and approaches developed so far and how biomaterials can be further implemented to help and shape the future of cancer immunotherapy. Conclusion The bioengineering strategies here presented constitute a powerful toolkit to develop safe and successful novel cancer immunotherapies.
Collapse
|
16
|
Vasquez JC, Huttner A, Zhang L, Marks A, Chan A, Baehring JM, Kahle KT, Dhodapkar KM. SOX2 immunity and tissue resident memory in children and young adults with glioma. J Neurooncol 2017. [PMID: 28620836 DOI: 10.1007/s11060-017-2515-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Therapies targeting immune checkpoints are effective in tumors with a high mutation burden that express multiple neo-antigens. However, glial tumors including those seen in children carry fewer mutations and there is an unmet need to identify new antigenic targets of anti-tumor immunity. SOX2 is an embryonal stem cell antigen implicated in the biology of glioma initiating cells. Expression of SOX2 by pediatric glial tumors and the capacity of the immune system in these patients to recognize SOX2 has not been previously studied. We examined the expression of SOX2 on archived paraffin-embedded tissue from pediatric glial tumors. The presence of T-cell immunity to SOX2 was examined in both blood and tumor-infiltrating T-cells in children and young adults with glioma. The nature of tumor-infiltrating immune cells was analyzed with a 37-marker panel using single-cell mass cytometry. SOX2 is expressed by tumor cells but not surrounding normal tissue in pediatric gliomas of all grades. T-cells against this antigen can be detected in blood and tumor tissue in glioma patients. Glial tumors are enriched for CD8/CD4 T-cells with tissue resident memory (TRM; CD45RO+, CD69+, CCR7-) phenotype, which co-express multiple inhibitory checkpoints including PD-1, PD-L1 and TIGIT. Tumors also contain natural killer cells with reduced expression of lytic granzyme. Our data demonstrate immunogenicity of SOX2, which is specifically overexpressed on pediatric glial tumor cells. Harnessing tumor immunity in glioma will likely require the combined targeting of multiple inhibitory checkpoints.
Collapse
Affiliation(s)
- Juan C Vasquez
- Department of Pediatrics, Yale School of Medicine, 333 Cedar Street, LMP 2073, New Haven, CT, 06510, USA
| | - Anita Huttner
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Lin Zhang
- Department of Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Asher Marks
- Department of Pediatrics, Yale School of Medicine, 333 Cedar Street, LMP 2073, New Haven, CT, 06510, USA
| | - Amy Chan
- Department of Medicine, Yale School of Medicine, New Haven, CT, USA
| | | | | | - Kavita M Dhodapkar
- Department of Pediatrics, Yale School of Medicine, 333 Cedar Street, LMP 2073, New Haven, CT, 06510, USA.
| |
Collapse
|
17
|
Balan S, Finnigan J, Bhardwaj N. Dendritic Cell Strategies for Eliciting Mutation-Derived Tumor Antigen Responses in Patients. Cancer J 2017; 23:131-137. [PMID: 28410301 PMCID: PMC5520811 DOI: 10.1097/ppo.0000000000000251] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Dendritic cells (DCs) are equipped for sensing danger signals and capturing, processing, and presenting antigens to naive or effector cells and are critical in inducing humoral and adaptive immunity. Successful vaccinations are those that activate DCs to elicit both cellular and humoral responses, as well as long-lasting memory response against the target of interest. Recently, it has become apparent that tumor cells can provide new sources of antigens through nonsynonymous mutations or frame-shift mutations, leading to potentially hundreds of mutation-derived tumor antigens (MTAs) or neoantigens. T cells recognizing MTA have been detected in cancer patients and can even lead to tumor regression. Designing MTA-specific vaccination strategies will have to take into account the adjuvant activity of DC subsets and the best formulation to elicit an effective immune response. We discuss the potential of human DCs to prime MTA-specific responses.
