1
|
Zaccariotto GDC, Bistaffa MJ, Zapata AMM, Rodero C, Coelho F, Quitiba JV, Lima L, Sterman R, Cardoso VDO, Zucolotto V. Cancer Nanovaccines: Mechanisms, Design Principles, and Clinical Translation. ACS NANO 2025; 19:16204-16223. [PMID: 40202241 PMCID: PMC12060653 DOI: 10.1021/acsnano.4c15765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 04/01/2025] [Accepted: 04/01/2025] [Indexed: 04/10/2025]
Abstract
Cancer immunotherapy has transformed the landscape of oncological treatment by employing various strategies to teach the immune system to eliminate tumors. Among these, cancer nanovaccines are an emerging strategy that utilizes nanotechnology to enhance immune activation in response to tumor antigens. This review addresses the principles behind the different technologies in this field aimed at generating a robust and effective immune response. The diversity of strategies adopted for the design of nanovaccines is discussed, including the types of active agents, nanocarriers, their functionalizations, and the incorporation of adjuvants. Furthermore, strategies to optimize nanoparticle formulations to enhance the antigen presentation, target immune cells, and organs and promote strong and durable antitumor responses are explored. Finally, we analyze the current state of clinical application, highlighting ongoing clinical trials and the future potential of cancer nanovaccines. The insights presented in this review aim to guide future research and development efforts in the field, contributing to the advancement of more effective and targeted nanovaccines in the fight against cancer.
Collapse
Affiliation(s)
- Gabriel de Camargo Zaccariotto
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | - Maria Julia Bistaffa
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | - Angelica Maria Mazuera Zapata
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | - Camila Rodero
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | - Fernanda Coelho
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | - João Victor
Brandão Quitiba
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | - Lorena Lima
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | - Raquel Sterman
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | | | - Valtencir Zucolotto
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| |
Collapse
|
2
|
Im SH, Chung Y, Duskunovic N, Choi H, Park SH, Chung HJ. Oligonucleotide-Linked Lipid Nanoparticles as a Versatile mRNA Nanovaccine Platform. Adv Healthc Mater 2024; 13:e2401868. [PMID: 39363681 DOI: 10.1002/adhm.202401868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/07/2024] [Indexed: 10/05/2024]
Abstract
An effective delivery platform is crucial for the development of mRNA vaccines and therapeutics. Here, a versatile platform utilizing cholesterol-modified oligonucleotides (L-oligo) that bind to the mRNA within lipid nanoparticles (LNP), and enables the effective delivery of the mRNA into target cells is introduced. mRNA incorporated into LNPs via linkage with L-oligo, termed oligonucleotide-linked LNP (lnLNP), is superior in cellular uptake and transfection efficiency in target cells in vitro and in vivo, compared to the conventional LNP formulations. It is further applied lnLNP as an mRNA vaccine platform for SARS-CoV-2, demonstrating robust induction of neutralizing activity as well as polyfunctional SARS-CoV-2-specific T-cell response in vivo. The current strategy can be versatilely applied to different LNP platforms, for vaccine and therapeutic applications against various diseases, such as infections and cancers.
Collapse
Affiliation(s)
- San Hae Im
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Youseung Chung
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Nevena Duskunovic
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Heewon Choi
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Su-Hyung Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
- The Center for Epidemic Preparedness, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Hyun Jung Chung
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| |
Collapse
|
3
|
Bai Z, Wan D, Lan T, Hong W, Dong H, Wei Y, Wei X. Nanoplatform Based Intranasal Vaccines: Current Progress and Clinical Challenges. ACS NANO 2024; 18:24650-24681. [PMID: 39185745 PMCID: PMC11394369 DOI: 10.1021/acsnano.3c10797] [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: 11/01/2023] [Revised: 04/01/2024] [Accepted: 04/08/2024] [Indexed: 08/27/2024]
Abstract
Multiple vaccine platforms have been employed to develop the nasal SARS-CoV-2 vaccines in preclinical studies, and the dominating pipelines are viral vectored as protein-based vaccines. Among them, several viral vectored-based vaccines have entered clinical development. Nevertheless, some unsatisfactory results were reported in these clinical studies. In the face of such urgent situations, it is imperative to rapidly develop the next-generation intranasal COVID-19 vaccine utilizing other technologies. Nanobased intranasal vaccines have emerged as an approach against respiratory infectious diseases. Harnessing the power of nanotechnology, these vaccines offer a noninvasive yet potent defense against pathogens, including the threat of COVID-19. The improvements made in vaccine mucosal delivery technologies based on nanoparticles, such as lipid nanoparticles, polymeric nanoparticles, inorganic nanoparticles etc., not only provide stability and controlled release but also enhance mucosal adhesion, effectively overcoming the limitations of conventional vaccines. Hence, in this review, we overview the evaluation of intranasal vaccine and highlight the current barriers. Next, the modern delivery systems based on nanoplatforms are summarized. The challenges in clinical application of nanoplatform based intranasal vaccine are finally discussed.
Collapse
Affiliation(s)
| | | | | | - Weiqi Hong
- Laboratory of Aging Research
and Cancer Drug Target, State Key Laboratory of Biotherapy, National
Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Haohao Dong
- Laboratory of Aging Research
and Cancer Drug Target, State Key Laboratory of Biotherapy, National
Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Yuquan Wei
- Laboratory of Aging Research
and Cancer Drug Target, State Key Laboratory of Biotherapy, National
Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Xiawei Wei
- Laboratory of Aging Research
and Cancer Drug Target, State Key Laboratory of Biotherapy, National
Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| |
Collapse
|
4
|
Kim D, Javius-Jones K, Mamidi N, Hong S. Dendritic nanoparticles for immune modulation: a potential next-generation nanocarrier for cancer immunotherapy. NANOSCALE 2024; 16:10208-10220. [PMID: 38727407 DOI: 10.1039/d4nr00635f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
Immune activation, whether occurring from direct immune checkpoint blockade or indirectly as a result of chemotherapy, is an approach that has drastically impacted the way we treat cancer. Utilizing patients' own immune systems for anti-tumor efficacy has been translated to robust immunotherapies; however, clinically significant successes have been achieved in only a subset of patient populations. Dendrimers and dendritic polymers have recently emerged as a potential nanocarrier platform that significantly improves the therapeutic efficacy of current and next-generation cancer immunotherapies. In this paper, we highlight the recent progress in developing dendritic polymer-based therapeutics with immune-modulating properties. Specifically, dendrimers, dendrimer hybrids, and dendronized copolymers have demonstrated promising results and are currently in pre-clinical development. Despite their early stage of development, these nanocarriers hold immense potential to make profound impact on cancer immunotherapy and combination therapy. This overview provides insights into the potential impact of dendrimers and dendron-based polymers, offering a preview of their potential utilities for various aspects of cancer treatment.
Collapse
Affiliation(s)
- DaWon Kim
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, School of Pharmacy, Madison, WI, USA.
| | - Kaila Javius-Jones
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, School of Pharmacy, Madison, WI, USA.
| | - Narsimha Mamidi
- Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI, USA
| | - Seungpyo Hong
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, School of Pharmacy, Madison, WI, USA.
- Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI, USA
- Lachman Institute for Drug Development, University of Wisconsin-Madison, Madison, WI, USA
- Yonsei Frontier Lab, Yonsei University, Seoul, Korea
| |
Collapse
|
5
|
Wang Y, Suarez ER, Kastrunes G, de Campos NSP, Abbas R, Pivetta RS, Murugan N, Chalbatani GM, D'Andrea V, Marasco WA. Evolution of cell therapy for renal cell carcinoma. Mol Cancer 2024; 23:8. [PMID: 38195534 PMCID: PMC10775455 DOI: 10.1186/s12943-023-01911-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 12/05/2023] [Indexed: 01/11/2024] Open
Abstract
Treatment for renal cell carcinoma (RCC) has improved dramatically over the last decade, shifting from high-dose cytokine therapy in combination with surgical resection of tumors to targeted therapy, immunotherapy, and combination therapies. However, curative treatment, particularly for advanced-stage disease, remains rare. Cell therapy as a "living drug" has achieved hematological malignancy cures with a high response rate, and significant research efforts have been made to facilitate its translation to solid tumors. Herein, we overview the cellular therapies for RCC focusing on allogeneic hematopoietic stem cell transplantation, T cell receptor gene-modified T cells, chimeric antigen receptor (CAR) T cells, CAR natural killer (NK) cells, lymphokine-activated killer (LAK) cells, γδ T cells, and dendritic cell vaccination. We have also included perspectives for using other recent approaches, such as CAR macrophages, dendritic cell-cytokine induced killer cells and regulatory CAR-T cells to shed light on preclinical development of cell therapy and advancing cell therapy into clinic to achieve cures for RCC.
Collapse
Affiliation(s)
- Yufei Wang
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Harvard Medical School, Boston, MA, 02215, USA
| | - Eloah Rabello Suarez
- Center for Natural and Human Sciences, Federal University of ABC, Santo Andre, SP, 09210-580, Brazil
- Graduate Program in Medicine - Hematology and Oncology, Federal University of Sao Paulo, São Paulo, SP, 04023-062, Brazil
| | - Gabriella Kastrunes
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Najla Santos Pacheco de Campos
- Center for Natural and Human Sciences, Federal University of ABC, Santo Andre, SP, 09210-580, Brazil
- Graduate Program in Medicine - Hematology and Oncology, Federal University of Sao Paulo, São Paulo, SP, 04023-062, Brazil
| | - Rabia Abbas
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Renata Schmieder Pivetta
- Center for Natural and Human Sciences, Federal University of ABC, Santo Andre, SP, 09210-580, Brazil
- Graduate Program in Medicine - Hematology and Oncology, Federal University of Sao Paulo, São Paulo, SP, 04023-062, Brazil
| | - Nithyassree Murugan
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | | | - Vincent D'Andrea
- Department of Surgery, Brigham and Women's Hospital, Boston, MA, 02215, USA
| | - Wayne A Marasco
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA.
- Harvard Medical School, Boston, MA, 02215, USA.
| |
Collapse
|
6
|
Wang Z, Guo Y, Shen M, Wang Y, Shi X. Hyperbranched Polymer-Based Vaccines for Cancer Immunotherapy. Macromol Biosci 2023; 23:e2300188. [PMID: 37300444 DOI: 10.1002/mabi.202300188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 05/31/2023] [Indexed: 06/12/2023]
Abstract
Recently, several immunotherapeutic strategies are extensively studied and entered clinical investigation, suggesting their potential to lead a new generation of cancer therapy. Particularly, a cancer vaccine that combines tumor-associated antigens and immune adjuvants with a nanocarrier holds huge promise for inducing specific antitumor immune responses. Hyperbranched polymers, such as dendrimers and branched polyethylenimine (PEI) possessing abundant positively charged amine groups and inherent proton sponge effect are ideal carriers of antigens. Much effort is devoted to design dendrimer/branched PEI-based cancer vaccines. Herein, the recent advances in the design of dendrimer/branched PEI-based cancer vaccines for immunotherapy are reviewed. The future perspectives with regard to the development of dendrimer/branched PEI-based cancer vaccines are also briefly discussed.
