1
|
Jung C, Fichter M, Oberländer J, Schunke J, Bolduan V, Schneider P, Kang J, Koynov K, Mailänder V, Landfester K. Nanobodies Outperform Antibodies - Rapid Functionalization with Equal In Vivo Targeting Properties. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2412563. [PMID: 39468885 DOI: 10.1002/adma.202412563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/01/2024] [Indexed: 10/30/2024]
Abstract
Highly specific targeting of dendritic cells in vivo is crucial for the development of effective tumor nanovaccines. This group recently presented an antibody-functionalized nanocarrier system able to maintain its targeting properties when transferred from in vitro to in vivo studies. However, producing this system requires long synthesis times and involves high expenses due to the involved site-specific enzymatic multi-step modification procedure of the antibody. Consequently, improving the previously proposed system is necessary in order to accelerate the development. Here, a novel system utilizing nanobodies for the targeting of dendritic cells is presented. A C-terminal cysteine tag facilitates an easy attachment of the nanobody to the nanocarrier via a thiol-maleimide conjugation technique. This reduces the functionalization time from several days to mere hours. Using in vitro and in vivo assays, it is shown that the optimized system possesses equal targeting properties as the antibody-based system. As a result, nanobodies and the coupling chemistry are found to be a superior strategy for the in vivo targeting of dendritic cells when compared to antibodies, due to their rapid attachment to nanocarriers and equal targeting specificity. This would replace antibodies as the current "gold standard" of targeting moieties.
Collapse
Affiliation(s)
- Carina Jung
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Michael Fichter
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Jennifer Oberländer
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Jenny Schunke
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Vanessa Bolduan
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Paul Schneider
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Jinhong Kang
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Kaloian Koynov
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
| | - Volker Mailänder
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Katharina Landfester
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
| |
Collapse
|
2
|
Sorrentino C, Ciummo SL, Fieni C, Di Carlo E. Nanomedicine for cancer patient-centered care. MedComm (Beijing) 2024; 5:e767. [PMID: 39434967 PMCID: PMC11491554 DOI: 10.1002/mco2.767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/13/2024] [Accepted: 09/13/2024] [Indexed: 10/23/2024] Open
Abstract
Cancer is a leading cause of morbidity and mortality worldwide, and an increase in incidence is estimated in the next future, due to population aging, which requires the development of highly tolerable and low-toxicity cancer treatment strategies. The use of nanotechnology to tailor treatments according to the genetic and immunophenotypic characteristics of a patient's tumor, and to allow its targeted release, can meet this need, improving the efficacy of treatment and minimizing side effects. Nanomedicine-based approach for the diagnosis and treatment of cancer is a rapidly evolving field. Several nanoformulations are currently in clinical trials, and some have been approved and marketed. However, their large-scale production and use are still hindered by an in-depth debate involving ethics, intellectual property, safety and health concerns, technical issues, and costs. Here, we survey the key approaches, with specific reference to organ-on chip technology, and cutting-edge tools, such as CRISPR/Cas9 genome editing, through which nanosystems can meet the needs for personalized diagnostics and therapy in cancer patients. An update is provided on the nanopharmaceuticals approved and marketed for cancer therapy and those currently undergoing clinical trials. Finally, we discuss the emerging avenues in the field and the challenges to be overcome for the transfer of nano-based precision oncology into clinical daily life.
Collapse
Affiliation(s)
- Carlo Sorrentino
- Department of Medicine and Sciences of Aging“G. d'Annunzio” University” of Chieti‐PescaraChietiItaly
- Anatomic Pathology and Immuno‐Oncology Unit, Center for Advanced Studies and Technology (CAST)“G. d'Annunzio” University of Chieti‐PescaraChietiItaly
| | - Stefania Livia Ciummo
- Department of Medicine and Sciences of Aging“G. d'Annunzio” University” of Chieti‐PescaraChietiItaly
- Anatomic Pathology and Immuno‐Oncology Unit, Center for Advanced Studies and Technology (CAST)“G. d'Annunzio” University of Chieti‐PescaraChietiItaly
| | - Cristiano Fieni
- Department of Medicine and Sciences of Aging“G. d'Annunzio” University” of Chieti‐PescaraChietiItaly
- Anatomic Pathology and Immuno‐Oncology Unit, Center for Advanced Studies and Technology (CAST)“G. d'Annunzio” University of Chieti‐PescaraChietiItaly
| | - Emma Di Carlo
- Department of Medicine and Sciences of Aging“G. d'Annunzio” University” of Chieti‐PescaraChietiItaly
- Anatomic Pathology and Immuno‐Oncology Unit, Center for Advanced Studies and Technology (CAST)“G. d'Annunzio” University of Chieti‐PescaraChietiItaly
| |
Collapse
|
3
|
Hale G. Living in LALA land? Forty years of attenuating Fc effector functions. Immunol Rev 2024; 328:422-437. [PMID: 39158044 PMCID: PMC11659930 DOI: 10.1111/imr.13379] [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] [Indexed: 08/20/2024]
Abstract
The Fc region of antibodies is vital for most of their physiological functions, many of which are engaged through binding to a range of Fc receptors. However, these same interactions are not always helpful or wanted when therapeutic antibodies are directed against self-antigens, and can sometimes cause catastrophic adverse reactions. Over the past 40 years, there have been intensive efforts to "silence" unwanted binding to Fc-gamma receptors, resulting in at least 45 different variants which have entered clinical trials. One of the best known is "LALA" (L234A/L235A). However, neither this, nor most of the other variants in clinical use are completely silenced, and in addition, the biophysical properties of many of them are compromised. I review the development of different variants to see what we can learn from their biological properties and use in the clinic. With the rise of powerful new uses of antibody therapy such as bispecific T-cell engagers, antibody-drug conjugates, and checkpoint inhibitors, it is increasingly important to optimize the Fc region as well as the antibody binding site in order to achieve the best combination of safety and efficacy.
Collapse
|
4
|
Sui C, Wu H, Li X, Wang Y, Wei J, Yu J, Wu X. Cancer immunotherapy and its facilitation by nanomedicine. Biomark Res 2024; 12:77. [PMID: 39097732 PMCID: PMC11297660 DOI: 10.1186/s40364-024-00625-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 07/22/2024] [Indexed: 08/05/2024] Open
Abstract
Cancer immunotherapy has sparked a wave of cancer research, driven by recent successful proof-of-concept clinical trials. However, barriers are emerging during its rapid development, including broad adverse effects, a lack of reliable biomarkers, tumor relapses, and drug resistance. Integration of nanomedicine may ameliorate current cancer immunotherapy. Ultra-large surface-to-volume ratio, extremely small size, and easy modification surface of nanoparticles enable them to selectively detect cells and kill cancer cells in vivo. Exciting synergistic applications of the two approaches have emerged in treating various cancers at the intersection of cancer immunotherapy and cancer nanomedicine, indicating the potential that the combination of these two therapeutic modalities can lead to new paradigms in the treatment of cancer. This review discusses the status of current immunotherapy and explores the possible opportunities that the nanomedicine platform can make cancer immunotherapy more powerful and precise by synergizing the two approaches.
Collapse
Affiliation(s)
- Chao Sui
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, 1500 East Duarte, Los Angeles, CA, 91010, USA
| | - Heqing Wu
- The First Affiliated Hospital of Soochow University, Suzhou, China
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Xinxin Li
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an Shaanxi, 710072, China
| | - Yuhang Wang
- The First Affiliated Hospital of Soochow University, Suzhou, China
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Jiaqi Wei
- The First Affiliated Hospital of Soochow University, Suzhou, China
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Jianhua Yu
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, 1500 East Duarte, Los Angeles, CA, 91010, USA.
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, 91010, USA.
| | - Xiaojin Wu
- The First Affiliated Hospital of Soochow University, Suzhou, China.
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, China.
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.
| |
Collapse
|
5
|
Li F, Ouyang J, Chen Z, Zhou Z, Milon Essola J, Ali B, Wu X, Zhu M, Guo W, Liang XJ. Nanomedicine for T-Cell Mediated Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2301770. [PMID: 36964936 DOI: 10.1002/adma.202301770] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/14/2023] [Indexed: 06/18/2023]
Abstract
T-cell immunotherapy offers outstanding advantages in the treatment of various diseases, and with the selection of appropriate targets, efficient disease treatment can be achieved. T-cell immunotherapy has made great progress, but clinical results show that only a small proportion of patients can benefit from T-cell immunotherapy. The extensive mechanistic work outlines a blueprint for using T cells as a new option for immunotherapy, but also presents new challenges, including the balance between different fractions of T cells, the inherent T-cell suppression patterns in the disease microenvironment, the acquired loss of targets, and the decline of T-cell viability. The diversity, flexibility, and intelligence of nanomedicines give them great potential for enhancing T-cell immunotherapy. Here, how T-cell immunotherapy strategies can be adapted with different nanomaterials to enhance therapeutic efficacy is discussed. For two different pathological states, immunosuppression and immune activation, recent advances in nanomedicines for T-cell immunotherapy in diseases such as cancers, rheumatoid arthritis, systemic lupus erythematosus, ulcerative colitis, and diabetes are summarized. With a focus on T-cell immunotherapy, this review highlights the outstanding advantages of nanomedicines in disease treatment, and helps advance one's understanding of the use of nanotechnology to enhance T-cell immunotherapy.
Collapse
Affiliation(s)
- Fangzhou Li
- Department of Minimally Invasive Interventional Radiology, the State Key Laboratory of Respiratory Disease, School of Biomedical Engineering & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, P. R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
| | - Jiang Ouyang
- Department of Minimally Invasive Interventional Radiology, the State Key Laboratory of Respiratory Disease, School of Biomedical Engineering & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, P. R. China
| | - Zuqin Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
| | - Ziran Zhou
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Julien Milon Essola
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Barkat Ali
- Department of Minimally Invasive Interventional Radiology, the State Key Laboratory of Respiratory Disease, School of Biomedical Engineering & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, P. R. China
- Food Sciences Research Institute, Pakistan Agricultural Research Council, 44000, Islamabad, Pakistan
| | - Xinyue Wu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Mengliang Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
| | - Weisheng Guo
- Department of Minimally Invasive Interventional Radiology, the State Key Laboratory of Respiratory Disease, School of Biomedical Engineering & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, P. R. China
| | - Xing-Jie Liang
- Department of Minimally Invasive Interventional Radiology, the State Key Laboratory of Respiratory Disease, School of Biomedical Engineering & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, P. R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, P. R. China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
6
|
Jiang X, Wu L, Zhang M, Zhang T, Chen C, Wu Y, Yin C, Gao J. Biomembrane nanostructures: Multifunctional platform to enhance tumor chemoimmunotherapy via effective drug delivery. J Control Release 2023; 361:510-533. [PMID: 37567505 DOI: 10.1016/j.jconrel.2023.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/02/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023]
Abstract
Chemotherapeutic drugs have been found to activate the immune response against tumors by inducing immunogenic cell death, in addition to their direct cytotoxic effects toward tumors, therefore broadening the application of chemotherapy in tumor immunotherapy. The combination of other therapeutic strategies, such as phototherapy or radiotherapy, could further strengthen the therapeutic effects of immunotherapy. Nanostructures can facilitate multimodal tumor therapy by integrating various active agents and combining multiple types of therapeutics in a single nanostructure. Biomembrane nanostructures (e.g., exosomes and cell membrane-derived nanostructures), characterized by superior biocompatibility, intrinsic targeting ability, intelligent responsiveness and immune-modulating properties, could realize superior chemoimmunotherapy and represent next-generation nanostructures for tumor immunotherapy. This review summarizes recent advances in biomembrane nanostructures in tumor chemoimmunotherapy and highlights different types of engineering approaches and therapeutic mechanisms. A series of engineering strategies for combining different biomembrane nanostructures, including liposomes, exosomes, cell membranes and bacterial membranes, are summarized. The combination strategy can greatly enhance the targeting, intelligence and functionality of biomembrane nanostructures for chemoimmunotherapy, thereby serving as a stronger tumor therapeutic method. The challenges associated with the clinical translation of biomembrane nanostructures for chemoimmunotherapy and their future perspectives are also discussed.