Collapse
Affiliation(s)
- Sreekumar Balan
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, NY NY
| | - John Finnigan
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, NY NY
| | - Nina Bhardwaj
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, NY NY
| |
Collapse
|
18
|
Qiu H, Min Y, Rodgers Z, Zhang L, Wang AZ. Nanomedicine approaches to improve cancer immunotherapy. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2017; 9. [PMID: 28296286 DOI: 10.1002/wnan.1456] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 10/28/2016] [Accepted: 12/17/2016] [Indexed: 01/10/2023]
Abstract
Significant advances have been made in the field of cancer immunotherapy by orchestrating the body's immune system to eradicate cancer cells. However, safety and efficacy concerns stemming from the systemic delivery of immunomodulatory compounds limits cancer immunotherapies expansion and application. In this context, nanotechnology presents a number of advantages, such as targeted delivery to immune cells, enhanced clinical outcomes, and reduced adverse events, which may aid in the delivery of cancer vaccines and immunomodulatory agents. With this in mind, a diverse range of nanomaterials with different physicochemical characteristics have been developed to stimulate the immune system and battle cancer. In this review, we will focus on some recent developments and the potential advantages of utilizing nanotechnology within the field of cancer immunotherapy. WIREs Nanomed Nanobiotechnol 2017, 9:e1456. doi: 10.1002/wnan.1456 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Hui Qiu
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine; Department of Radiation Oncology, University of North Carolina, Chapel Hill, NC, USA.,Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yuanzeng Min
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine; Department of Radiation Oncology, University of North Carolina, Chapel Hill, NC, USA
| | - Zach Rodgers
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine; Department of Radiation Oncology, University of North Carolina, Chapel Hill, NC, USA
| | - Longzhen Zhang
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Andrew Z Wang
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine; Department of Radiation Oncology, University of North Carolina, Chapel Hill, NC, USA.,Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
19
|
Grippin AJ, Sayour EJ, Mitchell DA. Translational nanoparticle engineering for cancer vaccines. Oncoimmunology 2017; 6:e1290036. [PMID: 29123947 PMCID: PMC5665077 DOI: 10.1080/2162402x.2017.1290036] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 01/23/2017] [Accepted: 01/26/2017] [Indexed: 01/03/2023] Open
Abstract
Conventional cancer treatments remain insufficient to treat many therapy-resistant tumors.1 Cancer vaccines attempt to overcome this resistance by activating the patient's immune system to eliminate tumor cells without the toxicity of systemic chemotherapy and radiation. Nanoparticles (NPs) are promising as customizable, immunostimulatory carriers to protect and deliver antigen. Although many NP vaccines have been investigated in preclinical settings, a few have advanced into clinical application, and still fewer have demonstrated clinical benefit. This review incorporates observations from NP vaccines that have been evaluated in early phase clinical trials to make recommendations for the next generation of NP-based cancer vaccines.
Collapse
Affiliation(s)
- Adam J Grippin
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida Brain Tumor Immunotherapy Program, McKnight Brain Institute, Department of Neurosurgery, University of Florida, Gainesville, FL, USA.,J. Crayton Pruitt Family Department of Biomedical Engineering, Biomedical Sciences Building, University of Florida, Gainesville, FL, USA
| | - Elias J Sayour
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida Brain Tumor Immunotherapy Program, McKnight Brain Institute, Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Duane A Mitchell
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida Brain Tumor Immunotherapy Program, McKnight Brain Institute, Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| |
Collapse
|
20
|
Fytianos K, Chortarea S, Rodriguez-Lorenzo L, Blank F, von Garnier C, Petri-Fink A, Rothen-Rutishauser B. Aerosol Delivery of Functionalized Gold Nanoparticles Target and Activate Dendritic Cells in a 3D Lung Cellular Model. ACS NANO 2017; 11:375-383. [PMID: 27973764 DOI: 10.1021/acsnano.6b06061] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Nanocarrier design combined with pulmonary drug delivery holds great promise for the treatment of respiratory tract disorders. In particular, targeting of dendritic cells that are key immune cells to enhance or suppress an immune response in the lung is a promising approach for the treatment of allergic diseases. Fluorescently encoded poly(vinyl alcohol) (PVA)-coated gold nanoparticles, functionalized with either negative (-COO-) or positive (-NH3+) surface charges, were functionalized with a DC-SIGN antibody on the particle surface, enabling binding to a dendritic cell surface receptor. A 3D coculture model consisting of epithelial and immune cells (macrophages and dendritic cells) mimicking the human lung epithelial tissue barrier was employed to assess the effects of aerosolized AuNPs. PVA-NH2 AuNPs showed higher uptake compared to that of their -COOH counterparts, with the highest uptake recorded in macrophages, as shown by flow cytometry. None of the AuNPs induced cytotoxicity or necrosis or increased cytokine secretion, whereas only PVA-NH2 AuNPs induced higher apoptosis levels. DC-SIGN AuNPs showed significantly increased uptake by monocyte-derived dendritic cells (MDDCs) with subsequent activation compared to non-antibody-conjugated control AuNPs, independent of surface charge. Our results show that DC-SIGN conjugation to the AuNPs enhanced MDDC targeting and activation in a complex 3D lung cell model. These findings highlight the potential of immunoengineering approaches to the targeting and activation of immune cells in the lung by nanocarriers.