Collapse
Affiliation(s)
- Zhiqiang Wang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Yunqi Guo
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Mingwu Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Yong Wang
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| |
Collapse
|
7
|
Danaeifar M, Negahdari B, Eslam HM, Zare H, Ghanaat M, Koushali SS, Malekshahi ZV. Polymeric nanoparticles for DNA vaccine-based cancer immunotherapy: a review. Biotechnol Lett 2023; 45:1053-1072. [PMID: 37335426 DOI: 10.1007/s10529-023-03383-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 03/28/2023] [Accepted: 04/11/2023] [Indexed: 06/21/2023]
Abstract
Cancer is one of the leading causes of death and mortality in the world. There is an essential need to develop new drugs or therapeutic approaches to manage treatment-resistant cancers. Cancer immunotherapy is a type of cancer treatment that uses the power of the body's immune system to prevent, control, and eliminate cancer. One of the materials used as a vaccine in immunotherapy is DNA. The application of polymeric nanoparticles as carriers for DNA vaccines could be an effective therapeutic approach to activate immune responses and increase antigen presentation efficiency. Various materials have been used as polymeric nanoparticles, including: chitosan, poly (lactic-co-glycolic acid), Polyethylenimine, dendrimers, polypeptides, and polyesters. Application of these polymer nanoparticles has several advantages, including increased vaccine delivery, enhanced antigen presentation, adjuvant effects, and more sustainable induction of the immune system. Besides many clinical trials and commercial products that were developed based on polymer nanoparticles, there is still a need for more comprehensive studies to increase the DNA vaccine efficiency in cancer immunotherapy using this type of carrier.
Collapse
Affiliation(s)
- Mohsen Danaeifar
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Negahdari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Houra Mobaleghol Eslam
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Zare
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Momeneh Ghanaat
- Department of Microbiology, Ayatollah Amoli Branch, Islamic Azad University, Amol, Iran
| | - Sekinehe Shokouhi Koushali
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Ziba Veisi Malekshahi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Crintea A, Motofelea AC, Șovrea AS, Constantin AM, Crivii CB, Carpa R, Duțu AG. Dendrimers: Advancements and Potential Applications in Cancer Diagnosis and Treatment-An Overview. Pharmaceutics 2023; 15:pharmaceutics15051406. [PMID: 37242648 DOI: 10.3390/pharmaceutics15051406] [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: 03/05/2023] [Revised: 04/17/2023] [Accepted: 04/29/2023] [Indexed: 05/28/2023] Open
Abstract
Cancer is a leading cause of death worldwide, and the main treatment methods for this condition are surgery, chemotherapy, and radiotherapy. These treatment methods are invasive and can cause severe adverse reactions among organisms, so nanomaterials are increasingly used as structures for anticancer therapies. Dendrimers are a type of nanomaterial with unique properties, and their production can be controlled to obtain compounds with the desired characteristics. These polymeric molecules are used in cancer diagnosis and treatment through the targeted distribution of some pharmacological substances. Dendrimers have the ability to fulfill several objectives in anticancer therapy simultaneously, such as targeting tumor cells so that healthy tissue is not affected, controlling the release of anticancer agents in the tumor microenvironment, and combining anticancer strategies based on the administration of anticancer molecules to potentiate their effect through photothermal therapy or photodynamic therapy. The purpose of this review is to summarize and highlight the possible uses of dendrimers regarding the diagnosis and treatment of oncological conditions.
Collapse
Affiliation(s)
- Andreea Crintea
- Department of Molecular Sciences, Faculty of Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Alexandru Cătălin Motofelea
- Department of Internal Medicine, Faculty of Medicine, Victor Babeș University of Medicine and Pharmacy, 300041 Timișoara, Romania
| | - Alina Simona Șovrea
- Department of Morphological Sciences, Faculty of Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400000 Cluj-Napoca, Romania
| | - Anne-Marie Constantin
- Department of Morphological Sciences, Faculty of Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400000 Cluj-Napoca, Romania
| | - Carmen-Bianca Crivii
- Department of Morphological Sciences, Faculty of Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400000 Cluj-Napoca, Romania
| | - Rahela Carpa
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Institute for Research-Development-Innovation in Applied Natural Sciences, Babeș-Bolyai University, 400084 Cluj-Napoca, Romania
| | - Alina Gabriela Duțu
- Department of Molecular Sciences, Faculty of Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| |
Collapse
|
9
|
Kisakova LA, Apartsin EK, Nizolenko LF, Karpenko LI. Dendrimer-Mediated Delivery of DNA and RNA Vaccines. Pharmaceutics 2023; 15:pharmaceutics15041106. [PMID: 37111593 PMCID: PMC10145063 DOI: 10.3390/pharmaceutics15041106] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/27/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
DNA and RNA vaccines (nucleic acid-based vaccines) are a promising platform for vaccine development. The first mRNA vaccines (Moderna and Pfizer/BioNTech) were approved in 2020, and a DNA vaccine (Zydus Cadila, India), in 2021. They display unique benefits in the current COVID-19 pandemic. Nucleic acid-based vaccines have a number of advantages, such as safety, efficacy, and low cost. They are potentially faster to develop, cheaper to produce, and easier to store and transport. A crucial step in the technology of DNA or RNA vaccines is choosing an efficient delivery method. Nucleic acid delivery using liposomes is the most popular approach today, but this method has certain disadvantages. Therefore, studies are actively underway to develop various alternative delivery methods, among which synthetic cationic polymers such as dendrimers are very attractive. Dendrimers are three-dimensional nanostructures with a high degree of molecular homogeneity, adjustable size, multivalence, high surface functionality, and high aqueous solubility. The biosafety of some dendrimers has been evaluated in several clinical trials presented in this review. Due to these important and attractive properties, dendrimers are already being used to deliver a number of drugs and are being explored as promising carriers for nucleic acid-based vaccines. This review summarizes the literature data on the development of dendrimer-based delivery systems for DNA and mRNA vaccines.
Collapse
Affiliation(s)
- Lyubov A. Kisakova
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Kol’tsovo, Russia
| | - Evgeny K. Apartsin
- CBMN, UMR 5248, CNRS, Bordeaux INP, University Bordeaux, F-33600 Pessac, France
| | - Lily F. Nizolenko
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Kol’tsovo, Russia
| | - Larisa I. Karpenko
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, 630559 Kol’tsovo, Russia
| |
Collapse
|
10
|
Ghafelehbashi R, Farshbafnadi M, Aghdam NS, Amiri S, Salehi M, Razi S. Nanoimmunoengineering strategies in cancer diagnosis and therapy. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2023; 25:78-90. [PMID: 36076122 DOI: 10.1007/s12094-022-02935-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 08/22/2022] [Indexed: 01/07/2023]
Abstract
Cancer immunotherapy strategies in combination with engineered nanosystems have yielded beneficial results in the treatment of cancer and their application is increasing day by day. The pivotal role of stimuli-responsive nanosystems and nanomedicine-based cancer immunotherapy, as a subsidiary discipline in the field of immunology, cannot be ignored. Today, rapid advances in nanomedicine are used as a platform for exploring new therapeutic applications and modern smart healthcare management strategies. The progress of nanomedicine in cancer treatment has confirmed the findings of immunotherapy in the medical research phase. This study concentrates on approaches connected to the efficacy of nanoimmunoengineering strategies for cancer immunotherapies and their applications. By assessing improved approaches, different aspects of the nanoimmunoengineering strategies for cancer therapies are discussed in this study.
Collapse
Affiliation(s)
- Robabehbeygom Ghafelehbashi
- Department of Materials and Textile Engineering, College of Engineering, Razi University, Kermanshah, Iran.,Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Melina Farshbafnadi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Shahin Amiri
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.,Student Research Committee, Pasteur Institute of Iran, Tehran, Iran
| | - Mitra Salehi
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Sepideh Razi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran. .,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran. .,School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
11
|
Fajardo De La Rosa A, Hernández-Meléndez O, Saniger-Blesa JM, Nicolás-Vázquez MI, Bárzana E. Theoretical–experimental prediction of the selectivity between polyamidoamine dendrimers and bioactive peptides derived from amaranth seeds. NEW J CHEM 2023; 47:12170-12181. [DOI: 10.1039/d3nj01344h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
The use of dendritic macromolecules in the selective separation of amaranth-derived bioactive peptides opens the door to new processes and applications in the use of biomolecules with important biological functions.
Collapse
Affiliation(s)
- Alejandro Fajardo De La Rosa
- Departamento de Ingeniería Química, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de Mexico, Mexico
| | - Oscar Hernández-Meléndez
- Departamento de Ingeniería Química, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de Mexico, Mexico
| | - J. Manuel Saniger-Blesa
- Departamento de Micro y Nanotecnología, Instituto de Ciencias Aplicadas y Tecnología, Universidad Nacional Autónoma de México, Ciudad de Mexico, Mexico
| | - Ma. Inés Nicolás-Vázquez
- Departamento de Ciencias Químicas, Campo 1, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Ciudad de Mexico, Mexico
| | - Eduardo Bárzana
- Departamento de Alimentos y Biotecnología, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de Mexico, Mexico
| |
Collapse
|
12
|
Sarangi MK, Padhi S, Rath G, Nanda SS, Yi DK. Success of nano-vaccines against COVID-19: a transformation in nanomedicine. Expert Rev Vaccines 2022; 21:1739-1761. [PMID: 36384360 DOI: 10.1080/14760584.2022.2148659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
INTRODUCTION The vaccines being used against COVID-19 are composed of either non-viral or viral nanoparticles (NPs). Nanotechnology-based vaccine technology was studied for its potentially transformative advancement of medicine. AREAS COVERED NPs protect the encapsulated mRNA in vaccines, thereby enhancing the stability of the ribonucleic acids and facilitating their intact delivery to their specific targets. Compared to liposomes, lipid nanoparticles (LNPs) are unique and, through their rigid morphology and better cellular penetrability, render enhanced cargo stability. To explore nanotechnology-mediated vaccine delivery and its potential in future pandemics, we assessed articles from various databases, such as PubMed, Embase, and Scopus, including editorial/research notes, expert opinions, and collections of data from several clinical research trials. In the current review, we focus on the nanoparticulate approach of the different SARS-CoV-2 vaccines and explore their success against the pandemic. EXPERT OPINION The mRNA-based vaccines, with their tremendous efficacy of ~95% (under phase III-IV clinical trials) and distinct nanocarriers (LNPs), represent a new medical front alongside DNA and siRNA-based vaccines.