Collapse
Affiliation(s)
- Xianghe Jiang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China; College of Life Science, Mudanjiang Medical University, Mudanjiang 157011, China
| | - Lili Wu
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Mengya Zhang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Tinglin Zhang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Cuimin Chen
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Yan Wu
- College of Life Science, Mudanjiang Medical University, Mudanjiang 157011, China.
| | - Chuan Yin
- Department of Gastroenterology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai 200003, China.
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
7
|
Roy M, Roy A, Rustagi S, Pandey N. An Overview of Nanomaterial Applications in Pharmacology. BIOMED RESEARCH INTERNATIONAL 2023; 2023:4838043. [PMID: 37388336 PMCID: PMC10307208 DOI: 10.1155/2023/4838043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 05/06/2023] [Accepted: 06/13/2023] [Indexed: 07/01/2023]
Abstract
Nanotechnology has become one of the most extensive fields of research. Nanoparticles (NPs) form the base for nanotechnology. Recently, nanomaterials (NMs) are widely used due to flexible chemical, biological, and physical characteristics with improved efficacy in comparison to bulk counterparts. The significance of each class of NMs is enhanced by identifying their properties. Day by day, there is an emergence of various applications of NMs, but the toxic effects associated with them cannot be avoided. NMs demonstrate therapeutic abilities by enhancing the drug delivery system, diagnosis, and therapeutic effects of numerous agents, but determining the benefits of NMs over other clinical applications (disease-specific) or substances is an ongoing investigation. This review is aimed at defining NMs and NPs and their types, synthesis, and pharmaceutical, biomedical, and clinical applications.
Collapse
Affiliation(s)
- Madhura Roy
- Centre for Translational and Clinical Research, School of Chemical and Life Sciences, Jamia Hamdard, India
| | - Arpita Roy
- Department of Biotechnology, Sharda School of Engineering & Technology, Sharda University, Greater Noida, India
| | - Sarvesh Rustagi
- School of Applied and Life sciences, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Neha Pandey
- Department of Biotechnology, Graphic Era Deemed to Be University, Dehradun, Uttarakhand, India
| |
Collapse
|
8
|
Dong H, Li Q, Zhang Y, Ding M, Teng Z, Mou Y. Biomaterials Facilitating Dendritic Cell-Mediated Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301339. [PMID: 37088780 PMCID: PMC10288267 DOI: 10.1002/advs.202301339] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/22/2023] [Indexed: 05/03/2023]
Abstract
Dendritic cell (DC)-based cancer immunotherapy has exhibited remarkable clinical prospects because DCs play a central role in initiating and regulating adaptive immune responses. However, the application of traditional DC-mediated immunotherapy is limited due to insufficient antigen delivery, inadequate antigen presentation, and high levels of immunosuppression. To address these challenges, engineered biomaterials have been exploited to enhance DC-mediated immunotherapeutic effects. In this review, vital principal components that can enhance DC-mediated immunotherapeutic effects are first introduced. The parameters considered in the rational design of biomaterials, including targeting modifications, size, shape, surface, and mechanical properties, which can affect biomaterial optimization of DC functions, are further summarized. Moreover, recent applications of various engineered biomaterials in the field of DC-mediated immunotherapy are reviewed, including those serve as immune component delivery platforms, remodel the tumor microenvironment, and synergistically enhance the effects of other antitumor therapies. Overall, the present review comprehensively and systematically summarizes biomaterials related to the promotion of DC functions; and specifically focuses on the recent advances in biomaterial designs for DC activation to eradicate tumors. The challenges and opportunities of treatment strategies designed to amplify DCs via the application of biomaterials are discussed with the aim of inspiring the clinical translation of future DC-mediated cancer immunotherapies.
Collapse
Affiliation(s)
- Heng Dong
- Nanjing Stomatological HospitalAffiliated Hospital of Medical School, Nanjing University30 Zhongyang RoadNanjingJiangsu210008P. R. China
| | - Qiang Li
- Nanjing Stomatological HospitalAffiliated Hospital of Medical School, Nanjing University30 Zhongyang RoadNanjingJiangsu210008P. R. China
| | - Yu Zhang
- Nanjing Stomatological HospitalAffiliated Hospital of Medical School, Nanjing University30 Zhongyang RoadNanjingJiangsu210008P. R. China
| | - Meng Ding
- Nanjing Stomatological HospitalAffiliated Hospital of Medical School, Nanjing University30 Zhongyang RoadNanjingJiangsu210008P. R. China
| | - Zhaogang Teng
- Key Laboratory for Organic Electronics and Information DisplaysJiangsu Key Laboratory for BiosensorsInstitute of Advanced MaterialsJiangsu National Synergetic Innovation Centre for Advanced MaterialsNanjing University of Posts and Telecommunications9 Wenyuan RoadNanjingJiangsu210023P. R. China
| | - Yongbin Mou
- Nanjing Stomatological HospitalAffiliated Hospital of Medical School, Nanjing University30 Zhongyang RoadNanjingJiangsu210008P. R. China
| |
Collapse
|
9
|
Meng L, Teng Z, Yang S, Wang N, Guan Y, Chen X, Liu Y. Biomimetic nanoparticles for DC vaccination: a versatile approach to boost cancer immunotherapy. NANOSCALE 2023; 15:6432-6455. [PMID: 36916703 DOI: 10.1039/d2nr07071e] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Cancer immunotherapy, which harnesses the immune system to fight cancer, has begun to make a breakthrough in clinical applications. Dendritic cells (DCs) are the bridge linking innate and adaptive immunity and the trigger of tumor immune response. Considering the cumbersome process and poor efficacy of classic DC vaccines, there has been interest in transferring the field of in vitro-generated DC vaccines to nanovaccines. Conventional nanoparticles have insufficient targeting ability and are easily cleared by the reticuloendothelial system. Biological components have evolved very specific functions, which are difficult to fully reproduce with synthetic materials, making people interested in using the further understanding of biological systems to prepare nanoparticles with new and enhanced functions. Biomimetic nanoparticles are semi-biological or nature-derived delivery systems comprising one or more natural materials, which have a long circulation time in vivo and excellent performance of targeting DCs, and can mimic the antigen-presenting behavior of DCs. In this review, we introduce the classification, design, preparation, and challenges of different biomimetic nanoparticles, and discuss their application in activating DCs in vivo and stimulating T cell antitumor immunity. Incorporating biomimetic nanoparticles into cancer immunotherapy has shown outstanding advantages in precisely coaxing the immune system against cancer.
Collapse
Affiliation(s)
- Lingyang Meng
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, P.R. China.
| | - Zhuang Teng
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, P.R. China.
| | - Shuang Yang
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, P.R. China.
| | - Na Wang
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, P.R. China.
| | - YingHua Guan
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, P.R. China.
| | - Xiguang Chen
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, P.R. China.
- Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266000, P.R. China
| | - Ya Liu
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, P.R. China.
| |
Collapse
|
10
|
Hu Y, Zhang W, Chu X, Wang A, He Z, Si CL, Hu W. Dendritic cell-targeting polymer nanoparticle-based immunotherapy for cancer: A review. Int J Pharm 2023; 635:122703. [PMID: 36758880 DOI: 10.1016/j.ijpharm.2023.122703] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 02/01/2023] [Accepted: 02/04/2023] [Indexed: 02/10/2023]
Abstract
Cancer immunity is dependent on dynamic interactions between T cells and dendritic cells (DCs). Polymer-based nanoparticles target DC receptors to improve anticancer immune responses. In this paper, DC surface receptors and their specific coupling natural ligands and antibodies are reviewed and compared. Moreover, reaction mechanisms are described, and the synergistic effects of immune adjuvants are demonstrated. Also, extracellular-targeting antigen-delivery strategies and intracellular stimulus responses are reviewed to promote the rational design of polymer delivery systems.
Collapse
Affiliation(s)
- Yeye Hu
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China; Tianjin Key Laboratory of Pulp & Paper, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Wei Zhang
- School of Life Sciences, Huaiyin Normal University, Huaian 223300, China
| | - Xiaozhong Chu
- School of Chemistry & Chemical Engineering, Huaiyin Normal University, Huaian 223300, China
| | - Aoran Wang
- School of Chemistry & Chemical Engineering, Huaiyin Normal University, Huaian 223300, China
| | - Ziliang He
- School of Life Sciences, Huaiyin Normal University, Huaian 223300, China
| | - Chuan-Ling Si
- Tianjin Key Laboratory of Pulp & Paper, Tianjin University of Science & Technology, Tianjin 300457, China.
| | - Weicheng Hu
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China; Affiliated Hospital of Yangzhou University, Yangzhou 225009, China; Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, School of Medicine, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
11
|
Ruan S, Huang Y, He M, Gao H. Advanced Biomaterials for Cell-Specific Modulation and Restore of Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200027. [PMID: 35343112 PMCID: PMC9165523 DOI: 10.1002/advs.202200027] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/18/2022] [Indexed: 05/09/2023]
Abstract
The past decade has witnessed the explosive development of cancer immunotherapies. Nevertheless, low immunogenicity, limited specificity, poor delivery efficiency, and off-target side effects remain to be the major limitations for broad implementation of cancer immunotherapies to patient bedside. Encouragingly, advanced biomaterials offering cell-specific modulation of immunological cues bring new solutions for improving the therapeutic efficacy while relieving side effect risks. In this review, focus is given on how functional biomaterials can enable cell-specific modulation of cancer immunotherapy within the cancer-immune cycle, with particular emphasis on antigen-presenting cells (APCs), T cells, and tumor microenvironment (TME)-resident cells. By reviewing the current progress in biomaterial-based cancer immunotherapy, here the aim is to provide a better understanding of biomaterials' role in targeting modulation of antitumor immunity step-by-step and guidelines for rationally developing targeting biomaterials for more personalized cancer immunotherapy. Moreover, the current challenge and future perspective regarding the potential application and clinical translation will also be discussed.
Collapse
Affiliation(s)
- Shaobo Ruan
- Advanced Research Institute of Multidisciplinary ScienceBeijing Institute of TechnologyBeijing100081China
| | - Yuanyu Huang
- Advanced Research Institute of Multidisciplinary ScienceBeijing Institute of TechnologyBeijing100081China
| | - Mei He
- College of PharmacyUniversity of FloridaGainesvilleFL32610USA
| | - Huile Gao
- West China School of PharmacySichuan UniversityChengdu610041China
| |
Collapse
|
12
|
Wilkinson I, Anderson S, Fry J, Julien LA, Neville D, Qureshi O, Watts G, Hale G. Fc-engineered antibodies with immune effector functions completely abolished. PLoS One 2021; 16:e0260954. [PMID: 34932587 PMCID: PMC8691596 DOI: 10.1371/journal.pone.0260954] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 11/20/2021] [Indexed: 01/12/2023] Open
Abstract
Elimination of the binding of immunoglobulin Fc to Fc gamma receptors (FcγR) is highly desirable for the avoidance of unwanted inflammatory responses to therapeutic antibodies and fusion proteins. Many different approaches have been described in the literature but none of them completely eliminates binding to all of the Fcγ receptors. Here we describe a set of novel variants having specific amino acid substitutions in the Fc region at L234 and L235 combined with the substitution G236R. They show no detectable binding to Fcγ receptors or to C1q, are inactive in functional cell-based assays and do not elicit inflammatory cytokine responses. Meanwhile, binding to FcRn, manufacturability, stability and potential for immunogenicity are unaffected. These variants have the potential to improve the safety and efficacy of therapeutic antibodies and Fc fusion proteins.