Collapse
Affiliation(s)
| | | | | | - Fabian Blank
- Respiratory Medicine, Inselspital, University of Bern , 3012 Bern, Switzerland
| | | | | | | |
Collapse
|
21
|
Peres C, Matos AI, Conniot J, Sainz V, Zupančič E, Silva JM, Graça L, Sá Gaspar R, Préat V, Florindo HF. Poly(lactic acid)-based particulate systems are promising tools for immune modulation. Acta Biomater 2017; 48:41-57. [PMID: 27826003 DOI: 10.1016/j.actbio.2016.11.012] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Revised: 09/20/2016] [Accepted: 11/03/2016] [Indexed: 01/05/2023]
Abstract
Poly(lactic acid) (PLA) is one of the most successful and versatile polymers explored for controlled delivery of bioactive molecules. Its attractive properties of biodegradability and biocompatibility in vivo have contributed in a meaningful way to the approval of different products by the FDA and EMA for a wide range of biomedical and pharmaceutical applications, in the past two decades. This polymer has been widely used for the preparation of particles as delivery systems of several therapeutic molecules, including vaccines. These PLA vaccine carriers have shown to induce a sustained and targeted release of different bacterial, viral and tumor-associated antigens and adjuvants in vivo, triggering distinct immune responses. The present review intends to highlight and discuss the major advantages of PLA as a promising polymer for the development of potent vaccine delivery systems against pathogens and cancer. It aims to provide a critical discussion based on preclinical data to better understand the major effect of PLA-based carrier properties on their interaction with immune cells and thus their role in the modulation of host immunity. STATEMENT OF SIGNIFICANCE During the last decades, vaccination has had a great impact on global health with the control of many severe diseases. Polymeric nanosystems have emerged as promising strategies to stabilize vaccine antigens, promoting their controlled release to phagocytic cells, thus avoiding the need for multiple administrations. One of the most promising polymers are the aliphatic polyesters, which include the poly(lactic acid). This is a highly versatile biodegradable and biocompatible polymer. Products containing this polymer have already been approved for all food and some biomedical applications. Despite all favorable characteristics presented above, PLA has been less intensively discussed than other polymers, such as its copolymer PLGA, including regarding its application in vaccination and particularly in tumor immunotherapy. The present review discusses the major advantages of poly(lactic acid) for the development of potent vaccine delivery systems, providing a critical view on the main properties that determine their effect on the modulation of immune cells.
Collapse
Affiliation(s)
- Carina Peres
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal; Louvain Drug Research Institute, Advanced Drug Delivery & Biomaterials, Université Catholique de Louvain, 1200 Brussels, Belgium; Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | - Ana I Matos
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - João Conniot
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal; EPSRC Centre for Innovative Manufacturing in Emergent Macromolecular Therapies, UCL School of Pharmacy, London, UK
| | - Vanessa Sainz
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal; EPSRC Centre for Innovative Manufacturing in Emergent Macromolecular Therapies, UCL School of Pharmacy, London, UK
| | - Eva Zupančič
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Joana M Silva
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal; Louvain Drug Research Institute, Advanced Drug Delivery & Biomaterials, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Luís Graça
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | - Rogério Sá Gaspar
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Véronique Préat
- Louvain Drug Research Institute, Advanced Drug Delivery & Biomaterials, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Helena F Florindo
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal.
| |
Collapse
|
22
|
Tuettenberg A, Steinbrink K, Schuppan D. Myeloid cells as orchestrators of the tumor microenvironment: novel targets for nanoparticular cancer therapy. Nanomedicine (Lond) 2016; 11:2735-2751. [DOI: 10.2217/nnm-2016-0208] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Macrophages, myeloid-derived suppressor cells and tolerogenic dendritic cells are central players of a heterogeneous myeloid cell population, with the ability to suppress innate and adaptive immune responses and thus to promote tumor growth. Their influx and local proliferation are mainly induced by the cancers themselves, and their numbers in the tumor microenvironment and the peripheral blood correlate with decreased survival. Therapeutic targeting these innate immune cells, either aiming at their elimination or polarization toward tumor suppressive cells is an attractive novel approach to control tumor progression and block metastasis. We review the current understanding of cancer immunology including immune surveillance and immune editing in the context of these prominent innate suppressor cells, and their targetability by nanoparticular immunotherapy with small molecules or siRNA.
