1
|
Shirazi MMA, Saedi TA, Moghaddam ZS, Nemati M, Shiri R, Negahdari B, Goradel NH. Nanotechnology and nano-sized tools: Newer approaches to circumvent oncolytic adenovirus limitations. Pharmacol Ther 2024; 256:108611. [PMID: 38387653 DOI: 10.1016/j.pharmthera.2024.108611] [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: 10/19/2023] [Revised: 01/03/2024] [Accepted: 02/14/2024] [Indexed: 02/24/2024]
Abstract
Oncolytic adenoviruses (OAds), engineered Ads preferentially infect and lyse tumor cells, have attracted remarkable attention as immunotherapy weapons for the treatment of various malignancies. Despite hopeful successes in preclinical investigations and translation into clinical phases, they face some challenges that thwart their therapeutic effectiveness, including low infectivity of cancer cells, liver sequestration, pre-existing neutralizing antibodies, physical barriers to the spread of Ads, and immunosuppressive TME. Nanotechnology and nano-sized tools provide several advantages to overcome these limitations of OAds. Nano-sized tools could improve the therapeutic efficacy of OAds by enhancing infectivity and cellular uptake, targeting and protecting from pre-existing immune responses, masking and preventing liver tropism, and co-delivery with other therapeutic agents. Herein, we reviewed the constructs of various OAds and their application in clinical trials, as well as the limitations they have faced. Furthermore, we emphasized the potential applications of nanotechnology to solve the constraints of OAds to improve their anti-tumor activities.
Collapse
Affiliation(s)
| | - Tayebeh Azam Saedi
- Department of Genetics, Faculty of Science, Islamic Azad University, Tonekabon Branch, Tonekabon, Iran
| | - Zahra Samadi Moghaddam
- Department of Medical Biotechnology, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Mahnaz Nemati
- Amir Oncology Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Shiri
- Department of Basic Sciences, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Babak Negahdari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nasser Hashemi Goradel
- Department of Medical Biotechnology, Maragheh University of Medical Sciences, Maragheh, Iran; Arthropod-Borne Diseases Research Centre, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
2
|
Walsh MJ, Ali LR, Lenehan P, Kureshi CT, Kureshi R, Dougan M, Knipe DM, Dougan SK. Blockade of innate inflammatory cytokines TNF α, IL-1 β, or IL-6 overcomes virotherapy-induced cancer equilibrium to promote tumor regression. IMMUNOTHERAPY ADVANCES 2023; 3:ltad011. [PMID: 37461742 PMCID: PMC10349916 DOI: 10.1093/immadv/ltad011] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 06/30/2023] [Indexed: 07/20/2023] Open
Abstract
Cancer therapeutics can lead to immune equilibrium in which the immune response controls tumor cell expansion without fully eliminating the cancer. The factors involved in this equilibrium remain incompletely understood, especially those that would antagonize the anti-tumor immune response and lead to tumor outgrowth. We previously demonstrated that continuous treatment with a non-replicating herpes simplex virus 1 expressing interleukin (IL)-12 induces a state of cancer immune equilibrium highly dependent on interferon-γ. We profiled the IL-12 virotherapy-induced immune equilibrium in murine melanoma, identifying blockade of innate inflammatory cytokines, tumor necrosis factor alpha (TNFα), IL-1β, or IL-6 as possible synergistic interventions. Antibody depletions of each of these cytokines enhanced survival in mice treated with IL-12 virotherapy and helped to overcome equilibrium in some tumors. Single-cell RNA-sequencing demonstrated that blockade of inflammatory cytokines resulted in downregulation of overlapping inflammatory pathways in macrophages, shifting immune equilibrium towards tumor clearance, and raising the possibility that TNFα blockade could synergize with existing cancer immunotherapies.