Collapse
Affiliation(s)
- Ian Wilkinson
- Absolute Antibody Ltd, Wilton, United Kingdom
- mAbsolve Limited, Oxford, United Kingdom
| | | | - Jeremy Fry
- ProImmune Limited, Oxford, United Kingdom
| | | | - David Neville
- Reading Scientific Services Limited, Reading, United Kingdom
| | | | - Gary Watts
- Abzena Limited, Babraham, United Kingdom
| | - Geoff Hale
- mAbsolve Limited, Oxford, United Kingdom
- * E-mail:
| |
Collapse
|
13
|
Biomembrane-based nanostructures for cancer targeting and therapy: From synthetic liposomes to natural biomembranes and membrane-vesicles. Adv Drug Deliv Rev 2021; 178:113974. [PMID: 34530015 DOI: 10.1016/j.addr.2021.113974] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/29/2021] [Accepted: 09/08/2021] [Indexed: 12/14/2022]
Abstract
The translational success of liposomes in chemotherapeutics has already demonstrated the great potential of biomembrane-based nanostructure in effective drug delivery. Meanwhile, increasing efforts are being dedicated to the application of naturally derived lipid membranes, including cellular membranes and extracellular vesicles in anti-cancer therapies. While synthetic liposomes support superior multifunctional flexibility, natural biomembrane materials possess interesting biomimetic properties and can also be further engineered for intelligent design. Despite being remarkably different from each other in production and composition, the phospholipid bilayer structure in common allows liposomes, cell membrane-derived nanomaterials, and extracellular vesicles to be modified, functionalized, and exploited in many similar manners against challenges posed by tumor-targeted drug delivery. This review will summarize the recent advancements in engineering the membrane-derived nanostructures with "intelligent" modules to respond, regulate, and target tumor cells and the microenvironment to fight against malignancy. We will also discuss perspectives of combining engineered functionalities with naturally occurring activity for enhanced cancer therapy.
Collapse
|
14
|
Zhou L, Liu H, Liu K, Wei S. Gold Compounds and the Anticancer Immune Response. Front Pharmacol 2021; 12:739481. [PMID: 34588987 PMCID: PMC8473785 DOI: 10.3389/fphar.2021.739481] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 08/31/2021] [Indexed: 12/20/2022] Open
Abstract
Gold compounds are not only well-explored for cytotoxic effects on tumors, but are also known to interact with the cancer immune system. The immune system deploys innate and adaptive mechanisms to protect against pathogens and prevent malignant transformation. The combined action of gold compounds with the activated immune system has shown promising results in cancer therapy through in vivo and in vitro experiments. Gold compounds are known to induce innate immune responses; however, these responses may contribute to adaptive immune responses. Gold compounds play the role of a major hapten that acts synergistically in innate immunity. Gold compounds support cancer cell antigenicity and promote anti-tumor immune response by inducing the release of CRT, ATP, HMGB1, HSP, and NKG2D to enhance immunogenicity. Gold compounds affect various immune cells (including suppressor regulatory T cells), inhibit myeloid derived suppressor cells, and enhance the function and number of dendritic cells. Gold nanoparticles (AuNPs) have potential for improving the effect of immunotherapy and reducing the toxicity and side effects of the treatment process. Thus, AuNPs provide an ideal opportunity for exploring the combination of anticancer gold compounds and immunotherapeutic interventions.
Collapse
Affiliation(s)
- Ling Zhou
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan, China
| | - Huiguo Liu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan, China
| | - Kui Liu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan, China
| | - Shuang Wei
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
15
|
Chauhan A, Khan T, Omri A. Design and Encapsulation of Immunomodulators onto Gold Nanoparticles in Cancer Immunotherapy. Int J Mol Sci 2021; 22:8037. [PMID: 34360803 PMCID: PMC8347387 DOI: 10.3390/ijms22158037] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 07/20/2021] [Accepted: 07/23/2021] [Indexed: 02/06/2023] Open
Abstract
The aim of cancer immunotherapy is to reactivate autoimmune responses to combat cancer cells. To stimulate the immune system, immunomodulators, such as adjuvants, cytokines, vaccines, and checkpoint inhibitors, are extensively designed and studied. Immunomodulators have several drawbacks, such as drug instability, limited half-life, rapid drug clearance, and uncontrolled immune responses when used directly in cancer immunotherapy. Several strategies have been used to overcome these limitations. A simple and effective approach is the loading of immunomodulators onto gold-based nanoparticles (GNPs). As gold is highly biocompatible, GNPs can be administered intravenously, which aids in increasing cancer cell permeability and retention time. Various gold nanoplatforms, including nanospheres, nanoshells, nanorods, nanocages, and nanostars have been effectively used in cancer immunotherapy. Gold nanostars (GNS) are one of the most promising GNP platforms because of their unusual star-shaped geometry, which significantly increases light absorption and provides high photon-to-heat conversion efficiency due to the plasmonic effect. As a result, GNPs are a useful vehicle for delivering antigens and adjuvants that support the immune system in killing tumor cells by facilitating or activating cytotoxic T lymphocytes. This review represents recent progress in encapsulating immunomodulators into GNPs for utility in a cancer immunotherapeutic regimen.
Collapse
Affiliation(s)
- Akshita Chauhan
- Department of Quality Assurance, SVKM’s Dr. Bhanuben Nanavati College of Pharmacy, Mumbai 400056, Maharashtra, India;
| | - Tabassum Khan
- Department of Pharmaceutical Chemistry & Quality Assurance, SVKM’s Dr. Bhanuben Nanavati College of Pharmacy, Mumbai 400056, Maharashtra, India;
| | - Abdelwahab Omri
- The Novel Drug & Vaccine Delivery Systems Facility, Department of Chemistry and Biochemistry, Laurentian University, Sudbury, ON P3E 2C6, Canada
| |
Collapse
|
16
|
Nijen Twilhaar MK, Czentner L, van Nostrum CF, Storm G, den Haan JMM. Mimicking Pathogens to Augment the Potency of Liposomal Cancer Vaccines. Pharmaceutics 2021; 13:954. [PMID: 34202919 PMCID: PMC8308965 DOI: 10.3390/pharmaceutics13070954] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 01/02/2023] Open
Abstract
Liposomes have emerged as interesting vehicles in cancer vaccination strategies as their composition enables the inclusion of both hydrophilic and hydrophobic antigens and adjuvants. In addition, liposomes can be decorated with targeting moieties to further resemble pathogenic particles that allow for better engagement with the immune system. However, so far liposomal cancer vaccines have not yet reached their full potential in the clinic. In this review, we summarize recent preclinical studies on liposomal cancer vaccines. We describe the basic ingredients for liposomal cancer vaccines, tumor antigens, and adjuvants, and how their combined inclusion together with targeting moieties potentially derived from pathogens can enhance vaccine immunogenicity. We discuss newly identified antigen-presenting cells in humans and mice that pose as promising targets for cancer vaccines. The lessons learned from these preclinical studies can be applied to enhance the efficacy of liposomal cancer vaccination in the clinic.
Collapse
Affiliation(s)
- Maarten K. Nijen Twilhaar
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands;
| | - Lucas Czentner
- Department of Pharmaceutics, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands; (L.C.); (C.F.v.N.); (G.S.)
| | - Cornelus F. van Nostrum
- Department of Pharmaceutics, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands; (L.C.); (C.F.v.N.); (G.S.)
| | - Gert Storm
- Department of Pharmaceutics, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands; (L.C.); (C.F.v.N.); (G.S.)
- Department of Biomaterials, Science and Technology, Faculty of Science and Technology, University of Twente, 7522 NB Enschede, The Netherlands
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Joke M. M. den Haan
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands;
| |
Collapse
|
17
|
Donkor M, Jones HP. The Proposition of the Pulmonary Route as an Attractive Drug Delivery Approach of Nano-Based Immune Therapies and Cancer Vaccines to Treat Lung Tumors. FRONTIERS IN NANOTECHNOLOGY 2021. [DOI: 10.3389/fnano.2021.635194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Lung cancer is the leading cause of cancer related deaths globally, making it a major health concern. The lung’s permissive rich microenvironment is ideal for supporting outgrowth of disseminated tumors from pre-existing extra-pulmonary malignancies usually resulting in high mortality. Tumors occurring in the lungs are difficult to treat, necessitating the need for the development of advanced treatment modalities against primary tumors and secondary lung metastasis. In this review, we explore the pulmonary route as an attractive drug delivery approach to treat lung tumors. We also discuss the potential of pulmonary delivery of cancer vaccine vectors to induce mucosal immunity capable of preventing the seeding of tumors in the lung.
Collapse
|
18
|
Wang S, Sun Z, Hou Y. Engineering Nanoparticles toward the Modulation of Emerging Cancer Immunotherapy. Adv Healthc Mater 2021; 10:e2000845. [PMID: 32790039 DOI: 10.1002/adhm.202000845] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/09/2020] [Indexed: 12/16/2022]
Abstract
Cancer immunotherapy is a new therapeutic strategy to fight cancer by activating the patients' own immune system. At present, immunotherapy approaches such as cancer vaccines, immune checkpoint blockade (ICB), adoptive cell transfer (ACT), monoclonal antibodies (mAbs) therapy, and cytokines therapy have therapeutic potential in preclinical and clinical applications. However, the intrinsic limitations of conventional immunotherapy are difficulty of precise dosage control, insufficient enrichment in tumor tissues, partial immune response silencing or hyperactivity, and high cost. Engineering nanoparticles (NPs) have been emerging as a promising multifunctional platform to enhance conventional immunotherapy due to their intrinsic immunogenicity, convenient delivery function, controlled surface chemistry activity, multifunctional modifying potential, and intelligent targeting. This review presents the recent progress reflected by engineering NPs, including the diversified selection of functionalized NPs, the superiority of engineering NPs for enhancing conventional immunotherapy, and NP-mediated multiscale strategies for synergistic therapy consisting of compositions and their mechanism. Finally, the perspective on multifunctional NP-based cancer immunotherapy for boosting immunomodulation is discussed, which reveals the expanding landscape of engineering NPs in clinical translation.
Collapse
Affiliation(s)
- Shuren Wang
- Beijing Key Laboratory of Magnetoelectric Materials and Devices Department of Materials Science and Engineering College of Engineering Beijing Innovation Centre for Engineering Science and Advanced Technology Peking University Beijing 100871 China
| | - Zhaoli Sun
- Beijing Key Laboratory of Magnetoelectric Materials and Devices Department of Materials Science and Engineering College of Engineering Beijing Innovation Centre for Engineering Science and Advanced Technology Peking University Beijing 100871 China
- College of Life Sciences Peking University Beijing 100871 China
| | - Yanglong Hou
- Beijing Key Laboratory of Magnetoelectric Materials and Devices Department of Materials Science and Engineering College of Engineering Beijing Innovation Centre for Engineering Science and Advanced Technology Peking University Beijing 100871 China
| |
Collapse
|
19
|
Abstract
Currently, peptide-nanoparticle (NP) conjugates have been demonstrated to be efficient and powerful tools for the treatment and the diagnosis of various diseases as well as in the bioimaging application. Several bioconjugation strategies have been adopted to formulate the peptide-NP conjugates. In this review, we discuss the exciting applications of peptide-gold (Au) NP conjugates in the area of drug delivery, targeting, cancer therapy, brain diseases, vaccines, immune modulation, biosensor, colorimetric detection of heavy metals, and bio-labeling in vitro and in vivo models. Within this framework, various approaches such as radiotherapy, photothermal therapy, photodynamic therapy and chemo-photothermal therapy have been demonstrated for the treatment of several diseases. Moreover, we highlight how the morphology, size, density of peptide and the protein corona influence the biological activity, biodistribution and biological fate of peptide-AuNP conjugates. In the end, we discuss the future outlook and the challenges being faced in the clinical translation of the peptide-AuNP conjugates. Overall, this review emphasizes that the peptide-AuNP conjugates might be used as potential theranostic agents for the treatment of life-threatening diseases in an economical fashion in the future.