Collapse
Affiliation(s)
- Manoj Kumar Sarangi
- Department of Pharmacy, School of Pharmaceutical Sciences, Sardar Bhagwan Singh University, Dehradun, India
| | - Sasmita Padhi
- Department of Pharmacy, School of Pharmaceutical Sciences, Sardar Bhagwan Singh University, Dehradun, India
| | - Gautam Rath
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan University, Bhubaneswar, India
| | | | - Dong Kee Yi
- Department of Chemistry, Myongji University, Yongin, South Korea
| |
Collapse
|
13
|
Jeng LB, Liao LY, Shih FY, Teng CF. Dendritic-Cell-Vaccine-Based Immunotherapy for Hepatocellular Carcinoma: Clinical Trials and Recent Preclinical Studies. Cancers (Basel) 2022; 14:cancers14184380. [PMID: 36139542 PMCID: PMC9497058 DOI: 10.3390/cancers14184380] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/06/2022] [Accepted: 09/06/2022] [Indexed: 11/16/2022] Open
Abstract
Although many surgical and nonsurgical therapeutic options have been well-established, hepatocellular carcinoma (HCC) remains the third most common cause of cancer-related death worldwide. Therefore, the discovery of novel potential therapeutic strategies is still urgently required for improving survival and prognosis of HCC patients. As the most potent antigen-presenting cells in the human immune system, dendritic cells (DCs) play an important role in activating not only innate but also adaptive immune responses to specifically destroy tumor cells. As a result, DC-based vaccines, which are prepared by different tumor-antigen-pulsing strategies or maturation-stimulating reagents, either alone or in combination with various anticancer therapies and/or immune effector cells, have been developed as a promising personalized cancer immunotherapy. This review provides a comprehensive summary of the evidence from clinical trials evaluating the safety, feasibility, and efficacy of DC-based vaccines in treating HCC patients and highlights the data from recent preclinical studies regarding the development of promising strategies for optimizing the efficacy of DC-vaccine-based immunotherapy for HCC.
Collapse
Affiliation(s)
- Long-Bin Jeng
- Organ Transplantation Center, China Medical University Hospital, Taichung 404, Taiwan
- Cell Therapy Center, China Medical University Hospital, Taichung 404, Taiwan
| | - Li-Ying Liao
- Development of Plastic and Reconstructive Surgery, China Medical University Hospital, Taichung 404, Taiwan
| | - Fu-Ying Shih
- Ph.D. Program for Biotech Pharmaceutical Industry, School of Pharmacy, China Medical University, Taichung 404, Taiwan
| | - Chiao-Fang Teng
- Organ Transplantation Center, China Medical University Hospital, Taichung 404, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404, Taiwan
- Program for Cancer Biology and Drug Development, China Medical University, Taichung 404, Taiwan
- Research Center for Cancer Biology, China Medical University, Taichung 404, Taiwan
- Correspondence: ; Tel.: +886-4-2205-2121
| |
Collapse
|
14
|
Kumar M, Dogra R, Mandal UK. Nanomaterial-based delivery of vaccine through nasal route: Opportunities, challenges, advantages, and limitations. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103533] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
15
|
Rawding PA, Bu J, Wang J, Kim D, Drelich AJ, Kim Y, Hong S. Dendrimers for cancer immunotherapy: Avidity-based drug delivery vehicles for effective anti-tumor immune response. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1752. [PMID: 34414690 PMCID: PMC9485970 DOI: 10.1002/wnan.1752] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/25/2021] [Accepted: 07/29/2021] [Indexed: 12/19/2022]
Abstract
Cancer immunotherapy, or the utilization of a patient's own immune system to treat cancer, has shifted the paradigm of cancer treatment. Despite meaningful responses being observed in multiple studies, currently available immunotherapy platforms have only proven effective to a small subset of patients. To address this, nanoparticles have been utilized as a novel carrier for immunotherapeutic drugs, achieving robust anti-tumor effects with increased adaptive and durable responses. Specifically, dendrimer nanoparticles have attracted a great deal of scientific interest due to their versatility in various therapeutic applications, resulting from their unique physicochemical properties and chemically well-defined architecture. This review offers a comprehensive overview of dendrimer-based immunotherapy technologies, including their formulations, biological functionalities, and therapeutic applications. Common formulations include: (1) modulators of cytokine secretion of immune cells (adjuvants); (2) facilitators of the recognition of tumorous antigens (vaccines); (3) stimulators of immune effectors to selectively attack cells expressing specific antigens (antibodies); and (4) inhibitors of immune-suppressive responses (immune checkpoint inhibitors). On-going works and prospects of dendrimer-based immunotherapies are also discussed. Overall, this review provides a critical overview on rapidly growing dendrimer-based immunotherapy technologies and serves as a guideline for researchers and clinicians who are interested in this field. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Piper A Rawding
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA,Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jiyoon Bu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA,Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jianxin Wang
- Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - DaWon Kim
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA,Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Adam J Drelich
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA,Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Youngsoo Kim
- Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Seungpyo Hong
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA,Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI 53705, USA,Yonsei Frontier Lab and Department of Pharmacy, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
16
|
Lamrayah M, Phelip C, Coiffier C, Lacroix C, Willemin T, Trimaille T, Verrier B. A Polylactide-Based Micellar Adjuvant Improves the Intensity and Quality of Immune Response. Pharmaceutics 2022; 14:pharmaceutics14010107. [PMID: 35057003 PMCID: PMC8778782 DOI: 10.3390/pharmaceutics14010107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/24/2021] [Accepted: 12/27/2021] [Indexed: 11/21/2022] Open
Abstract
Micelles from amphiphilic polylactide-block-poly(N-acryloxysuccinimide-co-N-vinylpyrrolidone) (PLA-b-P(NAS-co-NVP)) block copolymers of 105 nm in size were characterized and evaluated in a vaccine context. The micelles were non-toxic in vitro (both in dendritic cells and HeLa cells). In vitro fluorescence experiments combined with in vivo fluorescence tomography imaging, through micelle loading with the DiR near infrared probe, suggested an efficient uptake of the micelles by the immune cells. The antigenic protein p24 of the HIV-1 was successfully coupled on the micelles using the reactive N-succinimidyl ester groups on the micelle corona, as shown by SDS-PAGE analyses. The antigenicity of the coupled antigen was preserved and even improved, as assessed by the immuno-enzymatic (ELISA) test. Then, the performances of the micelles in immunization were investigated and compared to different p24-coated PLA nanoparticles, as well as Alum and MF59 gold standards, following a standardized HIV-1 immunization protocol in mice. The humoral response intensity (IgG titers) was substantially similar between the PLA micelles and all other adjuvants over an extended time range (one year). More interestingly, this immune response induced by PLA micelles was qualitatively higher than the gold standards and PLA nanoparticles analogs, expressed through an increasing avidity index over time (>60% at day 365). Taken together, these results demonstrate the potential of such small-sized micellar systems for vaccine delivery.
Collapse
Affiliation(s)
- Myriam Lamrayah
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), Institut de Biologie et Chimie des Protéines (IBCP), CNRS UMR 5305, Université Lyon 1, Université de Lyon, 69367 Lyon, France; (C.P.); (C.C.); (C.L.); (T.W.); (B.V.)
- Correspondence: (M.L.); (T.T.)
| | - Capucine Phelip
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), Institut de Biologie et Chimie des Protéines (IBCP), CNRS UMR 5305, Université Lyon 1, Université de Lyon, 69367 Lyon, France; (C.P.); (C.C.); (C.L.); (T.W.); (B.V.)
| | - Céline Coiffier
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), Institut de Biologie et Chimie des Protéines (IBCP), CNRS UMR 5305, Université Lyon 1, Université de Lyon, 69367 Lyon, France; (C.P.); (C.C.); (C.L.); (T.W.); (B.V.)
| | - Céline Lacroix
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), Institut de Biologie et Chimie des Protéines (IBCP), CNRS UMR 5305, Université Lyon 1, Université de Lyon, 69367 Lyon, France; (C.P.); (C.C.); (C.L.); (T.W.); (B.V.)
| | - Thibaut Willemin
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), Institut de Biologie et Chimie des Protéines (IBCP), CNRS UMR 5305, Université Lyon 1, Université de Lyon, 69367 Lyon, France; (C.P.); (C.C.); (C.L.); (T.W.); (B.V.)
| | - Thomas Trimaille
- Laboratoire Ingénierie des Matériaux Polymères (IMP), CNRS UMR 5223, Université Lyon 1, Université de Lyon, 69622 Villeurbanne, France
- Correspondence: (M.L.); (T.T.)
| | - Bernard Verrier
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), Institut de Biologie et Chimie des Protéines (IBCP), CNRS UMR 5305, Université Lyon 1, Université de Lyon, 69367 Lyon, France; (C.P.); (C.C.); (C.L.); (T.W.); (B.V.)
| |
Collapse
|
17
|
Peptide-Functionalized Dendrimer Nanocarriers for Targeted Microdystrophin Gene Delivery. Pharmaceutics 2021; 13:pharmaceutics13122159. [PMID: 34959441 PMCID: PMC8708248 DOI: 10.3390/pharmaceutics13122159] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/07/2021] [Accepted: 12/12/2021] [Indexed: 12/13/2022] Open
Abstract
Gene therapy is a good alternative for determined congenital disorders; however, there are numerous limitations for gene delivery in vivo including targeted cellular uptake, intracellular trafficking, and transport through the nuclear membrane. Here, a modified G5 polyamidoamine (G5 PAMAM) dendrimer-DNA complex was developed, which will allow cell-specific targeting to skeletal muscle cells and transport the DNA through the intracellular machinery and the nuclear membrane. The G5 PAMAM nanocarrier was modified with a skeletal muscle-targeting peptide (SMTP), a DLC8-binding peptide (DBP) for intracellular transport, and a nuclear localization signaling peptide (NLS) for nuclear uptake, and polyplexed with plasmid DNA containing the GFP-tagged microdystrophin (µDys) gene. The delivery of µDys has been considered as a therapeutic modality for patients suffering from a debilitating Duchenne muscular dystrophy (DMD) disorder. The nanocarrier-peptide-DNA polyplexes were prepared with different charge ratios and characterized for stability, size, surface charge, and cytotoxicity. Using the optimized nanocarrier polyplexes, the transfection efficiency in vitro was determined by demonstrating the expression of the GFP and the µDys protein using fluorescence and Western blotting studies, respectively. Protein expression in vivo was determined by injecting an optimal nanocarrier polyplex formulation to Duchenne model mice, mdx4Cv. Ultimately, these nanocarrier polyplexes will allow targeted delivery of the microdystrophin gene to skeletal muscle cells and result in improved muscle function in Duchenne muscular dystrophy patients.
Collapse
|
18
|
Kharwade R, Badole P, Mahajan N, More S. Toxicity And Surface Modification Of Dendrimers: A Critical Review. Curr Drug Deliv 2021; 19:451-465. [PMID: 34674620 DOI: 10.2174/1567201818666211021160441] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 08/21/2021] [Accepted: 10/01/2021] [Indexed: 11/22/2022]
Abstract
As compared to other nano polymers, dendrimers have novel three dimensional, synthetic hyperbranched, nano-polymeric structures. The characteristic of these supramolecular dendritic structures has a high degree of significant surface as well as core functionality in the transportation of drugs for targeted therapy, specifically in host-guest response, gene transfer therapy and imaging of biological systems. However, there are conflicting shreds of evidence regarding biological safety and dendrimers toxicity due to their positive charge at the surface. It includes cytotoxicity, hemolytic toxicity, haematological toxicity, immunogenicity and in vivo toxicity. Therefore to resolve these problems surface modification of the dendrimer group is one of the methods. From that point, this review involves different strategies which reduce the toxicity and improve the biocompatibility of different types of dendrimers. From that viewpoint, we broaden the structural and safe characteristics of the dendrimers in the biomedical and pharmaceutical fields.