Collapse
Affiliation(s)
- Andrea Tuettenberg
- Department of Dermatology & Research Center for Immunotherapy (FZI) University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Kerstin Steinbrink
- Department of Dermatology & Research Center for Immunotherapy (FZI) University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Detlef Schuppan
- Institute of Translational Immunology & Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg-University, Mainz, Germany
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
23
|
Hutten TJA, Thordardottir S, Fredrix H, Janssen L, Woestenenk R, Tel J, Joosten B, Cambi A, Heemskerk MHM, Franssen GM, Boerman OC, Bakker LBH, Jansen JH, Schaap N, Dolstra H, Hobo W. CLEC12A-Mediated Antigen Uptake and Cross-Presentation by Human Dendritic Cell Subsets Efficiently Boost Tumor-Reactive T Cell Responses. THE JOURNAL OF IMMUNOLOGY 2016; 197:2715-25. [DOI: 10.4049/jimmunol.1600011] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 07/31/2016] [Indexed: 01/19/2023]
|
24
|
Siefert AL, Caplan MJ, Fahmy TM. Artificial bacterial biomimetic nanoparticles synergize pathogen-associated molecular patterns for vaccine efficacy. Biomaterials 2016; 97:85-96. [PMID: 27162077 DOI: 10.1016/j.biomaterials.2016.03.039] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 03/16/2016] [Accepted: 03/28/2016] [Indexed: 11/19/2022]
Abstract
Antigen-presenting cells (APCs) sense microorganisms via pathogen-associated molecular patterns (PAMPs) by both extra- and intracellular Toll-like Receptors (TLRs), initiating immune responses against invading pathogens. Bacterial PAMPs include extracellular lipopolysaccharides and intracellular unmethylated CpG-rich oligodeoxynucleotides (CpG). We hypothesized that a biomimetic approach involving antigen-loaded nanoparticles (NP) displaying Monophosphoryl Lipid A (MPLA) and encapsulating CpG may function as an effective "artificial bacterial" biomimetic vaccine platform. This hypothesis was tested in vitro and in vivo using NP assembled from biodegradable poly(lactic-co-glycolic acid) (PLGA) polymer, surface-modified with MPLA, and loaded with CpG and model antigen Ovalbumin (OVA). First, CpG potency, characterized by cytokine profiles, titers, and antigen-specific T cell responses, was enhanced when CpG was encapsulated in NP compared to equivalent concentrations of surface-presented CpG, highlighting the importance of biomimetic presentation of PAMPs. Second, NP synergized surface-bound MPLA with encapsulated CpG in vitro and in vivo, inducing greater pro-inflammatory, antigen-specific T helper 1 (Th1)-skewed cellular and antibody-mediated responses compared to single PAMPs or soluble PAMP combinations. Importantly, NP co-presentation of CpG and MPLA was critical for CD8(+) T cell responses, as vaccination with a mixture of NP presenting either CpG or MPLA failed to induce cellular immunity. This work demonstrates a rational methodology for combining TLR ligands in a context-dependent manner for synergistic nanoparticulate vaccines.
Collapse
Affiliation(s)
- Alyssa L Siefert
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Michael J Caplan
- Department of Molecular and Cellular Physiology, Yale University, New Haven, CT 06520, USA
| | - Tarek M Fahmy
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA; Department of Chemical and Environmental Engineering, Yale University, New Haven, CT 06520, USA; Department of Immunobiology, Yale University, New Haven, CT 06520, USA.
| |
Collapse
|
25
|
Glass JJ, Kent SJ, De Rose R. Enhancing dendritic cell activation and HIV vaccine effectiveness through nanoparticle vaccination. Expert Rev Vaccines 2016; 15:719-29. [PMID: 26783186 DOI: 10.1586/14760584.2016.1141054] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Novel vaccination approaches are needed to prevent and control human immunodeficiency virus (HIV) infection. A growing body of literature demonstrates the potential of nanotechnology to modulate the human immune system and generate targeted, controlled immune responses. In this Review, we summarize important advances in how 'nanovaccinology' can be used to develop safe and effective vaccines for HIV. We highlight the central role of dendritic cells in the immune response to vaccination and describe how nanotechnology can be used to enhance delivery to and activation of these important antigen-presenting cells. Strategies employed to improve biodistribution are discussed, including improved lymph node delivery and mucosal penetration concepts, before detailing methods to enhance the humoral and/or cellular immune response to vaccines. We conclude with a commentary on the current state of nanovaccinology.