Collapse
Affiliation(s)
- Akhilesh Rai
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Lino Ferreira
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
20
|
Dykman LA. Gold nanoparticles for preparation of antibodies and vaccines against infectious diseases. Expert Rev Vaccines 2020; 19:465-477. [PMID: 32306785 PMCID: PMC7196924 DOI: 10.1080/14760584.2020.1758070] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 04/16/2020] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Vaccination remains very effective in stimulating protective immune responses against infections. An important task in antibody and vaccine preparation is to choose an optimal carrier that will ensure a high immune response. Particularly promising in this regard are nanoscale particle carriers. An antigen that is adsorbed or encapsulated by nanoparticles can be used as an adjuvant to optimize the immune response during vaccination. a very popular antigen carrier used for immunization and vaccination is gold nanoparticles, with are being used to make new vaccines against viral, bacterial, and parasitic infections. AREAS COVERED This review summarizes what is currently known about the use of gold nanoparticles as an antigen carrier and adjuvant to prepare antibodies in vivo and design vaccines against viral, bacterial, and parasitic infections. The basic principles, recent advances, and current problems in the use of gold nanoparticles are discussed. EXPERT OPINION Gold nanoparticles can be used as adjuvants to increase the effectiveness of vaccines by stimulating antigen-presenting cells and ensuring controlled antigen release. Studying the characteristics of the immune response obtained from the use of gold nanoparticles as a carrier and an adjuvant will permit the particles' potential for vaccine design to be increased.
Collapse
Affiliation(s)
- Lev A. Dykman
- Institute of Biochemistry and Physiology of Plants and Microorganisms, Russian Academy of Sciences, Saratov, Russia
| |
Collapse
|
21
|
Shields CW, Wang LLW, Evans MA, Mitragotri S. Materials for Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1901633. [PMID: 31250498 DOI: 10.1002/adma.201901633] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 04/17/2019] [Indexed: 05/20/2023]
Abstract
Breakthroughs in materials engineering have accelerated the progress of immunotherapy in preclinical studies. The interplay of chemistry and materials has resulted in improved loading, targeting, and release of immunomodulatory agents. An overview of the materials that are used to enable or improve the success of immunotherapies in preclinical studies is presented, from immunosuppressive to proinflammatory strategies, with particular emphasis on technologies poised for clinical translation. The materials are organized based on their characteristic length scale, whereby the enabling feature of each technology is organized by the structure of that material. For example, the mechanisms by which i) nanoscale materials can improve targeting and infiltration of immunomodulatory payloads into tissues and cells, ii) microscale materials can facilitate cell-mediated transport and serve as artificial antigen-presenting cells, and iii) macroscale materials can form the basis of artificial microenvironments to promote cell infiltration and reprogramming are discussed. As a step toward establishing a set of design rules for future immunotherapies, materials that intrinsically activate or suppress the immune system are reviewed. Finally, a brief outlook on the trajectory of these systems and how they may be improved to address unsolved challenges in cancer, infectious diseases, and autoimmunity is presented.
Collapse
Affiliation(s)
- C Wyatt Shields
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
| | - Lily Li-Wen Wang
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Michael A Evans
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
| |
Collapse
|
22
|
Hashemi V, Farhadi S, Ghasemi Chaleshtari M, Seashore-Ludlow B, Masjedi A, Hojjat-Farsangi M, Namdar A, Ajjoolabady A, Mohammadi H, Ghalamfarsa G, Jadidi-Niaragh F. Nanomedicine for improvement of dendritic cell-based cancer immunotherapy. Int Immunopharmacol 2020; 83:106446. [PMID: 32244048 DOI: 10.1016/j.intimp.2020.106446] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/24/2020] [Accepted: 03/24/2020] [Indexed: 02/08/2023]
Abstract
Dendritic cell (DC)-based cancer immunotherapy has shown impressive outcomes, including the development of the first FDA-approved anti-cancer vaccine. However, the clinical application of DC-based cancer immunotherapy is associated with various challenges. Promising novel tools for the administration of cancer vaccines has emerged from recent developments in nanoscale biomaterials. One current strategy to enhance targeted drug delivery, while minimizing drug-related toxicities, is the use of nanoparticles (NPs). These can be utilized for antigen delivery into DCs, which have been shown to provide potent T cell-stimulating effects. Therefore, NP delivery represents one promising approach for creating an effective and stable immune response without toxic side effects. The current review surveys cancer immunotherapy with particular attention toward NP-based delivery methods that target DCs.
Collapse
Affiliation(s)
- Vida Hashemi
- Department of Basic Science, Faculty of Medicine, Maragheh University of Medical Sciences, Maragheh, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shohreh Farhadi
- Student Research Committee, Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Brinton Seashore-Ludlow
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institute, Stockholm, Sweden
| | - Ali Masjedi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Hojjat-Farsangi
- Bioclinicum, Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden; The Persian Gulf Marine Biotechnology Medicine Research Center, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Afshin Namdar
- Department of Oncology, Cross Cancer Institute, The University of Alberta, Edmonton, Alberta, Canada
| | - Amir Ajjoolabady
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Mohammadi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Ghasem Ghalamfarsa
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran.
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
23
|
Bhargava A, Srivastava RK, Mishra DK, Tiwari RR, Sharma RS, Mishra PK. Dendritic cell engineering for selective targeting of female reproductive tract cancers. Indian J Med Res 2019; 148:S50-S63. [PMID: 30964081 PMCID: PMC6469378 DOI: 10.4103/ijmr.ijmr_224_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Female reproductive tract cancers (FRCs) are considered as one of the most frequently occurring malignancies and a foremost cause of death among women. The late-stage diagnosis and limited clinical effectiveness of currently available mainstay therapies, primarily due to the developed drug resistance properties of tumour cells, further increase disease severity. In the past decade, dendritic cell (DC)-based immunotherapy has shown remarkable success and appeared as a feasible therapeutic alternative to treat several malignancies, including FRCs. Importantly, the clinical efficacy of this therapy is shown to be restricted by the established immunosuppressive tumour microenvironment. However, combining nanoengineered approaches can significantly assist DCs to overcome this tumour-induced immune tolerance. The prolonged release of nanoencapsulated tumour antigens helps improve the ability of DC-based therapeutics to selectively target and remove residual tumour cells. Incorporation of surface ligands and co-adjuvants may further aid DC targeting (in vivo) to overcome the issues associated with the short DC lifespan, immunosuppression and imprecise uptake. We herein briefly discuss the necessity and progress of DC-based therapeutics in FRCs. The review also sheds lights on the future challenges to design and develop clinically effective nanoparticles-DC combinations that can induce efficient anti-tumour immune responses and prolong patients’ survival.
Collapse
Affiliation(s)
- Arpit Bhargava
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | | | - Dinesh Kumar Mishra
- School of Pharmacy & Technology Management, Narsee Monjee Institute of Management & Studies, Shirpur, India
| | - Rajnarayan R Tiwari
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Radhey Shyam Sharma
- Division of Reproductive Biology, Maternal & Child Health, Indian Council of Medical Research, New Delhi, India
| | - Pradyumna Kumar Mishra
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| |
Collapse
|
24
|
Nandedkar-Kulkarni N, Vartak AR, Sucheck SJ, Wall KA, Quinn A, Morran MP, McInerney MF. Development of a Bioconjugate Platform for Modifying the Immune Response of Autoreactive Cytotoxic T Lymphocytes Involved in Type 1 Diabetes. Bioconjug Chem 2019; 30:2049-2059. [PMID: 31274300 DOI: 10.1021/acs.bioconjchem.9b00332] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Type 1 diabetes (T1D) is an autoimmune disorder characterized by autoimmune cell mediated destruction of pancreatic beta cells. Pancreatic beta cells are the only source of insulin in the body. T1D patients then have to depend on insulin injections for their lifetime. Insulin injection can modulate the blood sugar levels, but insulin has little effect on the autoimmune process. Altered peptide ligands (APL) derived from known autoantigens in T1D are able to induce tolerance in autoreactive cells in T1D animal models, but are currently unable to elicit this protection in humans. There is a need to improve immunogenicity of the APLs, as these short peptides can be easily degraded by enzymes in the blood. GAD546-554 is a dominant epitope recognized by autoreactive T cells in the nonobese diabetic (NOD) mouse model that can cause destruction of beta cells. Alanine substitution at the eighth position of GAD546-554 peptide (APL9) induced tolerance in a GAD546-554 specific cytotoxic T lymphocyte clone. To improve the antigen presentation and endosomal escape of APL9, we developed a bioconjugate platform that consists of a liposome containing a bioconjugate of APL9 and toll-like receptor 2 ligand Pam3CysSK4 as well as an antibody against macrophage protein F4/80. APL9 bioconjugate liposome with F4/80 antibody was able to induce tolerance in a GAD 546-554 specific clone. Diabetic NOD splenocytes pretreated with APL9 bioconjugate were also not able to transfer diabetes into prediabetic NOD recipient mice. This work is beneficial to prevent T1D as an immunotherapy strategy to render autoreactive immune cells more tolerant of beta cells.
Collapse
Affiliation(s)
- Neha Nandedkar-Kulkarni
- Department of Medicinal and Biological Chemistry , University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Health Science Campus , 3000 Arlington Avenue , Toledo , Ohio 43614 , United States
| | - Abhishek R Vartak
- Department of Chemistry and Biochemistry , University of Toledo, College of Natural Sciences and Mathematics , Main Campus, 2801 West Bancroft Street , Toledo , Ohio 43606 , United States
| | - Steven J Sucheck
- Department of Chemistry and Biochemistry , University of Toledo, College of Natural Sciences and Mathematics , Main Campus, 2801 West Bancroft Street , Toledo , Ohio 43606 , United States
| | - Katherine A Wall
- Department of Medicinal and Biological Chemistry , University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Health Science Campus , 3000 Arlington Avenue , Toledo , Ohio 43614 , United States
| | - Anthony Quinn
- Department of Biological Sciences , University of Toledo, College of Natural Sciences and Mathematics , Main Campus, 2801 West Bancroft Street , Toledo , Ohio 43606 , United States
| | - Michael P Morran
- Department of Medicinal and Biological Chemistry , University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Health Science Campus , 3000 Arlington Avenue , Toledo , Ohio 43614 , United States
| | - Marcia F McInerney
- Department of Medicinal and Biological Chemistry , University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Health Science Campus , 3000 Arlington Avenue , Toledo , Ohio 43614 , United States.,Center for Diabetes and Endocrine Research , University of Toledo, Health Science Campus , 3000 Arlington Avenue , Toledo , Ohio 43614 , United States
| |
Collapse
|
25
|
Dykman LA, Khlebtsov NG. Gold nanoparticles in chemo-, immuno-, and combined therapy: review [Invited]. BIOMEDICAL OPTICS EXPRESS 2019; 10:3152-3182. [PMID: 31467774 PMCID: PMC6706047 DOI: 10.1364/boe.10.003152] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 05/30/2019] [Accepted: 05/30/2019] [Indexed: 05/19/2023]
Abstract
Functionalized gold nanoparticles (GNPs) with controlled geometrical and optical properties have been the subject of intense research and biomedical applications. This review summarizes recent data and topical problems in nanomedicine that are related to the use of variously sized, shaped, and structured GNPs. We focus on three topical fields in current nanomedicine: (1) use of GNP-based nanoplatforms for the targeted delivery of anticancer and antimicrobial drugs and of genes; (2) GNP-based cancer immunotherapy; and (3) combined chemo-, immuno-, and phototherapy. We present a summary of the available literature data and a short discussion of future work.