Collapse
Affiliation(s)
- Rohini Kharwade
- Dadasaheb Balpande College of Pharmacy, Besa, Nagpur, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur, (MS). India
| | - Payal Badole
- Dadasaheb Balpande College of Pharmacy, Besa, Nagpur, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur, (MS). India
| | - Nilesh Mahajan
- Dadasaheb Balpande College of Pharmacy, Besa, Nagpur, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur, (MS). India
| | - Sachin More
- Dadasaheb Balpande College of Pharmacy, Besa, Nagpur, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur, (MS). India
| |
Collapse
|
19
|
Sa-nguanmoo N, Namdee K, Khongkow M, Ruktanonchai U, Zhao Y, Liang XJ. Review: Development of SARS-CoV-2 immuno-enhanced COVID-19 vaccines with nano-platform. NANO RESEARCH 2021; 15:2196-2225. [PMID: 34659650 PMCID: PMC8501370 DOI: 10.1007/s12274-021-3832-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/19/2021] [Accepted: 08/19/2021] [Indexed: 05/04/2023]
Abstract
Vaccination is the most effective way to prevent coronavirus disease 2019 (COVID-19). Vaccine development approaches consist of viral vector vaccines, DNA vaccine, RNA vaccine, live attenuated virus, and recombinant proteins, which elicit a specific immune response. The use of nanoparticles displaying antigen is one of the alternative approaches to conventional vaccines. This is due to the fact that nano-based vaccines are stable, able to target, form images, and offer an opportunity to enhance the immune responses. The diameters of ultrafine nanoparticles are in the range of 1-100 nm. The application of nanotechnology on vaccine design provides precise fabrication of nanomaterials with desirable properties and ability to eliminate undesirable features. To be successful, nanomaterials must be uptaken into the cell, especially into the target and able to modulate cellular functions at the subcellular levels. The advantages of nano-based vaccines are the ability to protect a cargo such as RNA, DNA, protein, or synthesis substance and have enhanced stability in a broad range of pH, ambient temperatures, and humidity for long-term storage. Moreover, nano-based vaccines can be engineered to overcome biological barriers such as nonspecific distribution in order to elicit functions in antigen presenting cells. In this review, we will summarize on the developing COVID-19 vaccine strategies and how the nanotechnology can enhance antigen presentation and strong immunogenicity using advanced technology in nanocarrier to deliver antigens. The discussion about their safe, effective, and affordable vaccines to immunize against COVID-19 will be highlighted.
Collapse
Affiliation(s)
- Nawamin Sa-nguanmoo
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Beijing, 100190 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Katawut Namdee
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani, 12120 Thailand
| | - Mattaka Khongkow
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani, 12120 Thailand
| | - Uracha Ruktanonchai
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani, 12120 Thailand
| | - YongXiang Zhao
- National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumour Theranostics and Therapy, Guangxi Medical University, Nanning, 530021 China
| | - Xing-Jie Liang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Beijing, 100190 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| |
Collapse
|
20
|
Rezaei T, Davoudian E, Khalili S, Amini M, Hejazi M, de la Guardia M, Mokhtarzadeh A. Strategies in DNA vaccine for melanoma cancer. Pigment Cell Melanoma Res 2021; 34:869-891. [PMID: 33089665 DOI: 10.1111/pcmr.12933] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 09/05/2020] [Accepted: 09/22/2020] [Indexed: 11/29/2022]
Abstract
According to reports of the international agency for cancer on research, although malignant melanoma shows less prevalence than nonmelanoma skin cancers, it is the major cause of skin cancer mortality. Given that, the production of effective vaccines to control melanoma is eminently required. In this regard, DNA-based vaccines have been extensively investigated for melanoma therapy. DNA vaccines are capable of inducing both cellular and humoral branches of immune responses. These vaccines possess some valuable advantages such as lack of severe side effects and high stability compared to conventional vaccination methods. The ongoing studies are focused on novel strategies in the development of DNA vaccines encoding artificial polyepitope immunogens based on the multiple melanoma antigens, the inclusion of molecular adjuvants to increase the level of immune responses, and the improvement of delivery approaches. In this review, we have outlined the recent advances in the field of melanoma DNA vaccines and described their implications in clinical trials as a strong strategy in the prevention and control of melanoma.
Collapse
Affiliation(s)
- Tayebeh Rezaei
- Department of Molecular Medicine and Biotechnology, Faculty of Medicine, Arak University of Medical Science, Arak, Iran
| | - Elham Davoudian
- Department of Microbiology, School of Paramedical Sciences, Ilam University of Medical Sciences, Ilam, Iran
| | - Saeed Khalili
- Department of Biology Sciences, Shahid Rajaee Teacher Training University, Tehran, Iran
| | - Mohammad Amini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Hejazi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
21
|
Eusébio D, Neves AR, Costa D, Biswas S, Alves G, Cui Z, Sousa Â. Methods to improve the immunogenicity of plasmid DNA vaccines. Drug Discov Today 2021; 26:2575-2592. [PMID: 34214667 DOI: 10.1016/j.drudis.2021.06.008] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/31/2021] [Accepted: 06/22/2021] [Indexed: 02/06/2023]
Abstract
DNA vaccines have emerged as innovative approaches that have great potential to overcome the limitations of current conventional vaccines. Plasmid DNA vaccines are often safer than other vaccines because they carry only antigen genetic information, are more stable and easier to produce, and can stimulate both humoral and cellular immune responses. Although the results of ongoing clinical trials are very promising, some limitations compromise the immunogenicity of these vaccines. Thus, this review describes different strategies that can be explored to improve the immunogenicity of plasmid DNA vaccines, including the optimization of the plasmid vector backbone, the use of different methods for vaccine delivery, the use of alternative administration routes and the inclusion of adjuvants. In combination, these improvements could lead to the successful clinical use of plasmid DNA vaccines.
Collapse
Affiliation(s)
- Dalinda Eusébio
- CICS-UBI - Health Science Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Ana R Neves
- CICS-UBI - Health Science Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Diana Costa
- CICS-UBI - Health Science Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Swati Biswas
- Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Shameerpet, Hyderabad 500078, Telangana, India
| | - Gilberto Alves
- CICS-UBI - Health Science Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Zhengrong Cui
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX 78712, USA
| | - Ângela Sousa
- CICS-UBI - Health Science Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal.
| |
Collapse
|
22
|
Preclinical models and technologies to advance nanovaccine development. Adv Drug Deliv Rev 2021; 172:148-182. [PMID: 33711401 DOI: 10.1016/j.addr.2021.03.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 12/13/2022]
Abstract
The remarkable success of targeted immunotherapies is revolutionizing cancer treatment. However, tumor heterogeneity and low immunogenicity, in addition to several tumor-associated immunosuppression mechanisms are among the major factors that have precluded the success of cancer vaccines as targeted cancer immunotherapies. The exciting outcomes obtained in patients upon the injection of tumor-specific antigens and adjuvants intratumorally, reinvigorated interest in the use of nanotechnology to foster the delivery of vaccines to address cancer unmet needs. Thus, bridging nano-based vaccine platform development and predicted clinical outcomes the selection of the proper preclinical model will be fundamental. Preclinical models have revealed promising outcomes for cancer vaccines. However, only few cases were associated with clinical responses. This review addresses the major challenges related to the translation of cancer nano-based vaccines to the clinic, discussing the requirements for ex vivo and in vivo models of cancer to ensure the translation of preclinical success to patients.
Collapse
|
23
|
Karpenko LI, Apartsin EK, Dudko SG, Starostina EV, Kaplina ON, Antonets DV, Volosnikova EA, Zaitsev BN, Bakulina AY, Venyaminova AG, Ilyichev AA, Bazhan SI. Cationic Polymers for the Delivery of the Ebola DNA Vaccine Encoding Artificial T-Cell Immunogen. Vaccines (Basel) 2020; 8:vaccines8040718. [PMID: 33271964 PMCID: PMC7760684 DOI: 10.3390/vaccines8040718] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/23/2020] [Accepted: 11/27/2020] [Indexed: 11/16/2022] Open
Abstract
Background: According to current data, an effective Ebola virus vaccine should induce both humoral and T-cell immunity. In this work, we focused our efforts on methods for delivering artificial T-cell immunogen in the form of a DNA vaccine, using generation 4 polyamidoamine dendrimers (PAMAM G4) and a polyglucin:spermidine conjugate (PG). Methods: Optimal conditions were selected for obtaining complexes of previously developed DNA vaccines with cationic polymers. The sizes, mobility and surface charge of the complexes with PG and PAMAM 4G have been determined. The immunogenicity of the obtained vaccine constructs was investigated in BALB/c mice. Results: It was shown that packaging of DNA vaccine constructs both in the PG envelope and the PAMAM 4G envelope results in an increase in their immunogenicity as compared with the group of mice immunized with the of vector plasmid pcDNA3.1 (a negative control). The highest T-cell responses were shown in mice immunized with complexes of DNA vaccines with PG and these responses significantly exceeded those in the groups of animals immunized with both the combination of naked DNAs and the combination DNAs coated with PAMAM 4G. In the group of animals immunized with complexes of the DNA vaccines with PAMAM 4G, no statistical differences were found in the ability to induce T-cell responses, as compared with the group of mice immunized with the combination of naked DNAs. Conclusions: The PG conjugate can be considered as a promising and safe means to deliver DNA-based vaccines. The use of PAMAM requires further optimization.
Collapse
Affiliation(s)
- Larisa I. Karpenko
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, 630559 Novosibirsk Region, Russia; (S.G.D.); (E.V.S.); (O.N.K.); (D.V.A.); (E.A.V.); (B.N.Z.); (A.Y.B.); (A.A.I.)
- Correspondence: (L.I.K.); (S.I.B.); Tel.: +7-383-363-47-00 (ext. 2001) (L.I.K. & S.I.B.)
| | - Evgeny K. Apartsin
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.K.A.); (A.G.V.)