Collapse
Affiliation(s)
- Joshua J Glass
- a ARC Centre of Excellence in Convergent Bio-Nano Science and Technology , The University of Melbourne , Melbourne , Australia.,b Department of Microbiology and Immunology , Peter Doherty Institute for Infection and Immunity, The University of Melbourne , Melbourne , Australia
| | - Stephen J Kent
- a ARC Centre of Excellence in Convergent Bio-Nano Science and Technology , The University of Melbourne , Melbourne , Australia.,b Department of Microbiology and Immunology , Peter Doherty Institute for Infection and Immunity, The University of Melbourne , Melbourne , Australia.,c Melbourne Sexual Health Centre and Department of Infectious Diseases , Alfred Health, Central Clinical School, Monash University , Melbourne , Australia
| | - Robert De Rose
- a ARC Centre of Excellence in Convergent Bio-Nano Science and Technology , The University of Melbourne , Melbourne , Australia.,b Department of Microbiology and Immunology , Peter Doherty Institute for Infection and Immunity, The University of Melbourne , Melbourne , Australia
| |
Collapse
|
26
|
Shevtsov MA, Nikolaev BP, Yakovleva LY, Parr MA, Marchenko YY, Eliseev I, Yudenko A, Dobrodumov AV, Zlobina O, Zhakhov A, Ischenko AM, Pitkin E, Multhoff G. 70-kDa heat shock protein coated magnetic nanocarriers as a nanovaccine for induction of anti-tumor immune response in experimental glioma. J Control Release 2015; 220:329-340. [PMID: 26522072 DOI: 10.1016/j.jconrel.2015.10.051] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 10/15/2015] [Accepted: 10/28/2015] [Indexed: 10/22/2022]
Abstract
Nanovaccines based on superparamagnetic iron oxide nanoparticles (SPIONs) provide a novel approach to induce the humoral and cell-based immune system to fight cancer. Herein, we increased the immunostimulatory capacity of SPIONs by coating them with recombinant heat shock protein 70 (Hsp70) which is known to chaperone antigenic peptides. After binding, Hsp70-SPIONs deliver immunogenic peptides from tumor lysates to dendritiс cells (DCs) and thus stimulate a tumor-specific, CD8+ cytotoxic T cell response. We could show that binding activity of Hsp70-SPIONs to the substrate-binding domain (SBD) is highly dependent on the ATPase activity of its nucleotide-binding domain NBD), as shown by (31)P NMR spectroscopy. Immunization of C6 glioma-bearing rats with DCs pulsed with Hsp70-SPIONs and tumor lysates resulted in a delayed tumor progression (as measured by MRI) and an increased overall survival. In parallel an increased IFNγ secretion were detected in the serum of these animals and immunohistological analysis of subsequent cryosections of the glioma revealed an enhanced infiltration of memory CD45RO+ and cytotoxic CD8+ T cells. Taken together the study demonstrates that magnetic nanocarriers such as SPIONs coated with Hsp70 can be applied as a platform for boosting anti-cancer immune responses.
Collapse
Affiliation(s)
- Maxim A Shevtsov
- Institute of Cytology of the Russian Academy of Sciences (RAS), Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia; I.P. Pavlov State Medical University of St. Petersburg, Lev Tolstoy str. 6/8, 197022 St. Petersburg, Russia; A.L. Polenov Russian Research Scientific Institute of Neurosurgery, Mayakovsky str. 12, 191014 St. Petersburg, Russia; Technische Universität München, Klinikum rechts der Isar, Ismaniger Str. 22, 81675 Munich, Germany.