Collapse
Affiliation(s)
- L A Dykman
- Institute of Biochemistry and Physiology of Plants and Microorganisms, Russian Academy of Sciences, 13 Prospekt Entuziastov, Saratov 410049, Russia
| | - N G Khlebtsov
- Institute of Biochemistry and Physiology of Plants and Microorganisms, Russian Academy of Sciences, 13 Prospekt Entuziastov, Saratov 410049, Russia
- Saratov National Research State University, 83 Ulitsa Astrakhanskaya, Saratov 410012, Russia
| |
Collapse
|
26
|
Cruz LJ, Tacken PJ, van der Schoot JMS, Rueda F, Torensma R, Figdor CG. ICAM3-Fc Outperforms Receptor-Specific Antibodies Targeted Nanoparticles to Dendritic Cells for Cross-Presentation. Molecules 2019; 24:molecules24091825. [PMID: 31083610 PMCID: PMC6540027 DOI: 10.3390/molecules24091825] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/07/2019] [Accepted: 05/08/2019] [Indexed: 12/27/2022] Open
Abstract
Optimal targeting of nanoparticles (NP) to dendritic cells (DCs) receptors to deliver cancer-specific antigens is key to the efficient induction of anti-tumour immune responses. Poly (lactic-co-glycolic acid) (PLGA) nanoparticles containing tètanus toxoid and gp100 melanoma-associated antigen, toll-like receptor adjuvants were targeted to the DC-SIGN receptor in DCs by specific humanized antibodies or by ICAM3-Fc fusion proteins, which acts as the natural ligand. Despite higher binding and uptake efficacy of anti-DC-SIGN antibody-targeted NP vaccines than ICAM3-Fc ligand, no difference were observed in DC activation markers CD80, CD83, CD86 and CCR7 induced. DCs loaded with NP coated with ICAM3-Fc appeared more potent in activating T cells via cross-presentation than antibody-coated NP vaccines. This fact could be very crucial in the design of new cancer vaccines.
Collapse
Affiliation(s)
- Luis J Cruz
- Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands.
| | - Paul J Tacken
- Department of Tumor Immunology, Radboud Insititute for Molecular Life Sciences, Radboud University Medical Center, Postbox 9101, 6500 HB Nijmegen, The Netherlands.
| | - Johan M S van der Schoot
- Department of Tumor Immunology, Radboud Insititute for Molecular Life Sciences, Radboud University Medical Center, Postbox 9101, 6500 HB Nijmegen, The Netherlands.
| | - Felix Rueda
- Department of Biochemistry and Molecular Biology, University of Barcelona, Diagonal 643, 08028 Barcelona, Spain.
| | - Ruurd Torensma
- Department of Tumor Immunology, Radboud Insititute for Molecular Life Sciences, Radboud University Medical Center, Postbox 9101, 6500 HB Nijmegen, The Netherlands.
| | - Carl G Figdor
- Department of Tumor Immunology, Radboud Insititute for Molecular Life Sciences, Radboud University Medical Center, Postbox 9101, 6500 HB Nijmegen, The Netherlands.
| |
Collapse
|
27
|
Hossain MK, Vartak A, Sucheck SJ, Wall KA. Liposomal Fc Domain Conjugated to a Cancer Vaccine Enhances Both Humoral and Cellular Immunity. ACS OMEGA 2019; 4:5204-5208. [PMID: 30949616 PMCID: PMC6441943 DOI: 10.1021/acsomega.9b00029] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 02/27/2019] [Indexed: 06/09/2023]
Abstract
Targeted delivery of antigens to antigen-presenting cells (APCs) by utilizing natural anticarbohydrate antibodies is a promising approach for selective uptake and enhanced antigen presentation. Previously, we reported that in the presence of a natural antibody, anti-rhamnose antibody (anti-Rha), the bacterial sugar rhamnose conjugated with liposomal cancer antigen MUC1-Tn enhances antigen presentation by APCs such as dendritic cells by targeting Fc gamma receptors. The idea was to utilize the natural human anti-Rha antibodies present in human serum for targeted delivery of cancer-specific antigens. Recently, we found that the IgG3 antibody isotype was the most prevalent anti-Rha antibody generated in mice immunized with rhamnose-Ficoll (Rha-Ficoll) antigen. In this manuscript, we have conjugated the murine IgG3-Fc with a MUC1-containing cancer vaccine and compared the humoral and cellular immune response to this vaccine with one targeted via the human anti-Rha antibody and to the MUC1 vaccine alone. This Fc approach enhanced antibody production and T-cell proliferation almost to the same level as using the anti-Rha antibody. These results suggest that targeting Fc directly to dendritic cells can be an alternative approach to human anti-Rha for generating effective antigen-primed T-cells.
Collapse
Affiliation(s)
- Md Kamal Hossain
- Department
of Medicinal and Biological Chemistry and Department of Chemistry and Biochemistry, University of Toledo, Toledo, Ohio 43606, United States
| | - Abhishek Vartak
- Department
of Medicinal and Biological Chemistry and Department of Chemistry and Biochemistry, University of Toledo, Toledo, Ohio 43606, United States
| | - Steven J. Sucheck
- Department
of Medicinal and Biological Chemistry and Department of Chemistry and Biochemistry, University of Toledo, Toledo, Ohio 43606, United States
| | - Katherine A. Wall
- Department
of Medicinal and Biological Chemistry and Department of Chemistry and Biochemistry, University of Toledo, Toledo, Ohio 43606, United States
| |
Collapse
|
28
|
Junco JA, Rodríguez R, Fuentes F, Baladrón I, Castro MD, Calzada L, Valenzuela C, Bover E, Pimentel E, Basulto R, Arteaga N, Cid-Arregui A, Sariol F, González L, Porres-Fong L, Medina M, Rodríguez A, Garay AH, Reyes O, López M, de Quesada L, Alvarez A, Martínez C, Marrero M, Molero G, Guerra A, Rosales P, Capote C, Acosta S, Vela I, Arzuaga L, Campal A, Ruiz E, Rubio E, Cedeño P, Sánchez MC, Cardoso P, Morán R, Fernández Y, Campos M, Touduri H, Bacardi D, Feria I, Ramirez A, Cosme K, Saura PL, Quintana M, Muzio V, Bringas R, Ayala M, Mendoza M, Fernández LE, Carr A, Herrera L, Guillén G. Safety and Therapeutic Profile of a GnRH-Based Vaccine Candidate Directed to Prostate Cancer. A 10-Year Follow-Up of Patients Vaccinated With Heberprovac. Front Oncol 2019; 9:49. [PMID: 30859088 PMCID: PMC6397853 DOI: 10.3389/fonc.2019.00049] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 01/17/2019] [Indexed: 12/25/2022] Open
Abstract
Heberprovac is a GnRH based vaccine candidate containing 2.4 mg of the GnRHm1-TT peptide as the main active principle; 245 μg of the very small size proteoliposomes adjuvant (VSSP); and 350 μL of Montanide ISA 51 VG oil adjuvant. The aim of this study was to assess the safety and tolerance of the Heberprovac in advanced prostate cancer patients as well as its capacity to induce anti-GnRH antibodies, the subsequent effects on serum levels of testosterone and PSA and the patient overall survival. The study included eight patients with histologically-proven advanced prostate cancer with indication for hormonal therapy, who received seven intramuscular immunizations with Heberprovac within 18 weeks. Anti-GnRH antibody titers, testosterone and PSA levels, as well as clinical parameters were recorded and evaluated. The vaccine was well tolerated. Significant reductions in serum levels of testosterone and PSA were seen after four immunizations. Castrate levels of testosterone were observed in all patients at the end of the immunization schedule, which remained at the lowest level for at least 20 months. In a 10-year follow-up three out of six patients who completed the entire trial survived. In contrast only one out eight patients survived in the same period in a matched randomly selected group receiving standard anti-hormonal treatment. Heberprovac vaccination showed a good security profile, as well as immunological, biochemical and, most importantly, clinical benefit. The vaccinated group displayed survival advantage compared with the reference group that received standard treatment. These results warrant further clinical trials with Heberprovac involving a larger cohort.
Collapse
Affiliation(s)
- Jesús A. Junco
- Center for Genetic Engineering and Biotechnology of Camaguey, Camagüey, Cuba
| | - Ranfis Rodríguez
- Uro-oncology Department of National Institute of Oncology and Radiobiology (INOR), Havana, Cuba
| | - Franklin Fuentes
- Center for Genetic Engineering and Biotechnology of Camaguey, Camagüey, Cuba
| | - Idania Baladrón
- Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Maria D. Castro
- Center for Genetic Engineering and Biotechnology of Camaguey, Camagüey, Cuba
| | - Lesvia Calzada
- Center for Genetic Engineering and Biotechnology of Camaguey, Camagüey, Cuba
| | | | - Eddy Bover
- Center for Genetic Engineering and Biotechnology of Camaguey, Camagüey, Cuba
| | | | - Roberto Basulto
- Center for Genetic Engineering and Biotechnology of Camaguey, Camagüey, Cuba
| | - Niurka Arteaga
- Center for Genetic Engineering and Biotechnology of Camaguey, Camagüey, Cuba
| | | | | | | | | | - María Medina
- Oncologic Hospital of Camaguey, Marie Curie, Camagüey, Cuba
| | - Ayni Rodríguez
- Department of Pharmacology of Camaguey Medical University, Camagüey, Cuba
| | - A. Hilda Garay
- Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Osvaldo Reyes
- Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Matilde López
- Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | | | | | | | | | | | - Alfredo Guerra
- Department of Pharmacology of Camaguey Medical University, Camagüey, Cuba
| | - Pedro Rosales
- Oncologic Hospital of Camaguey, Marie Curie, Camagüey, Cuba
| | - Carlos Capote
- Amalia Simoni Clinical-Surgical Hospital, Camagüey, Cuba
| | - Sahily Acosta
- Oncologic Hospital of Camaguey, Marie Curie, Camagüey, Cuba
| | - Idania Vela
- Oncologic Hospital of Camaguey, Marie Curie, Camagüey, Cuba
| | - Lina Arzuaga
- Oncologic Hospital of Camaguey, Marie Curie, Camagüey, Cuba
| | - Ana Campal
- Center for Genetic Engineering and Biotechnology of Camaguey, Camagüey, Cuba
| | - Erlán Ruiz
- Oncologic Hospital of Camaguey, Marie Curie, Camagüey, Cuba
| | - Elier Rubio
- Oncologic Hospital of Camaguey, Marie Curie, Camagüey, Cuba
| | - Pável Cedeño
- Oncologic Hospital of Camaguey, Marie Curie, Camagüey, Cuba
| | - María Carmen Sánchez
- Clinical Laboratory of the Oncologic Hospital of Camaguey, Marie Curie, Camagüey, Cuba
| | - Pedro Cardoso
- Oncologic Hospital of Camaguey, Marie Curie, Camagüey, Cuba
| | - Rolando Morán
- Center for Genetic Engineering and Biotechnology of Camaguey, Camagüey, Cuba
| | - Yairis Fernández
- Department of Pharmacology of Camaguey Medical University, Camagüey, Cuba
| | - Magalys Campos
- Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Henio Touduri
- Department of Pharmacology of Camaguey Medical University, Camagüey, Cuba
| | - Dania Bacardi
- Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Indalecio Feria
- Clinical Trials Department of Oncologic Hospital Marie Curie of Camaguey, Marie Curie, Camagüey, Cuba
| | - Amilcar Ramirez
- Department of Pharmacology of Camaguey Medical University, Camagüey, Cuba
| | - Karelia Cosme
- Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | | | | | - Verena Muzio
- Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Ricardo Bringas
- Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Marta Ayala
- Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Mario Mendoza
- Oncologic Hospital of Camaguey, Marie Curie, Camagüey, Cuba
| | | | | | - Luis Herrera
- Center for Genetic Engineering and Biotechnology, Havana, Cuba
- BioCubafarma, Havana, Cuba
| | - Gerardo Guillén
- Center for Genetic Engineering and Biotechnology, Havana, Cuba
| |
Collapse
|
29
|
Englinger B, Pirker C, Heffeter P, Terenzi A, Kowol CR, Keppler BK, Berger W. Metal Drugs and the Anticancer Immune Response. Chem Rev 2018; 119:1519-1624. [DOI: 10.1021/acs.chemrev.8b00396] [Citation(s) in RCA: 174] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Bernhard Englinger
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Christine Pirker
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Petra Heffeter
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Alessio Terenzi
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 42, A-1090 Vienna, Austria
| | - Christian R. Kowol
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 42, A-1090 Vienna, Austria
| | - Bernhard K. Keppler
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 42, A-1090 Vienna, Austria
| | - Walter Berger
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
| |
Collapse
|
30
|
Recent advances in applying nanotechnologies for cancer immunotherapy. J Control Release 2018; 288:239-263. [PMID: 30223043 DOI: 10.1016/j.jconrel.2018.09.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/11/2018] [Accepted: 09/12/2018] [Indexed: 12/13/2022]
Abstract
Cancer immunotherapy aimed at boosting cancer-specific immunoresponses to eradicate tumor cells has evolved as a new treatment modality. Nanoparticles incorporating antigens and immunomodulatory agents can activate immune cells and modulate the tumor microenvironment to enhance anti-tumor immunity. The nanotechnology approach has been demonstrated to be superior to standard formulations in in-vivo settings. In this article, we focus on recent advances made within the last 5 years in nanoparticle-based cancer immunotherapy, including peptide- and nucleic acid-based nanovaccines, nanomedicines containing an immunoadjuvant to activate anti-tumor immunity, nanoparticle delivery of immune checkpoint inhibitors and the combination of the above approaches. Encouraging results and new emerging nanotechnologies in drug delivery promise the continuous growth of this field and ultimately clinical translation of enhanced immunotherapy of cancer.