Collapse
Affiliation(s)
- Michael J Walsh
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Harvard Program in Virology, Boston, MA, USA
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Lestat R Ali
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Patrick Lenehan
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Courtney T Kureshi
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Rakeeb Kureshi
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Michael Dougan
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - David M Knipe
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Stephanie K Dougan
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
3
|
Piroonpan T, Rimdusit P, Taechutrakul S, Pasanphan W. pH-Responsive Water-Soluble Chitosan Amphiphilic Core–Shell Nanoparticles: Radiation-Assisted Green Synthesis and Drug-Controlled Release Studies. Pharmaceutics 2023; 15:pharmaceutics15030847. [PMID: 36986708 PMCID: PMC10052151 DOI: 10.3390/pharmaceutics15030847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/22/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023] Open
Abstract
This work aims to apply water radiolysis-mediated green synthesis of amphiphilic core–shell water-soluble chitosan nanoparticles (WCS NPs) via free radical graft copolymerization in an aqueous solution using irradiation. Robust grafting poly(ethylene glycol) monomethacrylate (PEGMA) comb-like brushes were established onto WCS NPs modified with hydrophobic deoxycholic acid (DC) using two aqueous solution systems, i.e., pure water and water/ethanol. The degree of grafting (DG) of the robust grafted poly(PEGMA) segments was varied from 0 to ~250% by varying radiation-absorbed doses from 0 to 30 kGy. Using reactive WCS NPs as a water-soluble polymeric template, a high amount of DC conjugation and a high degree of poly(PEGMA) grafted segments brought about high moieties of hydrophobic DC and a high DG of the poly(PEGMA) hydrophilic functions; meanwhile, the water solubility and NP dispersion were also markedly improved. The DC-WCS-PG building block was excellently self-assembled into the core–shell nanoarchitecture. The DC-WCS-PG NPs efficiently encapsulated water-insoluble anticancer and antifungal drugs, i.e., paclitaxel (PTX) and berberine (BBR) (~360 mg/g). The DC-WCS-PG NPs met the role of controlled release with a pH-responsive function due to WCS compartments, and they showed a steady state for maintaining drugs for up to >10 days. The DC-WCS-PG NPs prolonged the inhibition capacity of BBR against the growth of S. ampelinum for 30 days. In vitro cytotoxicity results of the PTX-loaded DC-WCS-PG NPs with human breast cancer cells and human skin fibroblast cells proved the role of the DC-WCS-PG NPs as a promising nanoplatform for controlling drug release and reducing the side effects of the drugs on normal cells.
Collapse
Affiliation(s)
- Thananchai Piroonpan
- Center of Radiation Processing for Polymer Modification and Nanotechnology (CRPN), Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
| | - Pakjira Rimdusit
- Center of Radiation Processing for Polymer Modification and Nanotechnology (CRPN), Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
| | - Saowaluk Taechutrakul
- Center of Radiation Processing for Polymer Modification and Nanotechnology (CRPN), Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
- Department of Materials Science, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
| | - Wanvimol Pasanphan
- Center of Radiation Processing for Polymer Modification and Nanotechnology (CRPN), Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
- Department of Materials Science, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
- Correspondence: ; Tel.: +662-577-5555 (ext. 646515)
| |
Collapse
|
4
|
The Dilemma of HSV-1 Oncolytic Virus Delivery: The Method Choice and Hurdles. Int J Mol Sci 2023; 24:ijms24043681. [PMID: 36835091 PMCID: PMC9962028 DOI: 10.3390/ijms24043681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/03/2023] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
Oncolytic viruses (OVs) have emerged as effective gene therapy and immunotherapy drugs. As an important gene delivery platform, the integration of exogenous genes into OVs has become a novel path for the advancement of OV therapy, while the herpes simplex virus type 1 (HSV-1) is the most commonly used. However, the current mode of administration of HSV-1 oncolytic virus is mainly based on the tumor in situ injection, which limits the application of such OV drugs to a certain extent. Intravenous administration offers a solution to the systemic distribution of OV drugs but is ambiguous in terms of efficacy and safety. The main reason is the synergistic role of innate and adaptive immunity of the immune system in the response against the HSV-1 oncolytic virus, which is rapidly cleared by the body's immune system before it reaches the tumor, a process that is accompanied by side effects. This article reviews different administration methods of HSV-1 oncolytic virus in the process of tumor treatment, especially the research progress in intravenous administration. It also discusses immune constraints and solutions of intravenous administration with the intent to provide new insights into HSV-1 delivery for OV therapy.