| | - Boris P Nikolaev
- Research Institute of Highly Pure Biopreparations, Pudozhskaya str. 12, 191014 St. Petersburg, Russia
| | - Liudmila Y Yakovleva
- Research Institute of Highly Pure Biopreparations, Pudozhskaya str. 12, 191014 St. Petersburg, Russia
| | - Marina A Parr
- V.F. Fock Institute of Physics, St. Petersburg State University, Universitetskaya str. 7-9, 199034 St. Petersburg, Russia
| | - Yaroslav Y Marchenko
- Research Institute of Highly Pure Biopreparations, Pudozhskaya str. 12, 191014 St. Petersburg, Russia
| | - Igor Eliseev
- Academic University of the Russian Academy of Sciences (RAS), Hlopina str. 8, 194021 St. Petersburg, Russia
| | - Anna Yudenko
- Academic University of the Russian Academy of Sciences (RAS), Hlopina str. 8, 194021 St. Petersburg, Russia
| | - Anatolii V Dobrodumov
- Institute of Macromolecular Compounds of the Russian Academy of Sciences (RAS), Bolshoi pr. 31, 199004 St. Petersburg, Russia
| | - Olga Zlobina
- Research Institute of Highly Pure Biopreparations, Pudozhskaya str. 12, 191014 St. Petersburg, Russia
| | - Alexander Zhakhov
- Research Institute of Highly Pure Biopreparations, Pudozhskaya str. 12, 191014 St. Petersburg, Russia
| | - Alexander M Ischenko
- Research Institute of Highly Pure Biopreparations, Pudozhskaya str. 12, 191014 St. Petersburg, Russia
| | - Emil Pitkin
- The Wharton School, University of Pennsylvania, 3730 Walnut St., Philadelphia, PA 19104, USA
| | - Gabriele Multhoff
- Technische Universität München, Klinikum rechts der Isar, Ismaniger Str. 22, 81675 Munich, Germany
| |
Collapse
|
27
|
Pros and Cons of Antigen-Presenting Cell Targeted Tumor Vaccines. J Immunol Res 2015; 2015:785634. [PMID: 26583156 PMCID: PMC4637118 DOI: 10.1155/2015/785634] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 08/26/2015] [Accepted: 09/03/2015] [Indexed: 01/08/2023] Open
Abstract
In therapeutic antitumor vaccination, dendritic cells play the leading role since they decide if, how, when, and where a potent antitumor immune response will take place. Since the disentanglement of the complexity and merit of different antigen-presenting cell subtypes, antitumor immunotherapeutic research started to investigate the potential benefit of targeting these subtypes in situ. This review will discuss which antigen-presenting cell subtypes are at play and how they have been targeted and finally question the true meaning of targeting antitumor-based vaccines.
Collapse
|
28
|
Mocan T, Matea C, Tabaran F, Iancu C, Orasan R, Mocan L. In Vitro Administration of Gold Nanoparticles Functionalized with MUC-1 Protein Fragment Generates Anticancer Vaccine Response via Macrophage Activation and Polarization Mechanism. J Cancer 2015; 6:583-92. [PMID: 26000051 PMCID: PMC4439945 DOI: 10.7150/jca.11567] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 04/13/2015] [Indexed: 12/17/2022] Open
Abstract
Therapeutic cancer vaccines (or active immunotherapy) aim to guide the patient's personal immune system to eradicate cancer cells. An exciting approach to cancer vaccines has been offered by nanoscale drug delivery systems containing tumor associated antigens (TAAs). Their capacity to stimulate the immune system has been suggested during late years. However, the role of the macrophages as key-elements in antigen-presentation process following TAAs-containing nanosystems is not completely understood. We aimed to evaluate the effect of gold nanoparticles functionalized with mucin-1 peptide (MUC-1) on murine peritoneal macrophages. Gold nanoparticles, obtained using a modified Turkevich method, were functionalized with MUC-1 protein using Clealand's reagent. The obtained GNP-MUC-1 solution was used to treat at various concentrations monolayers of peritoneum-derived macrophages that were further analyzed using confocal and hyperspectral microscopy, ELISA assays and spectroscopic techniques. The GNP-MUC-1 nano-construct had proven to function as a powerful macrophage activator with consequent release of cytokines such as: TNF-ɑ, IL-6, IL-10 and IL-12 on peritoneal macrophages we have isolated from mice. Our results demonstrate optimization of antigen-presenting process and predominant M1 polarization following exposure GNP-MUC-1. To our best knowledge this is the first study to evaluate the anticancer effects of a newly designed nano-biocompound on the complex antigen- processing apparatus of peritoneal macrophages.