Collapse
|
31
|
Combinatory therapy adopting nanoparticle-based cancer vaccination with immune checkpoint blockade for treatment of post-surgical tumor recurrences. J Control Release 2018; 285:56-66. [DOI: 10.1016/j.jconrel.2018.07.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 07/05/2018] [Accepted: 07/05/2018] [Indexed: 12/20/2022]
|
32
|
Hossain MK, Vartak A, Karmakar P, Sucheck SJ, Wall KA. Augmenting Vaccine Immunogenicity through the Use of Natural Human Anti-rhamnose Antibodies. ACS Chem Biol 2018; 13:2130-2142. [PMID: 29916701 PMCID: PMC6103300 DOI: 10.1021/acschembio.8b00312] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
![]()
Utilizing
natural antibodies to augment vaccine immunogenicity
is a promising approach toward cancer immunotherapy. Anti-rhamnose
(anti-Rha) antibodies are some of the most common natural anti-carbohydrate
antibodies present in human serum. Therefore, rhamnose can be utilized
as a targeting moiety for a rhamnose-containing vaccine to prepare
an effective vaccine formulation. It was shown previously that anti-Rha
antibody generated in mice binds effectively with Rha-conjugated vaccine
and is picked up by antigen presenting cells (APCs) through stimulatory
Fc receptors. This leads to the effective uptake and processing of
antigen and eventually presentation by major histocompatibility complex
(MHC) molecules. In this article, we show that natural human anti-Rha
antibodies can also be used in a similar mechanism and immunogenicity
can be enhanced by targeting Rha-conjugated antigens. In doing so,
we have purified human anti-Rha antibodies from human serum using
a rhamnose affinity column. In vitro, human anti-Rha
antibodies are shown to enhance the uptake of a model antigen, Rha-ovalbumin
(Rha-Ova), by APCs. In vivo, they improved the priming
of CD4+ T cells to Rha-Ova in comparison to non-anti-Rha human antibodies.
Additionally, increased priming of both CD4+ and CD8+ T cells toward
the cancer antigen MUC1-Tn was observed in mice that received human
anti-Rha antibodies prior to vaccination with a rhamnose-modified
MUC1-Tn cancer vaccine. The vaccine conjugate contained Pam3CysSK4, a Toll-like receptor (TLR) agonist linked via copper-free cycloaddition chemistry to a 20-amino-acid
glycopeptide derived from the tumor marker MUC-1 containing the tumor-associated
carbohydrate antigen α-N-acetyl galactosamine
(GalNAc). The primed CD8+ T cells released IFN-γ and killed
tumor cells. Therefore, we have confirmed that human anti-Rha antibodies
can be effectively utilized as a targeting moiety for making an effective
vaccine.
Collapse
Affiliation(s)
- Md Kamal Hossain
- Dept. of Medicinal and Biological Chemistry, University of Toledo, Toledo, Ohio 43606, United States
| | - Abhishek Vartak
- Dept. of Chemistry and Biochemistry, University of Toledo, Toledo, Ohio 43606, United States
| | - Partha Karmakar
- Dept. of Chemistry and Biochemistry, University of Toledo, Toledo, Ohio 43606, United States
| | - Steven J. Sucheck
- Dept. of Chemistry and Biochemistry, University of Toledo, Toledo, Ohio 43606, United States
| | - Katherine A. Wall
- Dept. of Medicinal and Biological Chemistry, University of Toledo, Toledo, Ohio 43606, United States
| |
Collapse
|
33
|
Martinez-Gil L, Goff PH, Tan GS. The Role of Self-Assembling Lipid Molecules in Vaccination. ADVANCES IN BIOMEMBRANES AND LIPID SELF-ASSEMBLY 2018. [PMCID: PMC7147077 DOI: 10.1016/bs.abl.2017.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The advent of vaccines represents one of the most significant advances in medical history. The protection provided by vaccines has greatly contributed in reducing the number of cases of infections and most notably to the eradication of small pox. A large number of new technologies and approaches in vaccine development are currently being investigated with the goal of providing the basis for the next generation of prophylactics against an ever-expanding list of emerging and reemerging pathogens. In this chapter, we will focus on the role of lipids and lipid self-assembling vesicles in new and promising vaccination approaches. We will start by describing how lipids can induce activation of the innate immune system and focus on some lipid-derived vaccine adjuvants. Next, we will review current lipid-based self-assembling particles used as vaccine platforms, specifically liposomes and virus-like particles, and how virus-like particles have facilitated research of highly pathogenic viruses such as Ebola.
Collapse
|
34
|
Chen P, Liu X, Sun Y, Zhou P, Wang Y, Zhang Y. Dendritic cell targeted vaccines: Recent progresses and challenges. Hum Vaccin Immunother 2017; 12:612-22. [PMID: 26513200 DOI: 10.1080/21645515.2015.1105415] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Dendritic cells (DCs) are known to be a set of morphology, structure and function of heterogeneous professional antigen presenting cells (APCs), as well as the strongest functional antigen presenting cells, which can absorb, process and present antigens. As the key regulators of innate and adaptive immune responses, DCs are at the center of the immune system and capable of interacting with both B cells and T cells, thereby manipulating the humoral and cellular immune responses. DCs provide an essential link between the innate and adaptive immunity, and the strong immune activation function of DCs and their properties of natural adjuvants, make them a valuable target for antigen delivery. Targeting antigens to DC-specific endocytic receptors in combination with the relevant antibodies or ligands along with immunostimulatory adjuvants has been recently recognized as a promising strategy for designing an effective vaccine that elicits a strong and durable T cell response against intracellular pathogens and cancer. This opinion article provides a brief summary of the rationales, superiorities and challenges of existing DC-targeting approaches.
Collapse
Affiliation(s)
- Pengfei Chen
- a State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences , Lanzhou , China
| | - Xinsheng Liu
- a State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences , Lanzhou , China
| | - Yuefeng Sun
- a State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences , Lanzhou , China
| | - Peng Zhou
- a State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences , Lanzhou , China
| | - Yonglu Wang
- a State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences , Lanzhou , China
| | - Yongguang Zhang
- a State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences , Lanzhou , China
| |
Collapse
|
35
|
Song W, Musetti SN, Huang L. Nanomaterials for cancer immunotherapy. Biomaterials 2017; 148:16-30. [PMID: 28961532 DOI: 10.1016/j.biomaterials.2017.09.017] [Citation(s) in RCA: 183] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Revised: 09/07/2017] [Accepted: 09/17/2017] [Indexed: 02/08/2023]
Abstract
Cancer immunotherapy is quickly growing to be the fourth most important cancer therapy, after surgery, radiation therapy, and chemotherapy. Immunotherapy is the most promising cancer management strategy because it orchestrates the body's own immune system to target and eradicate cancer cells, which may result in durable antitumor responses and reduce metastasis and recurrence more than traditional treatments. Nanomaterials hold great promise in further improving the efficiency of cancer immunotherapy - in many cases, they are even necessary for effective delivery. In this review, we briefly summarize the basic principles of cancer immunotherapy and explain why and where to apply nanomaterials in cancer immunotherapy, with special emphasis on cancer vaccines and tumor microenvironment modulation.
Collapse
Affiliation(s)
- Wantong Song
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China
| | - Sara N Musetti
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Leaf Huang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
36
|
Wang C, Ye Y, Hu Q, Bellotti A, Gu Z. Tailoring Biomaterials for Cancer Immunotherapy: Emerging Trends and Future Outlook. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29. [PMID: 28556553 DOI: 10.1002/adma.201606036] [Citation(s) in RCA: 194] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 03/04/2017] [Indexed: 05/05/2023]
Abstract
Cancer immunotherapy, as a paradigm shift in cancer treatment, has recently received tremendous attention. The active cancer vaccination, immune checkpoint blockage (ICB) and chimeric antigen receptor (CAR) for T-cell-based adoptive cell transfer are among these developments that have achieved a significant increase in patient survival in clinical trials. Despite these advancements, emerging research at the interdisciplinary interface of cancer biology, immunology, bioengineering, and materials science is important to further enhance the therapeutic benefits and reduce side effects. Here, an overview of the latest studies on engineering biomaterials for the enhancement of anticancer immunity is given, including the perspectives of delivery of immunomodulatory therapeutics, engineering immune cells, and constructing immune-modulating scaffolds. The opportunities and challenges in this field are also discussed.
Collapse
Affiliation(s)
- Chao Wang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27695, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Yanqi Ye
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27695, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Quanyin Hu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27695, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Adriano Bellotti
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27695, USA
- Department of Medicine University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Zhen Gu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27695, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Medicine University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| |
Collapse
|
37
|
Effect of TLR ligands co-encapsulated with multiepitopic antigen in nanoliposomes targeted to human DCs via Fc receptor for cancer vaccines. Immunobiology 2017. [PMID: 28624137 DOI: 10.1016/j.imbio.2017.06.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Nanoliposomes (NLs) hold promise as new highly specific nanomedicine for anti-tumor vaccines, since they could be targeted to specific receptors on dendritic cell (DC) to induce maturation and activation and increase the anti-tumor immune response. Here we studied a NLs formulation targeted or not to FcR (the receptor for the IgG Fc fragment) for the treatment of androgen-responsive prostate cancer. Luteinizing-hormone-releasing hormone (LHRH) peptide (B- and T-cell epitopes), in tandem with a tetanus toxoid T-helper epitope (830-844 region) and several TLR (Toll-Like Receptor) ligands as adjuvants were co-encapsulated. Specific uptake in vitro of LHRH-TT liposomes targeted to the FcRs of human DCs was enhanced. DC maturation/activation, cytokine production and lymphocyte activation were consistently higher in targeted than non-targeted liposomes. Similar increase was observed as more adjuvants were administrated. Targeting to specific receptor and co-encapsulation of several TLR adjuvants are essential factors for the immune response in peptide based liposome vaccine.
Collapse
|
38
|
Gunay G, Sardan Ekiz M, Ferhati X, Richichi B, Nativi C, Tekinay AB, Guler MO. Antigenic GM3 Lactone Mimetic Molecule Integrated Mannosylated Glycopeptide Nanofibers for the Activation and Maturation of Dendritic Cells. ACS APPLIED MATERIALS & INTERFACES 2017; 9:16035-16042. [PMID: 28445638 DOI: 10.1021/acsami.7b04094] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The ability of dendritic cells to coordinate innate and adaptive immune responses makes them essential targets for vaccination strategies. Presentation of specific antigens by dendritic cells is required for the activation of the immune system against many pathogens and tumors, and nanoscale materials can be functionalized for active targeting of dendritic cells. In this work, we integrated an immunogenic, carbohydrate melanoma-associated antigen-mimetic GM3-lactone molecule into mannosylated peptide amphiphile nanofibers to target dendritic cells through DC-SIGN receptor. Based on morphological and functional analyses, when dendritic cells were treated with peptide nanofiber carriers, they showed significant increase in antigen internalization and a corresponding increase in the surface expression of the activation and maturation markers CD86, CD83 and HLA-DR, in addition to exhibiting a general morphology consistent with dendritic cell maturation. These results indicate that mannosylated peptide amphiphile nanofiber carriers are promising candidates to target dendritic cells for antigen delivery.