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
- Laboratoire de Chimie de Coordination, CNRS, 31077 Toulouse, France
| | - Sergei G. Dudko
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, 630559 Novosibirsk Region, Russia; (S.G.D.); (E.V.S.); (O.N.K.); (D.V.A.); (E.A.V.); (B.N.Z.); (A.Y.B.); (A.A.I.)
| | - Ekaterina V. Starostina
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, 630559 Novosibirsk Region, Russia; (S.G.D.); (E.V.S.); (O.N.K.); (D.V.A.); (E.A.V.); (B.N.Z.); (A.Y.B.); (A.A.I.)
| | - Olga N. Kaplina
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, 630559 Novosibirsk Region, Russia; (S.G.D.); (E.V.S.); (O.N.K.); (D.V.A.); (E.A.V.); (B.N.Z.); (A.Y.B.); (A.A.I.)
| | - Denis V. Antonets
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, 630559 Novosibirsk Region, Russia; (S.G.D.); (E.V.S.); (O.N.K.); (D.V.A.); (E.A.V.); (B.N.Z.); (A.Y.B.); (A.A.I.)
| | - Ekaterina A. Volosnikova
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, 630559 Novosibirsk Region, Russia; (S.G.D.); (E.V.S.); (O.N.K.); (D.V.A.); (E.A.V.); (B.N.Z.); (A.Y.B.); (A.A.I.)
| | - Boris N. Zaitsev
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, 630559 Novosibirsk Region, Russia; (S.G.D.); (E.V.S.); (O.N.K.); (D.V.A.); (E.A.V.); (B.N.Z.); (A.Y.B.); (A.A.I.)
| | - Anastasiya Yu. Bakulina
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, 630559 Novosibirsk Region, Russia; (S.G.D.); (E.V.S.); (O.N.K.); (D.V.A.); (E.A.V.); (B.N.Z.); (A.Y.B.); (A.A.I.)
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Aliya G. Venyaminova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.K.A.); (A.G.V.)
| | - Alexander A. Ilyichev
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, 630559 Novosibirsk Region, Russia; (S.G.D.); (E.V.S.); (O.N.K.); (D.V.A.); (E.A.V.); (B.N.Z.); (A.Y.B.); (A.A.I.)
| | - Sergei I. Bazhan
- State Research Center of Virology and Biotechnology “Vector”, Koltsovo, 630559 Novosibirsk Region, Russia; (S.G.D.); (E.V.S.); (O.N.K.); (D.V.A.); (E.A.V.); (B.N.Z.); (A.Y.B.); (A.A.I.)
- Correspondence: (L.I.K.); (S.I.B.); Tel.: +7-383-363-47-00 (ext. 2001) (L.I.K. & S.I.B.)
| |
Collapse
|
24
|
Lowell JA, Dikici E, Joshi PM, Landgraf R, Lemmon VP, Daunert S, Izenwasser S, Daftarian P. Vaccination against cocaine using a modifiable dendrimer nanoparticle platform. Vaccine 2020; 38:7989-7997. [PMID: 33158592 DOI: 10.1016/j.vaccine.2020.10.041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 09/29/2020] [Accepted: 10/12/2020] [Indexed: 10/23/2022]
Abstract
Pharmacological therapies for the treatment of cocaine addiction have had disappointing efficacy, and the lack of recent developments in the clinical care of cocaine-addicted patients indicates a need for novel treatment strategies. Recent studies have shown that vaccination against cocaine to elicit production of antibodies that reduce concentrations of free drug in the blood is a promising method to protect against the effects of cocaine and reduce rates of relapse. However, the poorly immunogenic nature of cocaine remains a major hurdle to active immunization. Therefore, we hypothesized that strategies to increase targeted exposure of cocaine to the immune system may produce a more effective vaccine. To specifically direct an immune response against cocaine, in the present study we have conjugated a cocaine analog to a dendrimer-based nanoparticle carrier with MHC II-binding moieties that previously has been shown to activate antigen-presenting cells necessary for antibody production. This strategy produced a rapid, prolonged, and high affinity anti-cocaine antibody response without the need for an adjuvant. Surprisingly, additional evaluation using multiple adjuvant formulations in two strains of inbred mice found adjuvants were either functionally redundant or deleterious in the vaccination against cocaine using this platform. The use of conditioned place preference in rats after administration of this vaccine provided proof of concept for the ability of this vaccine to diminish cocaine reward. Together these data demonstrate the intrinsic efficacy of an immune-targeting dendrimer-based cocaine vaccine, with a vast potential for design of future vaccines against other poorly immunogenic antigens by substitution of the conjugated cargo.
Collapse
Affiliation(s)
- Jeffrey A Lowell
- Miami Project to Cure Paralysis, University of Miami, 1095 NW 14th Terrace, Miami, FL 33136, United States
| | - Emre Dikici
- Department of Biochemistry and Molecular Biology, University of Miami, 1011 NW 15th Street, Miami, FL 33136, United States; Dr. JT Macdonald Foundation Biomedical Nanotechnology Institute, University of Miami, Life Science and Technology Park, 1951 Northwest 7th Avenue, Miami, FL 33136, United States
| | - Pratibha M Joshi
- Department of Biochemistry and Molecular Biology, University of Miami, 1011 NW 15th Street, Miami, FL 33136, United States; Dr. JT Macdonald Foundation Biomedical Nanotechnology Institute, University of Miami, Life Science and Technology Park, 1951 Northwest 7th Avenue, Miami, FL 33136, United States
| | - Ralf Landgraf
- Department of Biochemistry and Molecular Biology, University of Miami, 1011 NW 15th Street, Miami, FL 33136, United States
| | - Vance P Lemmon
- Miami Project to Cure Paralysis, University of Miami, 1095 NW 14th Terrace, Miami, FL 33136, United States; Department of Neurological Surgery, University of Miami, 1095 NW 14th Terrace, Miami, FL 33136, United States
| | - Sylvia Daunert
- Department of Biochemistry and Molecular Biology, University of Miami, 1011 NW 15th Street, Miami, FL 33136, United States; Dr. JT Macdonald Foundation Biomedical Nanotechnology Institute, University of Miami, Life Science and Technology Park, 1951 Northwest 7th Avenue, Miami, FL 33136, United States; Miami Clinical and Translational Science Institute, University of Miami, Clinical Research Building, 1120 NW 14th St., Miami, FL 33136, United States
| | - Sari Izenwasser
- Department of Psychiatry and Behavioral Sciences, University of Miami, 1600 NW 10(th) Avenue, Miami, FL 33136, United States.
| | - Pirouz Daftarian
- Department of Biochemistry and Molecular Biology, University of Miami, 1011 NW 15th Street, Miami, FL 33136, United States; Dr. JT Macdonald Foundation Biomedical Nanotechnology Institute, University of Miami, Life Science and Technology Park, 1951 Northwest 7th Avenue, Miami, FL 33136, United States.
| |
Collapse
|
25
|
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: 125] [Impact Index Per Article: 25.0] [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
|
26
|
Nanotechnology in the arena of cancer immunotherapy. Arch Pharm Res 2020; 43:58-79. [DOI: 10.1007/s12272-020-01207-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 01/09/2020] [Indexed: 12/14/2022]
|
27
|
Han S, Huang K, Gu Z, Wu J. Tumor immune microenvironment modulation-based drug delivery strategies for cancer immunotherapy. NANOSCALE 2020; 12:413-436. [PMID: 31829394 DOI: 10.1039/c9nr08086d] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The past years have witnessed promising clinical feedback for anti-cancer immunotherapies, which have become one of the hot research topics; however, they are limited by poor delivery kinetics, narrow patient response profiles, and systemic side effects. To the best of our knowledge, the development of cancer is highly associated with the immune system, especially the tumor immune microenvironment (TIME). Based on the comprehensive understanding of the complexity and diversity of TIME, drug delivery strategies focused on the modulation of TIME can be of great significance for directing and improving cancer immunotherapy. This review highlights the TIME modulation in cancer immunotherapy and summarizes the versatile TIME modulation-based cancer immunotherapeutic strategies, medicative principles and accessory biotechniques for further clinical transformation. Remarkably, the recent advances of cancer immunotherapeutic drug delivery systems and future prospects of TIME modulation-based drug delivery systems for much more controlled and precise cancer immunotherapy will be emphatically discussed.
Collapse
Affiliation(s)
- Shuyan Han
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province, School of Biomedical Engineering, Sun Yat-sen University, Guangzhou 510006, PR China.
| | | | | | | |
Collapse
|
28
|
Nanomaterials for direct and indirect immunomodulation: A review of applications. Eur J Pharm Sci 2020; 142:105139. [DOI: 10.1016/j.ejps.2019.105139] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 10/14/2019] [Accepted: 11/03/2019] [Indexed: 01/03/2023]
|
29
|
Hao Y, Zhou X, Li R, Song Z, Min Y. Advances of functional nanomaterials for cancer immunotherapeutic applications. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 12:e1574. [PMID: 31566896 DOI: 10.1002/wnan.1574] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 06/04/2019] [Accepted: 06/19/2019] [Indexed: 12/11/2022]
Abstract
Immunotherapy has made great progress by modulating the body's own immune system to fight against cancer cells. However, the low response rates of related drugs limit the development of immunotherapy strategies. Fortunately, the advantages of nanotechnology can just make up for this shortcoming. Nanocarriers of diverse systems are utilized to co-deliver antigens and adjuvants, combined with drugs for immunomodulatory, such as chemotherapy, radiotherapy, and photodynamic. Here we review recent studies on immunotherapy with biomimetic, organic, and inorganic nanomaterials. They are going to potentially overcome the drawbacks in cancer immunotherapy with delivering immunomodulatory drugs, delivering cancer vaccine, and monitoring the immune systems. This article is characterized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Yuhao Hao
- CAS Key Lab of Soft Matter Chemistry, University of Science and Technology of China, Hefei, China.,Department of Chemistry, University of Science and Technology of China, Hefei, China
| | - Xingyu Zhou
- CAS Key Lab of Soft Matter Chemistry, University of Science and Technology of China, Hefei, China.,Department of Chemistry, University of Science and Technology of China, Hefei, China
| | - Rui Li
- CAS Key Lab of Soft Matter Chemistry, University of Science and Technology of China, Hefei, China.,Department of Chemistry, University of Science and Technology of China, Hefei, China
| | - Zechenxi Song
- CAS Key Lab of Soft Matter Chemistry, University of Science and Technology of China, Hefei, China.,Department of Chemistry, University of Science and Technology of China, Hefei, China
| | - Yuanzeng Min
- CAS Key Lab of Soft Matter Chemistry, University of Science and Technology of China, Hefei, China.,Department of Chemistry, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Science at the Microscale, University of Science and Technology of China, Hefei, China
| |
Collapse
|
30
|
Trimaille T, Lacroix C, Verrier B. Self-assembled amphiphilic copolymers as dual delivery system for immunotherapy. Eur J Pharm Biopharm 2019; 142:232-239. [PMID: 31229673 DOI: 10.1016/j.ejpb.2019.06.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 05/03/2019] [Accepted: 06/19/2019] [Indexed: 01/07/2023]
Abstract
Subunit vaccines using recombinant antigens appear as the privileged vaccination technology for safety reasons but still require the development of carriers/adjuvants ensuring optimal immunogenicity and efficacy. Micelles from self-assembled amphiphilic copolymers have recently emerged as highly relevant and promising candidates owing to their ease of preparation, low size (entering in lymphatic capillaries for reaching lymph nodes), size/surface tunability and chemical versatility enabling introduction of stimuli (e.g. pH) responsive features and biofunctionalization with dedicated molecules. In particular, research efforts have increasingly focused on dendritic cells (DCs) targeting and activation by co-delivering (with antigen) ligands of pattern recognition receptors (PRRs, e.g. toll-like receptors). Such strategy has appeared as one of the most effective for eliciting CD 8+ T-cell response, which is crucial in the eradication of tumors and numerous infectious diseases. In this short review, we highlight the recent advances in such micelle-based carriers in subunit vaccination and how their precise engineering can be a strong asset for guiding and controlling immune responses.