Collapse
Affiliation(s)
- Teodora Mocan
- 2. Department of Nanomedicine, Institute of Gastroenterology and Hepatology, 19-21 Croitorilor St., 400162 Cluj-Napoca, Romania
- 3. Department of Physiology, “Iuliu Hatieganu” University of Medicine and Pharmacy, no. 1 Clinicilor Street., 400006 Cluj-Napoca, Romania
| | - Cristian Matea
- 1. 3rd Surgery Clinic, “Iuliu Hatieganu” University of Medicine and Pharmacy, 19-21 Croitorilor St., 400162 Cluj-Napoca, Romania
- 2. Department of Nanomedicine, Institute of Gastroenterology and Hepatology, 19-21 Croitorilor St., 400162 Cluj-Napoca, Romania
| | - Flaviu Tabaran
- 2. Department of Nanomedicine, Institute of Gastroenterology and Hepatology, 19-21 Croitorilor St., 400162 Cluj-Napoca, Romania
- 4. Department of Pathology, Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine, Cluj-Napoca, Romania; Calea Mănăștur 3-5, Cluj-Napoca 400372
| | - Cornel Iancu
- 1. 3rd Surgery Clinic, “Iuliu Hatieganu” University of Medicine and Pharmacy, 19-21 Croitorilor St., 400162 Cluj-Napoca, Romania
- 2. Department of Nanomedicine, Institute of Gastroenterology and Hepatology, 19-21 Croitorilor St., 400162 Cluj-Napoca, Romania
| | - Remus Orasan
- 3. Department of Physiology, “Iuliu Hatieganu” University of Medicine and Pharmacy, no. 1 Clinicilor Street., 400006 Cluj-Napoca, Romania
| | - Lucian Mocan
- 1. 3rd Surgery Clinic, “Iuliu Hatieganu” University of Medicine and Pharmacy, 19-21 Croitorilor St., 400162 Cluj-Napoca, Romania
- 2. Department of Nanomedicine, Institute of Gastroenterology and Hepatology, 19-21 Croitorilor St., 400162 Cluj-Napoca, Romania
| |
Collapse
|
29
|
Mody N, Dubey S, Sharma R, Agrawal U, Vyas SP. Dendritic cell-based vaccine research against cancer. Expert Rev Clin Immunol 2014; 11:213-32. [DOI: 10.1586/1744666x.2015.987663] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
30
|
Dhodapkar MV, Dhodapkar KM. Recent advances and new opportunities for targeting human dendritic cells in situ. Oncoimmunology 2014; 3:e954832. [PMID: 25610750 DOI: 10.4161/21624011.2014.954832] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 07/07/2014] [Indexed: 11/19/2022] Open
Abstract
Targeting antigens directly to dendritic cells (DCs) in situ has emerged as a promising strategy to potentiate immunity. A recent clinical trial of antibody-mediated targeting of tumor antigen NY-ESO1 to the DC receptor DEC-205 demonstrated an induction of strong cellular and humoral responses. Recent studies with DC-targeting via nanoparticles suggest that combinatorial targeting of multiple human DC subsets may further improve the efficacy of DC targeting.
Collapse
|
31
|
Sehgal K, Dhodapkar KM, Dhodapkar MV. Targeting human dendritic cells in situ to improve vaccines. Immunol Lett 2014; 162:59-67. [PMID: 25072116 DOI: 10.1016/j.imlet.2014.07.004] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2014] [Revised: 07/17/2014] [Accepted: 07/17/2014] [Indexed: 12/12/2022]
Abstract
Dendritic cells (DCs) provide a critical link between innate and adaptive immunity. The potent antigen presenting properties of DCs makes them a valuable target for the delivery of immunogenic cargo. Recent clinical studies describing in situ DC targeting with antibody-mediated targeting of DC receptor through DEC-205 provide new opportunities for the clinical application of DC-targeted vaccines. Further advances with nanoparticle vectors which can encapsulate antigens and adjuvants within the same compartment and be targeted against diverse DC subsets also represent an attractive strategy for targeting DCs. This review provides a brief summary of the rationale behind targeting dendritic cells in situ, the existing pre-clinical and clinical data on these vaccines and challenges faced by the next generation DC-targeted vaccines.
Collapse
Affiliation(s)
- Kartik Sehgal
- Department of Medicine, Yale University, New Haven, CT, United States
| | | | | |
Collapse
|