Collapse
Affiliation(s)
- Gokhan Gunay
- Institute of Materials Science and Nanotechnology, National Nanotechnology Research Center (UNAM), Bilkent University , Ankara 06800, Turkey
- Neuroscience Graduate Program, Bilkent University , Ankara 06800, Turkey
| | - Melis Sardan Ekiz
- Institute of Materials Science and Nanotechnology, National Nanotechnology Research Center (UNAM), Bilkent University , Ankara 06800, Turkey
| | - Xhenti Ferhati
- Department of Chemistry "Ugo Schiff", University of Florence , Sesto Fiorentino, Florence 50019, Italy
| | - Barbara Richichi
- Department of Chemistry "Ugo Schiff", University of Florence , Sesto Fiorentino, Florence 50019, Italy
| | - Cristina Nativi
- Department of Chemistry "Ugo Schiff", University of Florence , Sesto Fiorentino, Florence 50019, Italy
| | - Ayse B Tekinay
- Institute of Materials Science and Nanotechnology, National Nanotechnology Research Center (UNAM), Bilkent University , Ankara 06800, Turkey
- Neuroscience Graduate Program, Bilkent University , Ankara 06800, Turkey
| | - Mustafa O Guler
- Institute of Materials Science and Nanotechnology, National Nanotechnology Research Center (UNAM), Bilkent University , Ankara 06800, Turkey
- Institute for Molecular Engineering, University of Chicago , Chicago, Illinois 60637, United States
| |
Collapse
|
39
|
Hu X, Wu T, Bao Y, Zhang Z. Nanotechnology based therapeutic modality to boost anti-tumor immunity and collapse tumor defense. J Control Release 2017; 256:26-45. [PMID: 28434891 DOI: 10.1016/j.jconrel.2017.04.026] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 04/15/2017] [Accepted: 04/18/2017] [Indexed: 12/19/2022]
Abstract
Cancer is still the leading cause of death. While traditional treatments such as surgery, chemotherapy and radiotherapy play dominating roles, recent breakthroughs in cancer immunotherapy indicate that the influence of immune system on cancer development is virtually beyond our expectation. Manipulating the immune system to fight against cancer has been thriving in recent years. Further understanding of tumor anatomy provides opportunities to put a brake on immunosuppression by overcoming tumor intrinsic resistance or modulating tumor microenvironment. Nanotechnology which provides versatile engineered approaches to enhance therapeutic effects may potentially contribute to the development of future cancer treatment modality. In this review, we will focus on the application of nanotechnology both in boosting anti-tumor immunity and collapsing tumor defense.
Collapse
Affiliation(s)
| | | | - Yuling Bao
- Tongji School of Pharmacy, PR China; Department of Pharmacy, Tongji Hospital, PR China
| | - Zhiping Zhang
- Tongji School of Pharmacy, PR China; National Engineering Research Center for Nanomedicine, PR China; Hubei Engineering Research Center for Novel Drug Delivery System, HuaZhong University of Science and Technology, Wuhan 430030, PR China.
| |
Collapse
|
40
|
Dykman LA, Khlebtsov NG. Immunological properties of gold nanoparticles. Chem Sci 2017; 8:1719-1735. [PMID: 28451297 PMCID: PMC5396510 DOI: 10.1039/c6sc03631g] [Citation(s) in RCA: 154] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Accepted: 11/14/2016] [Indexed: 12/14/2022] Open
Abstract
In the past decade, gold nanoparticles have attracted strong interest from the nanobiotechnological community owing to the significant progress made in robust and easy-to-make synthesis technologies, in surface functionalization, and in promising biomedical applications. These include bioimaging, gene diagnostics, analytical sensing, photothermal treatment of tumors, and targeted delivery of various biomolecular and chemical cargos. For the last-named application, gold nanoparticles should be properly fabricated to deliver the cargo into the targeted cells through effective endocytosis. In this review, we discuss recent progress in understanding the selective penetration of gold nanoparticles into immune cells. The interaction of gold nanoparticles with immune cell receptors is discussed. As distinct from other published reviews, we present a summary of the immunological properties of gold nanoparticles. This review also summarizes what is known about the application of gold nanoparticles as an antigen carrier and adjuvant in immunization for the preparation of antibodies in vivo. For each of the above topics, the basic principles, recent advances, and current challenges are discussed. Thus, this review presents a detailed analysis of data on interaction of gold nanoparticles with immune cells. Emphasis is placed on the systematization of data over production of antibodies by using gold nanoparticles and adjuvant properties of gold nanoparticles. Specifically, we start our discussion with current data on interaction of various gold nanoparticles with immune cells. The next section describes existing technologies to improve production of antibodies in vivo by using gold nanoparticles conjugated with specific ligands. Finally, we describe what is known about adjuvant properties of bare gold or functionalized nanoparticles. In the Conclusion section, we present a short summary of reported data and some challenges and perspectives.
Collapse
Affiliation(s)
- Lev A Dykman
- Institute of Biochemistry and Physiology of Plants and Microorganisms , Russian Academy of Sciences , 13 Prospekt Entuziastov , Saratov 410049 , Russia . ;
| | - Nikolai G Khlebtsov
- Institute of Biochemistry and Physiology of Plants and Microorganisms , Russian Academy of Sciences , 13 Prospekt Entuziastov , Saratov 410049 , Russia . ;
- Saratov National Research State University , 83 Ulitsa Astrakhanskaya , Saratov 410012 , Russia
| |
Collapse
|
41
|
Chang TZ, Diambou I, Kim JR, Wang B, Champion JA. Host- and pathogen-derived adjuvant coatings on protein nanoparticle vaccines. Bioeng Transl Med 2017; 2:120-130. [PMID: 28516165 PMCID: PMC5412930 DOI: 10.1002/btm2.10052] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 12/02/2016] [Accepted: 12/03/2016] [Indexed: 02/01/2023] Open
Abstract
Nanoparticulate and molecular adjuvants have shown great efficacy in enhancing immune responses, and the immunogenic vaccines of the future will most likely contain both. To investigate the immunostimulatory effects of molecular adjuvants on nanoparticle vaccines, we have designed ovalbumin (OVA) protein nanoparticles coated with two different adjuvants-flagellin (FliC) and immunoglobulin M (IgM). These proteins, derived from Salmonella and mice, respectively, are representatives of pathogen- and host-derived molecules that can enhance immune responses. FliC-coated OVA nanoparticles, soluble FliC (sFliC) admixed with OVA nanoparticles, IgM-coated nanoparticles, and OVA-coated nanoparticles were assessed for immunogenicity in an in vivo mouse immunization study. IgM coatings on nanoparticles significantly enhanced both antibody and T cell responses, and promoted IgG2a class switching but not affinity maturation. FliC-coated nanoparticles and FliC-admixed with nanoparticles both triggered IgG2a class switching, but only FliC-coated nanoparticles enhanced antibody affinity maturation. Our findings that affinity maturation and class switching can be directed independently of one another suggest that adjuvant coatings on nanoparticles can be tailored to generate specific vaccine effector responses against different classes of pathogens.
Collapse
Affiliation(s)
- Timothy Z. Chang
- School of Chemical and Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGA 30332
| | - Ishatou Diambou
- School of Chemical and Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGA 30332
| | - Jong Rok Kim
- Institute for Biomedical SciencesGeorgia State UniversityAtlantaGA 30332
| | - Baozhong Wang
- Institute for Biomedical SciencesGeorgia State UniversityAtlantaGA 30332
| | - Julie A. Champion
- School of Chemical and Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGA 30332
| |
Collapse
|
42
|
Chang TZ, Stadmiller SS, Staskevicius E, Champion JA. Effects of ovalbumin protein nanoparticle vaccine size and coating on dendritic cell processing. Biomater Sci 2017; 5:223-233. [PMID: 27918020 PMCID: PMC5285395 DOI: 10.1039/c6bm00500d] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Nanoparticle vaccine delivery platforms are a promising technology for enhancing vaccine immunogenicity. Protein nanoparticles (PNPs), made entirely from antigen, have been shown to induce protective immune responses against influenza. However, the fundamental mechanisms by which PNPs enhance component protein immunogenicity are not understood. Here, we investigate the role of size and coating of model ovalbumin (OVA) PNPs on particle uptake and trafficking, as well as on inflammation and maturation factor expression in dendritic cells (DCs) in vitro. OVA PNPs enhance antigen uptake in a size-independent manner, and experience attenuated endosomal acidification as compared to soluble OVA. OVA PNPs also trigger Fc receptor upregulation. Expression of cytokines IL-1β and TNF-α were PNP size- and coating-dependent, with small (∼270 nm) nanoparticles triggering greater inflammatory cytokine production than large (∼560 nm) particles. IL-1β expression by DCs in response to PNP stimulation implies activation of the inflammasome, a pathway known to be activated by certain types of nanoparticulate adjuvants. The attenuated acidification and pro-inflammatory profile generated by PNPs in DCs demonstrate that physical biomaterial properties can modulate dendritic cell-mediated antigen processing and adjuvancy. In addition to nanoparticles' enhancement of DC antigen uptake, our work suggests that vaccine nanoparticle size and coating are uptake-independent modulators of immunogenicity.
Collapse
Affiliation(s)
- Timothy Z Chang
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Drive NW, Atlanta, GA 30332, USA.
| | - Samantha S Stadmiller
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Drive NW, Atlanta, GA 30332, USA.
| | - Erika Staskevicius
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Drive NW, Atlanta, GA 30332, USA.
| | - Julie A Champion
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Drive NW, Atlanta, GA 30332, USA.
| |
Collapse
|
43
|
Asadi N, Davaran S, Panahi Y, Hasanzadeh A, Malakootikhah J, Fallah Moafi H, Akbarzadeh A. Application of nanostructured drug delivery systems in immunotherapy of cancer: a review. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2016; 45:18-23. [PMID: 27196810 DOI: 10.1080/21691401.2016.1178136] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The cancer immunotherapy method uses the specificity of the immune system to provide a more effective than more conventional treatments, such as chemotherapy and radiotherapy. Immunotherapy has two main strategies (passive or active) to organize the immune system. Passive strategies use advantage of tumor-hyperpermeable cells, which have enhanced permeability and retention effects. Nanoparticles due to their better accumulation within tissues and cells of the immune system are well suitable for delivery of immune therapies such as vaccines or adjuvants. In this review, we explained application of nanotechnology in immunotherapy of cancer.