Collapse
Affiliation(s)
- Thomas Trimaille
- Aix Marseille Univ, CNRS, Institut de Chimie Radicalaire, Marseille, France.
| | - Céline Lacroix
- Université Lyon 1, CNRS, UMR 5305, Biologie Tissulaire et Ingénierie Thérapeutique, IBCP, 69367 Lyon, France
| | - Bernard Verrier
- Université Lyon 1, CNRS, UMR 5305, Biologie Tissulaire et Ingénierie Thérapeutique, IBCP, 69367 Lyon, France
| |
Collapse
|
31
|
Jativa SD, Thapar N, Broyles D, Dikici E, Daftarian P, Jiménez JJ, Daunert S, Deo SK. Enhanced Delivery of Plasmid DNA to Skeletal Muscle Cells using a DLC8-Binding Peptide and ASSLNIA-Modified PAMAM Dendrimer. Mol Pharm 2019; 16:2376-2384. [PMID: 30951315 DOI: 10.1021/acs.molpharmaceut.8b01313] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Skeletal muscle is ideally suited and highly desirable as a target for therapeutic gene delivery because of its abundance, high vascularization, and high levels of protein expression. However, efficient gene delivery to skeletal muscle remains a current challenge. Besides the major obstacle of cell-specific targeting, efficient intracellular trafficking, or the cytosolic transport of DNA to the nucleus, must be demonstrated. To overcome the challenge of cell-specific targeting, herein we develop a generation 5-polyamidoamine dendrimer (G5-PAMAM) functionalized with a skeletal muscle-targeted peptide, ASSLNIA (G5-SMTP). Specifically, to demonstrate the feasibility of our approach, we prepared a complex of our G5-SMTP dendrimer with a plasmid encoding firefly luciferase and investigated its delivery to skeletal muscle cells. Luciferase assays indicated a threefold increase in transfection efficiency of C2C12 murine skeletal muscle cells using G5-SMTP when compared with nontargeting nanocarriers using unmodified G5. To further improve the transfection yield, we employed a cationic dynein light chain 8 protein (DLC8)-binding peptide (DBP) containing an internal sequence known to bind to the DLC8 of the dynein motor protein complex. Complexation of DBP with our targeting nanocarrier, that is, G5-SMTP, and our luciferase plasmid cargo resulted in a functional nanocarrier that showed an additional sixfold increase in transfection efficiency compared with G5-SMTP transfection alone. To our knowledge, this is the first successful use of two different functional nanocarrier components that enable targeted skeletal muscle cell recognition and increased efficiency of intracellular trafficking to synergistically enhance gene delivery to skeletal muscle cells. This strategy of targeting and trafficking can also be universally applied to any cell/tissue type for which a recognition domain exists.
Collapse
Affiliation(s)
- Samuel D Jativa
- University of Miami Clinical and Translational Science Institute , Miami 33136 , United States
| | | | - David Broyles
- Dr. JT Macdonald Foundation Biomedical Nanotechnology Institute of the University of Miami , Miami 33136 , United States
| | - Emre Dikici
- Dr. JT Macdonald Foundation Biomedical Nanotechnology Institute of the University of Miami , Miami 33136 , United States
| | - Pirouz Daftarian
- JSR Micro, Life Sciences , 1280 North Matilda Avenue , Sunnyvale , California 94089 , United States
| | | | - Sylvia Daunert
- Dr. JT Macdonald Foundation Biomedical Nanotechnology Institute of the University of Miami , Miami 33136 , United States.,University of Miami Clinical and Translational Science Institute , Miami 33136 , United States
| | - Sapna K Deo
- Dr. JT Macdonald Foundation Biomedical Nanotechnology Institute of the University of Miami , Miami 33136 , United States.,University of Miami Clinical and Translational Science Institute , Miami 33136 , United States
| |
Collapse
|
32
|
Roy H, Nayak BS, Abdul Rahaman S. Nano Drugs. CHARACTERIZATION AND BIOLOGY OF NANOMATERIALS FOR DRUG DELIVERY 2019:445-475. [DOI: 10.1016/b978-0-12-814031-4.00016-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
33
|
Tomich JM, Wessel E, Choi J, Avila LA. Nonviral Gene Therapy: Peptiplexes. NUCLEIC ACID NANOTHERANOSTICS 2019:247-276. [DOI: 10.1016/b978-0-12-814470-1.00008-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
34
|
Gao S, Yang D, Fang Y, Lin X, Jin X, Wang Q, Wang X, Ke L, Shi K. Engineering Nanoparticles for Targeted Remodeling of the Tumor Microenvironment to Improve Cancer Immunotherapy. Theranostics 2019; 9:126-151. [PMID: 30662558 PMCID: PMC6332787 DOI: 10.7150/thno.29431] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 11/01/2018] [Indexed: 12/22/2022] Open
Abstract
Owing to the fast-paced growth and cross-infiltration of oncology, immunology and molecular biology, tumor immunotherapy technology represented by immune checkpoint blockade and chimeric antigen receptor (CAR) T cell therapy has lately made remarkable advancements. In comparison with traditional chemotherapy, immunotherapy has the potential to elicit a stronger sustained antitumor immune response in those patients who have advanced malignant malignancies. In spite of the advancements made, a significant number of clinical research works have validated that an extensive proportion of cancer patients still manifest insensitivity to immunotherapy, primarily because of the immunomodulatory interactions between tumor cells and the immunosuppressive tumor microenvironment (TME), together mediating the immune tolerance of tumors and accordingly impacting the positive response to immunotherapy. The intricate immunosuppressive networks formed by stromal cells, inflammatory cells, vasculature, extracellular matrix (ECM), and their secreted cytokines in the TME, play a pivotal role in tumor immune escape. Specific blocking of inhibition pathways in the TME is expected to effectively prevent immune escape and tolerance of tumor cells in addition to their metastasis, accordingly improving the antitumor immune response at various phases of tumor growth. Emerging nanoscale targeted drug carriers truly suit this specific requirement due to their specificity, biocompatibility, and convenience of production. This review emphasizes recent attempts to remodel the tumor immune microenvironment using novel nanoparticles, which include specifically eliminating immunosuppressive cells, reprogramming immune regulatory cells, promoting inflammatory cytokines and blocking immune checkpoints. Targeted remodeling of the immunosuppressive TME using well-designed and fabricated nanoparticles provides a promising strategy for improving the effectiveness of current immunotherapy and is greatly significant.
Collapse
Affiliation(s)
- Shan Gao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 117004, P. R. China
| | - Dongjuan Yang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 117004, P. R. China
| | - Yan Fang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 117004, P. R. China
| | - Xiaojie Lin
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 117004, P. R. China
| | - Xuechao Jin
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 117004, P. R. China
| | - Qi Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 117004, P. R. China
| | - Xiyan Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 117004, P. R. China
| | - Liyuan Ke
- Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, P. R. China
| | - Kai Shi
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 117004, P. R. China
| |
Collapse
|
35
|
Jiang L, Zhou S, Zhang X, Wu W, Jiang X. Dendrimer-based nanoparticles in cancer chemotherapy and gene therapy. SCIENCE CHINA MATERIALS 2018; 61:1404-1419. [DOI: 10.1007/s40843-018-9242-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 03/05/2018] [Indexed: 01/06/2025]
|
36
|
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: 53] [Impact Index Per Article: 7.6] [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
|
37
|
Colorimetric determination of BCR/ABL fusion genes using a nanocomposite consisting of Au@Pt nanoparticles covered with a PAMAM dendrimer and acting as a peroxidase mimic. Mikrochim Acta 2018; 185:401. [DOI: 10.1007/s00604-018-2940-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 07/30/2018] [Indexed: 12/22/2022]
|
38
|
Affiliation(s)
- Xuedan He
- University at Buffalo; State University of New York; Buffalo NY 14260 USA
| | - Scott I. Abrams
- Roswell Park Comprehensive Cancer Center; Department of Immunology; Buffalo NY 14263 USA
| | - Jonathan F. Lovell
- University at Buffalo; State University of New York; Buffalo NY 14260 USA
| |
Collapse
|
39
|
Men K, Huang R, Zhang X, Zhang R, Zhang Y, Peng Y, Tong R, Yang L, Wei Y, Duan X. Delivery of interleukin-22 binding protein (IL-22BP) gene by cationic micelle for colon cancer gene therapy. RSC Adv 2018; 8:16537-16548. [PMID: 35540501 PMCID: PMC9080254 DOI: 10.1039/c8ra02580k] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Accepted: 04/23/2018] [Indexed: 02/05/2023] Open
Abstract
Gene therapy has provided an alternative strategy for cancer therapy. As an important cytokine, interleukin-22 (IL-22) is not only critical in reinforcing innate immune defenses and tissue regeneration, but also involved in the initial establishment of tumors. A soluble-secreted receptor of the cytokine IL-22, IL-22 binding protein (IL-22BP), binds IL-22 and prevents its binding to the functional transmembrane receptor IL-22R1 complex, inhibiting IL-22-based intracellular cancer proliferation signal. In this work, a novel IL-22BP-based cancer gene therapy strategy was reported for the first time. It was established by delivering IL-22BP gene with a newly developed non-viral gene vector DMP. The DMP cationic micelles were prepared by modifying monomethoxy poly(ethylene glycol)-poly(ε-caprolactone) with DOTAP lipid through self-assembling. The anti-cancer efficacy of the DMP/IL-22BP complex was studied on a colon cancer model by intraperitoneal administration. Our results demonstrated that the secretory expressed IL-22BP cytokine effectively inhibited cancer growth both in vitro and in vivo. Multiple anti-cancer mechanisms including IL-22 blocking, apoptosis inducing, lymphocyte infiltration and angiogenesis inhibition were indicated to be involved while no pathology changes were observed in healthy tissues. These results suggest the DMP/IL-22BP complex to be a potential candidate for cancer gene therapy. Cationic DMP micelle delivered interleukin-22BP gene efficiently inhibits colon carcinoma growth, providing a novel strategy for cancer gene therapy.![]()
Collapse
|
40
|
Ganda IS, Zhong Q, Hali M, Albuquerque RLC, Padilha FF, da Rocha SRP, Whittum-Hudson JA. Dendrimer-conjugated peptide vaccine enhances clearance of Chlamydia trachomatis genital infection. Int J Pharm 2017; 527:79-91. [PMID: 28546072 PMCID: PMC5522616 DOI: 10.1016/j.ijpharm.2017.05.045] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 05/07/2017] [Accepted: 05/21/2017] [Indexed: 12/12/2022]
Abstract
Peptide-based vaccines have emerged in recent years as promising candidates in the prevention of infectious diseases. However, there are many challenges to maintaining in vivo peptide stability and enhancement of peptide immunogenicity to generate protective immunity which enhances clearance of infections. Here, a dendrimer-based carrier system is proposed for peptide-based vaccine delivery, and shows its anti-microbial feasibility in a mouse model of Chlamydia trachomatis. Chlamydiae are the most prevalent sexually transmitted bacteria worldwide, and also the causal agent of trachoma, the leading cause of preventable infectious blindness. In spite of the prevalence of this infectious agent and the many previous vaccine-related studies, there is no vaccine commercially available. The carrier system proposed consists of generation 4, hydroxyl-terminated, polyamidoamine (PAMAM) dendrimers (G4OH), to which a peptide mimic of a chlamydial glycolipid antigen-Peptide 4 (Pep4, AFPQFRSATLLL) was conjugated through an ester bond. The ester bond between G4OH and Pep4 is expected to break down mainly in the intracellular environment for antigen presentation. Pep4 conjugated to dendrimer induced Chlamydia-specific serum antibodies after subcutaneous immunizations. Further, this new vaccine formulation significantly protected immunized animals from vaginal challenge with infectious Chlamydia trachomatis, and it reduced infectious loads and tissue (genital tract) damage. Pep4 conjugated to G4OH or only mixed with peptide provided enhanced protection compared to Pep4 and adjuvant (i.e. alum), suggesting a potential adjuvant effect of the PAMAM dendrimer. Combined, these results demonstrate that hydroxyl-terminated PAMAM dendrimer is a promising polymeric nanocarrier platform for the delivery of peptide vaccines and this approach has potential to be expanded to other infectious intracellular bacteria and viruses of public health significance.
Collapse
Affiliation(s)
- Ingrid S Ganda
- Biomaterials Laboratory, Technology and Research Institute, Tiradentes University, Aracaju, SE, 49032-490, Brazil; Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI 48202, USA; Departments of Immunology and Microbiology, Internal Medicine (Rheumatology), and Ophthalmology, School of Medicine, Wayne State University, Detroit, MI 48201, USA.
| | - Qian Zhong
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI 48202, USA.
| | - Mirabela Hali
- Departments of Immunology and Microbiology, Internal Medicine (Rheumatology), and Ophthalmology, School of Medicine, Wayne State University, Detroit, MI 48201, USA.
| | - Ricardo L C Albuquerque
- Laboratory of Morphology and Structural Biology, Technology and Research Institute, Tiradentes University, Aracaju, SE, 49032-490, Brazil.
| | - Francine F Padilha
- Biomaterials Laboratory, Technology and Research Institute, Tiradentes University, Aracaju, SE, 49032-490, Brazil.
| | - Sandro R P da Rocha
- Biomaterials Laboratory, Technology and Research Institute, Tiradentes University, Aracaju, SE, 49032-490, Brazil; Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI 48202, USA.
| | - Judith A Whittum-Hudson
- Departments of Immunology and Microbiology, Internal Medicine (Rheumatology), and Ophthalmology, School of Medicine, Wayne State University, Detroit, MI 48201, USA.
| |
Collapse
|
41
|
Morris CJ, Aljayyoussi G, Mansour O, Griffiths P, Gumbleton M. Endocytic Uptake, Transport and Macromolecular Interactions of Anionic PAMAM Dendrimers within Lung Tissue. Pharm Res 2017; 34:2517-2531. [PMID: 28616685 PMCID: PMC5736778 DOI: 10.1007/s11095-017-2190-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 05/22/2017] [Indexed: 12/13/2022]
Abstract
Purpose Polyamidoamine (PAMAM) dendrimers are a promising class of nanocarrier with applications in both small and large molecule drug delivery. Here we report a comprehensive evaluation of the uptake and transport pathways that contribute to the lung disposition of dendrimers. Methods Anionic PAMAM dendrimers and control dextran probes were applied to an isolated perfused rat lung (IPRL) model and lung epithelial monolayers. Endocytosis pathways were examined in primary alveolar epithelial cultures by confocal microscopy. Molecular interactions of dendrimers with protein and lipid lung fluid components were studied using small angle neutron scattering (SANS). Results Dendrimers were absorbed across the intact lung via a passive, size-dependent transport pathway at rates slower than dextrans of similar molecular sizes. SANS investigations of concentration-dependent PAMAM transport in the IPRL confirmed no aggregation of PAMAMs with either albumin or dipalmitoylphosphatidylcholine lung lining fluid components. Distinct endocytic compartments were identified within primary alveolar epithelial cells and their functionality in the rapid uptake of fluorescent dendrimers and model macromolecular probes was confirmed by co-localisation studies. Conclusions PAMAM dendrimers display favourable lung biocompatibility but modest lung to blood absorption kinetics. These data support the investigation of dendrimer-based carriers for controlled-release drug delivery to the deep lung. Electronic supplementary material The online version of this article (doi:10.1007/s11095-017-2190-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Christopher J Morris
- School of Pharmacy, University of East Anglia, Norwich Research Park, NR4 7TJ, UK.
| | - Ghaith Aljayyoussi
- Cardiff School of Pharmacy & Pharmaceutical Sciences, Redwood Building, Cardiff, CF10 3NB, UK
| | - Omar Mansour
- Department of Pharmaceutical, Chemical and Environmental Science, University of Greenwich, Medway Campus, Kent, ME4 4TB, UK
| | - Peter Griffiths
- Department of Pharmaceutical, Chemical and Environmental Science, University of Greenwich, Medway Campus, Kent, ME4 4TB, UK
| | - Mark Gumbleton
- Cardiff School of Pharmacy & Pharmaceutical Sciences, Redwood Building, Cardiff, CF10 3NB, UK.
| |
Collapse
|
42
|
Zilio S, Vella JL, De la Fuente AC, Daftarian PM, Weed DT, Kaifer A, Marigo I, Leone K, Bronte V, Serafini P. 4PD Functionalized Dendrimers: A Flexible Tool for In Vivo Gene Silencing of Tumor-Educated Myeloid Cells. THE JOURNAL OF IMMUNOLOGY 2017; 198:4166-4177. [PMID: 28396317 DOI: 10.4049/jimmunol.1600833] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 03/13/2017] [Indexed: 12/11/2022]
Abstract
Myeloid cells play a key role in tumor progression and metastasis by providing nourishment and immune protection, as well as facilitating cancer invasion and seeding to distal sites. Although advances have been made in understanding the biology of these tumor-educated myeloid cells (TEMCs), their intrinsic plasticity challenges our further understanding of their biology. Indeed, in vitro experiments only mimic the in vivo setting, and current gene-knockout technologies do not allow the simultaneous, temporally controlled, and cell-specific silencing of multiple genes or pathways. In this article, we describe the 4PD nanoplatform, which allows the in vivo preferential transfection and in vivo tracking of TEMCs with the desired RNAs. This platform is based on the conjugation of CD124/IL-4Rα-targeting peptide with G5 PAMAM dendrimers as the loading surface and can convey therapeutic or experimental RNAs of interest. When injected i.v. in mice bearing CT26 colon carcinoma or B16 melanoma, the 4PD nanoparticles predominantly accumulate at the tumor site, transfecting intratumoral myeloid cells. The use of 4PD to deliver a combination of STAT3- and C/EBPβ-specific short hairpin RNA or miR-142-3p confirmed the importance of these genes and microRNAs in TEMC biology and indicates that silencing of both genes is necessary to increase the efficacy of immune interventions. Thus, the 4PD nanoparticle can rapidly and cost effectively modulate and assess the in vivo function of microRNAs and mRNAs in TEMCs.
Collapse
Affiliation(s)
- Serena Zilio
- Department of Microbiology and Immunology, University of Miami, Miami, FL, 33136
| | - Jennifer L Vella
- Department of Microbiology and Immunology, University of Miami, Miami, FL, 33136
| | | | - Pirouz M Daftarian
- Department of Microbiology and Immunology, University of Miami, Miami, FL, 33136
| | - Donald T Weed
- Department of Otolaryngology, University of Miami, Miami, FL, 33136
| | - Angel Kaifer
- Department of Chemistry, University of Miami, Coral Gables, FL, 33146
| | - Ilaria Marigo
- Istituto Oncologico Veneto-Istituto di Ricovero e Cura a Carattere Scientifico, IOV-IRCCS, 35128 Padova, Italy; and
| | - Kevin Leone
- Istituto Oncologico Veneto-Istituto di Ricovero e Cura a Carattere Scientifico, IOV-IRCCS, 35128 Padova, Italy; and
| | - Vincenzo Bronte
- Department of Medicine, Verona University Hospital, 37134 Verona, Italy
| | - Paolo Serafini
- Department of Microbiology and Immunology, University of Miami, Miami, FL, 33136;
| |
Collapse
|
43
|
Qiu H, Min Y, Rodgers Z, Zhang L, Wang AZ. Nanomedicine approaches to improve cancer immunotherapy. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2017; 9. [PMID: 28296286 DOI: 10.1002/wnan.1456] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 10/28/2016] [Accepted: 12/17/2016] [Indexed: 01/10/2023]
Abstract
Significant advances have been made in the field of cancer immunotherapy by orchestrating the body's immune system to eradicate cancer cells. However, safety and efficacy concerns stemming from the systemic delivery of immunomodulatory compounds limits cancer immunotherapies expansion and application. In this context, nanotechnology presents a number of advantages, such as targeted delivery to immune cells, enhanced clinical outcomes, and reduced adverse events, which may aid in the delivery of cancer vaccines and immunomodulatory agents. With this in mind, a diverse range of nanomaterials with different physicochemical characteristics have been developed to stimulate the immune system and battle cancer. In this review, we will focus on some recent developments and the potential advantages of utilizing nanotechnology within the field of cancer immunotherapy. WIREs Nanomed Nanobiotechnol 2017, 9:e1456. doi: 10.1002/wnan.1456 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Hui Qiu
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine; Department of Radiation Oncology, University of North Carolina, Chapel Hill, NC, USA.,Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yuanzeng Min
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine; Department of Radiation Oncology, University of North Carolina, Chapel Hill, NC, USA
| | - Zach Rodgers
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine; Department of Radiation Oncology, University of North Carolina, Chapel Hill, NC, USA
| | - Longzhen Zhang
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Andrew Z Wang
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine; Department of Radiation Oncology, University of North Carolina, Chapel Hill, NC, USA.,Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
44
|
Affiliation(s)
- Mingming Zhang
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, 236 Baidi Road, Nankai District, Tianjin 300192, China
| | - Yanhang Hong
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, 236 Baidi Road, Nankai District, Tianjin 300192, China
| | - Wenjuan Chen
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, 236 Baidi Road, Nankai District, Tianjin 300192, China
| | - Chun Wang
- Department
of Biomedical Engineering, University of Minnesota, 7-105 Hasselmo
Hall, 312 Church Street S. E., Minneapolis, Minnesota 55455, United States
| |
Collapse
|
45
|
Abstract
The use of gene delivery systems for the expression of antigenic proteins is an established means for activating a patient’s own immune system against the cancer they carry. Since tumor cells are poor antigen-presenting cells, cross-presentation of tumor antigens by dendritic cells (DCs) is essential for the generation of tumor-specific cytotoxic T-lymphocyte responses. A number of polymer-based nanomedicines have been developed to deliver genes into DCs, primarily by incorporating tumor-specific, antigen-encoding plasmid DNA with polycationic molecules to facilitate DNA loading and intracellular trafficking. Direct in vivo targeting of plasmid DNA to DC surface receptors can induce high transfection efficiency and long-term gene expression, essential for antigen loading onto major histocompatibility complex molecules and stimulation of T-cell responses. This chapter summarizes the physicochemical properties and biological information on polymer-based non-viral vectors used for targeting DCs, and discusses the main challenges for successful in vivo gene transfer into DCs.