Collapse
Affiliation(s)
- Nahideh Asadi
- a Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences , Tabriz University of Medical Sciences , Tabriz , Iran.,b Student Research Committee , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Soodabeh Davaran
- a Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Yunes Panahi
- c Chemical Injuries Research Center , Baqiyatallah University of Medical Sciences , Tehran , Iran
| | - Arash Hasanzadeh
- d Laboratory of Biochemistry, Department of Biology, Faculty of Natural Sciences , University of Tabriz , Tabriz , Iran
| | - Javad Malakootikhah
- e Department of New Sciences and Technologies , University of Tehran , Tehran , Iran
| | - Hadi Fallah Moafi
- f Department of Chemistry, Faculty of Science , University of Guilan , Rasht , Iran
| | - Abolfazl Akbarzadeh
- a Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences , Tabriz University of Medical Sciences , Tabriz , Iran.,b Student Research Committee , Tabriz University of Medical Sciences , Tabriz , Iran.,c Chemical Injuries Research Center , Baqiyatallah University of Medical Sciences , Tehran , Iran.,g Drug Applied Research Center , Tabriz University of Medical Sciences , Tabriz , Iran
| |
Collapse
|
44
|
Xu P, Tang S, Jiang L, Yang L, Zhang D, Feng S, Zhao T, Dong Y, He W, Wang R, Zhang J, Liang Z. Nanomaterial-dependent immunoregulation of dendritic cells and its effects on biological activities of contraceptive nanovaccines. J Control Release 2016; 225:252-68. [PMID: 26826303 DOI: 10.1016/j.jconrel.2016.01.043] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 01/19/2016] [Accepted: 01/24/2016] [Indexed: 01/08/2023]
Abstract
Nanovehicles are promising delivery systems for various vaccines. Nevertheless, different biophysicochemical properties of nanoparticles (NPs), dominating their in vitro and in vivo performances for vaccination, remain unclear. We attempted to elucidate the effects of NPs and their pH-sensitivity on in vitro and in vivo efficacy of resulting prophylactic nanovaccines containing a contraceptive peptide (FSHR). To this end, pH-responsive and non-responsive nanovaccines were produced using acetalated β-cyclodextrin (Ac-bCD) and poly(lactic-co-glycolic acid) (PLGA), respectively. Meanwhile, FSHR derived from an epitope of the follicle-stimulating hormone receptor was used as the model antigen. FSHR-containing Ac-bCD and PLGA NPs were successfully prepared by a nanoemulsion technique, leading to well-shaped nanovaccines with high loading efficiency. The pH-sensitivity of Ac-bCD and PLGA nanovaccines was examined by in vitro hydrolysis and antigen release studies. Nanovaccines could be effectively engulfed by dendritic cells (DCs) via endocytosis in both dose and time dependent manners, and their intracellular trafficking was closely related to the pH-sensitivity of the carrier materials. Furthermore, nanovaccines could induce the secretion of inflammatory cytokines by DCs and T cells co-cultured with the stimulated DCs. In vivo evaluations demonstrated that nanovaccines were more potent than that based on the complete Freund's adjuvant, with respect to inducing anti-FSHR antibody, reducing the sperm count, inhibiting the sperm motility, and increasing the teratosperm rate. Immunization of male mice with nanovaccines notably decreased the parturition incidence of the mated females. Consequently, both in vitro and in vivo activities of FSHR could be considerably augmented by NPs. More importantly, our studies indicated that the pH-responsive nanovaccine was not superior over the non-responsive counterpart for the examined peptide antigen.
Collapse
Affiliation(s)
- Pingping Xu
- Department of Obstetrics and Gynaecology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China; Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing 400038, China; Department of Obstetrics and Gynaecology, Hospital 81 of PLA, Nanjing 210002, China
| | - Shuai Tang
- Department of Obstetrics and Gynaecology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Luping Jiang
- Department of Obstetrics and Gynaecology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Lihua Yang
- Department of Obstetrics and Gynaecology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China; Department of Obstetrics and Gynaecology, Tangshan Workers' Hospital of Hebei Province, Tangshan 063000, China
| | - Dinglin Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Shibin Feng
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Tingting Zhao
- Institute of Immunology, Third Military Medical University, Chongqing 400038, China
| | - Yajun Dong
- Department of Obstetrics and Gynaecology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Wei He
- Department of Obstetrics and Gynaecology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Ruibing Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, China
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing 400038, China.
| | - Zhiqing Liang
- Department of Obstetrics and Gynaecology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
45
|
Bhargava A, Bunkar N, Khare NK, Mishra D, Mishra PK. Nanoengineered strategies to optimize dendritic cells for gastrointestinal tumor immunotherapy: from biology to translational medicine. Nanomedicine (Lond) 2015; 9:2187-202. [PMID: 25405796 DOI: 10.2217/nnm.14.115] [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] [Indexed: 02/07/2023] Open
Abstract
Nanomedicine may play an important role in improving the clinical efficacy of dendritic cell-based immunotherapy against GI tract malignancies. Dendritic cell-based vaccines have proven their effectiveness against different established GI tract tumors, yet their success is mainly hindered by the strong tumor-induced suppressive microenvironment. The sustained and targeted release of tumor antigens to dendritic cells using different nanoengineered approaches would be an efficient strategy to overcome established immune tolerance. Encapsulation would result in low diffusivity, restricted movement, effective crosspresentation and enhanced T-cell responses. These nanotherapy-based approaches will certainly help with the designing of clinically translatable dendritic cell-based therapeutic vaccines and facilitate the selective removal of residual disease in gastrointestinal cancer patients following standard treatments.
Collapse
Affiliation(s)
- Arpit Bhargava
- Translational Research Laboratory, School of Biological Sciences, Dr H. S. Gour Central University, Sagar, India
| | | | | | | | | |
Collapse
|
46
|
Fernández TD, Pearson JR, Leal MP, Torres MJ, Blanca M, Mayorga C, Le Guével X. Intracellular accumulation and immunological properties of fluorescent gold nanoclusters in human dendritic cells. Biomaterials 2015; 43:1-12. [DOI: 10.1016/j.biomaterials.2014.11.045] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 11/12/2014] [Accepted: 11/24/2014] [Indexed: 12/23/2022]
|
47
|
Yang E, Qian W, Cao Z, Wang L, Bozeman EN, Ward C, Yang B, Selvaraj P, Lipowska M, Wang YA, Mao H, Yang L. Theranostic nanoparticles carrying doxorubicin attenuate targeting ligand specific antibody responses following systemic delivery. Am J Cancer Res 2015; 5:43-61. [PMID: 25553097 PMCID: PMC4265747 DOI: 10.7150/thno.10350] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Accepted: 09/18/2014] [Indexed: 12/23/2022] Open
Abstract
Understanding the effects of immune responses on targeted delivery of nanoparticles is important for clinical translations of new cancer imaging and therapeutic nanoparticles. In this study, we found that repeated administrations of magnetic iron oxide nanoparticles (IONPs) conjugated with mouse or human derived targeting ligands induced high levels of ligand specific antibody responses in normal and tumor bearing mice while injections of unconjugated mouse ligands were weakly immunogenic and induced a very low level of antibody response in mice. Mice that received intravenous injections of targeted and polyethylene glycol (PEG)-coated IONPs further increased the ligand specific antibody production due to differential uptake of PEG-coated nanoparticles by macrophages and dendritic cells. However, the production of ligand specific antibodies was markedly inhibited following systemic delivery of theranostic nanoparticles carrying a chemotherapy drug, doxorubicin. Targeted imaging and histological analysis revealed that lack of the ligand specific antibodies led to an increase in intratumoral delivery of targeted nanoparticles. Results of this study support the potential of further development of targeted theranostic nanoparticles for the treatment of human cancers.
Collapse
|
48
|
Zhu M, Wang R, Nie G. Applications of nanomaterials as vaccine adjuvants. Hum Vaccin Immunother 2014; 10:2761-74. [PMID: 25483497 PMCID: PMC4977448 DOI: 10.4161/hv.29589] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 05/26/2014] [Accepted: 06/15/2014] [Indexed: 02/07/2023] Open
Abstract
Vaccine adjuvants are applied to amplify the recipient's specific immune responses against pathogen infection or malignancy. A new generation of adjuvants is being developed to meet the demands for more potent antigen-specific responses, specific types of immune responses, and a high margin of safety. Nanotechnology provides a multifunctional stage for the integration of desired adjuvant activities performed by the building blocks of tailor-designed nanoparticles. Using nanomaterials for antigen delivery can provide high bioavailability, sustained and controlled release profiles, and targeting and imaging properties resulting from manipulation of the nanomaterials' physicochemical properties. Moreover, the inherent immune-regulating activity of particular nanomaterials can further promote and shape the cellular and humoral immune responses toward desired types. The combination of both the delivery function and immunomodulatory effect of nanomaterials as adjuvants is thought to largely benefit the immune outcomes of vaccination. In this review, we will address the current achievements of nanotechnology in the development of novel adjuvants. The potential mechanisms by which nanomaterials impact the immune responses to a vaccine and how physicochemical properties, including size, surface charge and surface modification, impact their resulting immunological outcomes will be discussed. This review aims to provide concentrated information to promote new insights for the development of novel vaccine adjuvants.
Collapse
Affiliation(s)
- Motao Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety; National Center for Nanoscience and Technology of China; Beijing, PR China
- Center for Inflammation and Epigenetics; Houston Methodist Research Institute; Houston, TX USA
| | - Rongfu Wang
- Center for Inflammation and Epigenetics; Houston Methodist Research Institute; Houston, TX USA
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety; National Center for Nanoscience and Technology of China; Beijing, PR China
| |
Collapse
|
49
|
Cruz LJ, Tacken PJ, Zeelenberg IS, Srinivas M, Bonetto F, Weigelin B, Eich C, de Vries IJ, Figdor CG. Tracking targeted bimodal nanovaccines: immune responses and routing in cells, tissue, and whole organism. Mol Pharm 2014; 11:4299-313. [PMID: 25290882 DOI: 10.1021/mp400717r] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Dendritic cells (DCs) are the most potent antigen-presenting cells (APCs), involved in the induction of immunity and currently exploited for antitumor immunotherapies. An optimized noninvasive imaging modality capable of determining and quantifying DC-targeted nanoparticle (NP) trajectories could provide valuable information regarding therapeutic vaccine outcome. Here, targeted poly(d,l-lactide-co-glycolide) nanoparticles (PLGA NPs) recognizing DC receptors were equipped with superparamagnetic iron oxide particles (SPIO) or gold nanoparticles with fluorescently labeled antigen. The fluorescent label allowed for rapid analysis and quantification of DC-specific uptake of targeted PLGA NPs in comparison to uptake by other cells. Transmission electron microscopy (TEM) showed that a fraction of the encapsulated antigen reached the lysosomal compartment of DCs, where SPIO and gold were already partially released. However, part of the PLGA NPs localized within the cytoplasm, as confirmed by confocal microscopy. DCs targeted with NPs carrying SPIO or fluorescent antigen were detected within lymph nodes as early as 1 h after injection by magnetic resonance imaging (MRI). Despite the fact that targeting did not markedly affect PLGA NP biodistribution on organism and tissue level, it increased delivery of NPs to DCs residing in peripheral lymph nodes and resulted in enhanced T cell proliferation. In conclusion, two imaging agents within a single carrier allows tracking of targeted PLGA NPs at the subcellular, cellular, and organismal levels, thereby facilitating the rational design of in vivo targeted vaccination strategies.
Collapse
Affiliation(s)
- Luis J Cruz
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Furlan SN, Mandraju R, Brewer T, Roybal K, Troutman TD, Hu W, Palm NW, Unni A, Pasare C. Enhancement of anti-tumor CD8 immunity by IgG1-mediated targeting of Fc receptors. MAbs 2014; 6:108-18. [PMID: 24284965 DOI: 10.4161/mabs.27052] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Dendritic cells (DCs) function as professional antigen presenting cells and are critical for linking innate immune responses to the induction of adaptive immunity. Many current cancer DC vaccine strategies rely on differentiating DCs, feeding them tumor antigens ex vivo, and infusing them into patients. Importantly, this strategy relies on prior knowledge of suitable “tumor-specific” antigens to prime an effective anti-tumor response. DCs express a variety of receptors specific for the Fc region of immunoglobulins, and antigen uptake via Fc receptors is highly efficient and facilitates antigen presentation to T cells. Therefore, we hypothesized that expression of the mouse IgG1 Fc region on the surface of tumors would enhance tumor cell uptake by DCs and other myeloid cells and promote the induction of anti-tumor T cell responses. To test this, we engineered a murine lymphoma cell line expressing surface IgG1 Fc and discovered that such tumor cells were taken up rapidly by DCs, leading to enhanced cross-presentation of tumor-derived antigen to CD8+ T cells. IgG1-Fc tumors failed to grow in vivo and prophylactic vaccination of mice with IgG1-Fc tumors resulted in rejection of unmanipulated tumor cells. Furthermore, IgG1-Fc tumor cells were able to slow the growth of an unmanipulated primary tumor when used as a therapeutic tumor vaccine. Our data demonstrate that engagement of Fc receptors by tumors expressing the Fc region of IgG1 is a viable strategy to induce efficient and protective anti-tumor CD8+ T cell responses without prior knowledge of tumor-specific antigens.
Collapse
|