Collapse
Affiliation(s)
- Kenneth A. Howard
- Department of Molecular Biology and Gen, Interdisciplinary Nanoscience Center (i, Aarhus, Denmark
| | - Thomas Vorup-Jensen
- Department of Biomedicine, Biophysical I, Aarhus University, Aarhus, Denmark
| | - Dan Peer
- Britannia Bldg, 2nd Fl, Rm 226, Tel-Aviv Univ, Dept Cell Research, Tel-Aviv, Israel
| |
Collapse
|
46
|
Zhong Q, Merkel OM, Reineke JJ, da Rocha SRP. Effect of the Route of Administration and PEGylation of Poly(amidoamine) Dendrimers on Their Systemic and Lung Cellular Biodistribution. Mol Pharm 2016; 13:1866-78. [PMID: 27148629 DOI: 10.1021/acs.molpharmaceut.6b00036] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
There are many opportunities in the development of oral inhalation (oi) formulations for the delivery of small molecule therapeutics and biologics to and through the lungs. Nanocarriers have the potential to play a key role in advancing oi technologies and pushing the boundary of the pulmonary delivery market. In this work we investigate the effect of the route of administration and PEGylation on the systemic and lung cellular biodistribution of generation 3, amino-terminated poly(amidoamine) (PAMAM) dendrimers (G3NH2). Pharmacokinetic profiles show that the dendrimers reach their peak concentration in systemic circulation within a few hours after pulmonary delivery, independent of their chemistry (PEGylated or not), charge (+24 mV for G3NH2 vs -3.7 mV for G3NH2-24PEG1000), or size (5.1 nm for G3NH2 and 9.9 nm for G3NH2-24PEG1000). However, high density of surface modification with PEG enhances pulmonary absorption and the peak plasma concentration upon pulmonary delivery. The route of administration and PEGylation also significantly impact the whole body and local (lung cellular) distribution of the dendrimers. While ca. 83% of G3NH2 is found in the lungs upon pulmonary delivery at 6.5 h post administration, only 2% reached the lungs upon intravenous (iv) delivery. Moreover, no measurable concentration of either G3NH2 or G3NH2-24PEG1000 is found in the lymph nodes upon iv administration, while these are the tissues with the second highest mass distribution of dendrimers post pulmonary delivery. Dendrimer chemistry also significantly impacts the (cellular) distribution of the nanocarriers in the lung tissue. Upon pulmonary delivery, approximately 20% of the lung endothelial cells are seen to internalize G3NH2-24PEG1000, compared to only 6% for G3NH2. Conversely, G3NH2 is more readily taken up by lung epithelial cells (35%) when compared to its PEGylated counterpart (24%). The results shown here suggest that both the pulmonary route of administration and dendrimer chemistry combined can be used to passively target tissues and cell populations of great interest, and can thus be used as guiding principles in the development of dendrimer-based drug delivery strategies in the treatment of medically relevant diseases including lung ailments as well as systemic disorders.
Collapse
Affiliation(s)
- Qian Zhong
- Department of Chemical Engineering and Materials Science, College of Engineering, Wayne State University , Detroit, Michigan 48202, United States
| | - Olivia M Merkel
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy & Health Sciences, Wayne State University , Detroit, Michigan 48201, United States.,Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universität München , 81377 München, Germany
| | - Joshua J Reineke
- Department of Pharmaceutical Sciences, College of Pharmacy, South Dakota State University , Brookings, South Dakota 57007, United States
| | - Sandro R P da Rocha
- Department of Chemical Engineering and Materials Science, College of Engineering, Wayne State University , Detroit, Michigan 48202, United States
| |
Collapse
|
47
|
Brys AK, Gowda R, Loriaux DB, Robertson GP, Mosca PJ. Nanotechnology-based strategies for combating toxicity and resistance in melanoma therapy. Biotechnol Adv 2016; 34:565-577. [PMID: 26826558 DOI: 10.1016/j.biotechadv.2016.01.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Revised: 11/15/2015] [Accepted: 01/16/2016] [Indexed: 12/17/2022]
Abstract
Drug toxicity and resistance remain formidable challenges in cancer treatment and represent an area of increasing attention in the case of melanoma. Nanotechnology represents a paradigm-shifting field with the potential to mitigate drug resistance while improving drug delivery and minimizing toxicity. Recent clinical and pre-clinical studies have demonstrated how a diverse array of nanoparticles may be harnessed to circumvent known mechanisms of drug resistance in melanoma to improve therapeutic efficacy. In this review, we discuss known mechanisms of resistance to various melanoma therapies and possible nanotechnology-based strategies that could be used to overcome these barriers and improve the pharmacologic arsenal available to combat advanced stage melanoma.
Collapse
Affiliation(s)
- Adam K Brys
- Department of Surgery, Division of Surgical Oncology, Duke University Medical Center, Durham, NC 27710, United States
| | - Raghavendra Gowda
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Daniel B Loriaux
- Department of Surgery, Division of Surgical Oncology, Duke University Medical Center, Durham, NC 27710, United States
| | - Gavin P Robertson
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Paul J Mosca
- Department of Surgery, Division of Surgical Oncology, Duke University Medical Center, Durham, NC 27710, United States.
| |
Collapse
|
48
|
Kumar M, Kovalski L, Broyles D, Hunt EA, Daftarian P, Dikici E, Daunert S, Deo SK. Design and development of high bioluminescent resonance energy transfer efficiency hybrid-imaging constructs. Anal Biochem 2016; 498:1-7. [PMID: 26772160 DOI: 10.1016/j.ab.2015.11.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 11/06/2015] [Accepted: 11/23/2015] [Indexed: 12/22/2022]
Abstract
Here we describe the design and construction of an imaging construct with high bioluminescent resonance energy transfer (BRET) efficiency that is composed of multiple quantum dots (QDs; λem = 655 nm) self-assembled onto a bioluminescent protein, Renilla luciferase (Rluc). This is facilitated by the streptavidin-biotin interaction, allowing the facile formation of a hybrid-imaging construct (HIC) comprising up to six QDs (acceptor) grafted onto a light-emitting Rluc (donor) core. The resulting assembly of multiple acceptors surrounding a donor permits this construct to exhibit high resonance energy transfer efficiency (∼64.8%). The HIC was characterized using fluorescence excitation anisotropy measurements and high-resolution transmission electron microscopy. To demonstrate the application of our construct, a generation-5 (G5) polyamidoamine dendrimer (PAMAM) nanocarrier was loaded with our HIC for in vitro and in vivo imaging. We envision that this design of multiple acceptors and bioluminescent donor will lead to the development of new BRET-based systems useful in sensing, imaging, and other bioanalytical applications.
Collapse
Affiliation(s)
- Manoj Kumar
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; Indian Institute of Technology (BHU), Department of Chemical Engineering and School of Biomedical Engineering, Varanasi, UP 221005, India
| | - Letícia Kovalski
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - David Broyles
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Eric A Hunt
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Pirouz Daftarian
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Emre Dikici
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Sylvia Daunert
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Sapna K Deo
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA.
| |
Collapse
|
49
|
Abstract
In the two decades since their initial discovery, DNA vaccines technologies have come a long way. Unfortunately, when applied to human subjects inadequate immunogenicity is still the biggest challenge for practical DNA vaccine use. Many different strategies have been tested in preclinical models to address this problem, including novel plasmid vectors and codon optimization to enhance antigen expression, new gene transfection systems or electroporation to increase delivery efficiency, protein or live virus vector boosting regimens to maximise immune stimulation, and formulation of DNA vaccines with traditional or molecular adjuvants. Better understanding of the mechanisms of action of DNA vaccines has also enabled better use of the intrinsic host response to DNA to improve vaccine immunogenicity. This review summarizes recent advances in DNA vaccine technologies and related intracellular events and how these might impact on future directions of DNA vaccine development.
Collapse
Affiliation(s)
- Lei Li
- a Vaxine Pty Ltd, Bedford Park , Adelaide , Australia.,b Department of Diabetes and Endocrinology , Flinders University, Flinders Medical Centre , Adelaide , SA , Australia
| | - Nikolai Petrovsky
- a Vaxine Pty Ltd, Bedford Park , Adelaide , Australia.,b Department of Diabetes and Endocrinology , Flinders University, Flinders Medical Centre , Adelaide , SA , Australia
| |
Collapse
|
50
|
Genetic Immunization With In Vivo Dendritic Cell-targeting Liposomal DNA Vaccine Carrier Induces Long-lasting Antitumor Immune Response. Mol Ther 2015; 24:385-397. [PMID: 26666450 PMCID: PMC4817821 DOI: 10.1038/mt.2015.215] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 11/22/2015] [Indexed: 12/23/2022] Open
Abstract
A major limiting factor retarding the clinical success of dendritic cell (DC)-based genetic immunizations (DNA vaccination) is the scarcity of biologically safe and effective carrier systems for targeting the antigen-encoded DNA vaccines to DCs under in vivo settings. Herein, we report on a potent, mannose receptor selective in vivo DC-targeting liposomes of a novel cationic amphiphile with mannose-mimicking shikimoyl head-group. Flow cytometric experiments with cells isolated from draining lymph nodes of mice s.c. immunized with lipoplexes of pGFP plasmid (model DNA vaccine) using anti-CD11c antibody-labeled magnetic beads revealed in vivo DC-targeting properties of the presently described liposomal DNA vaccine carrier. Importantly, s.c. immunizations of mice with electrostatic complex of the in vivo DC-targeting liposome and melanoma antigen-encoded DNA vaccine (p-CMV-MART1) induced long-lasting antimelanoma immune response (100 days post melanoma tumor challenge) with remarkable memory response (more than 6 months after the second tumor challenge). The presently described direct in vivo DC-targeting liposomal DNA vaccine carrier is expected to find future exploitations toward designing effective vaccines for various infectious diseases and cancers.
Collapse
|