Collapse
|
5
|
Lin J, Sun S, Zhao K, Gao F, Wang R, Li Q, Zhou Y, Zhang J, Li Y, Wang X, Du L, Wang S, Li Z, Lu H, Lan Y, Song D, Guo W, Chen Y, Gao F, Zhao Y, Fan R, Guan J, He W. Oncolytic Parapoxvirus induces Gasdermin E-mediated pyroptosis and activates antitumor immunity. Nat Commun 2023; 14:224. [PMID: 36641456 PMCID: PMC9840172 DOI: 10.1038/s41467-023-35917-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/06/2023] [Indexed: 01/15/2023] Open
Abstract
The advantage of oncolytic viruses (OV) in cancer therapy is their dual effect of directly killing tumours while prompting anti-tumour immune response. Oncolytic parapoxvirus ovis (ORFV) and other OVs are thought to induce apoptosis, but apoptosis, being the immunogenically inert compared to other types of cell death, does not explain the highly inflamed microenvironment in OV-challenged tumors. Here we show that ORFV and its recombinant therapeutic derivatives are able to trigger tumor cell pyroptosis via Gasdermin E (GSDME). This effect is especially prominent in GSDME-low tumor cells, in which ORFV-challenge pre-stabilizes GSDME by decreasing its ubiquitination and subsequently initiates pyroptosis. Consistently, GSDME depletion reduces the proportion of intratumoral cytotoxic T lymphocytes, pyroptotic cell death and the success of tumor ORFV virotherapy. In vivo, the OV preferentially accumulates in the tumour upon systemic delivery and elicits pyroptotic tumor killing. Consequentially, ORFV sensitizes immunologically 'cold' tumors to checkpoint blockade. This study thus highlights the critical role of GSDME-mediated pyroptosis in oncolytic ORFV-based antitumor immunity and identifies combinatorial cancer therapy strategies.
Collapse
Affiliation(s)
- Jing Lin
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Shihui Sun
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Kui Zhao
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Fei Gao
- Department of Laboratory Animals, College of Animal Science, Jilin University, 130062, Changchun, China
| | - Renling Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Qi Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Yanlong Zhou
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Jing Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Yue Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Xinyue Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Le Du
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Shuai Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Zi Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Huijun Lu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Yungang Lan
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Deguang Song
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Wei Guo
- Department of Hematology, The first hospital of Jilin University, 130021, Changchun, China
| | - Yujia Chen
- Department of Gastrointestinal Surgery, The first hospital of Jilin University, 130021, Changchun, China
| | - Feng Gao
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China
| | - Yicheng Zhao
- Changchun University of Chinese Medicine, 130017, Changchun, China
| | - Rongrong Fan
- Department of Biosciences and Nutrition, Karolinska Institutet, 14183, Huddinge, Sweden
| | - Jiyu Guan
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China.
| | - Wenqi He
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062, Changchun, China.
| |
Collapse
|
6
|
Thambi T, Hong J, Yoon AR, Yun CO. Challenges and progress toward tumor-targeted therapy by systemic delivery of polymer-complexed oncolytic adenoviruses. Cancer Gene Ther 2022; 29:1321-1331. [PMID: 35444290 PMCID: PMC9576595 DOI: 10.1038/s41417-022-00469-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 03/10/2022] [Accepted: 03/29/2022] [Indexed: 11/22/2022]
Abstract
Oncolytic adenovirus (oAd) elicits antitumor activity by preferential viral replication in cancer cells. However, poor systemic administrability or suboptimal intratumoral retainment of the virus remains a major challenge toward maximizing the antitumor activity of oAd in a clinical environment. To surmount these issues, a variety of non-immunogenic polymers has been used to modify the surface of oAds chemically or physically. Complexation of oAd with polymers can effectively evade the host immune response and reduces nonspecific liver sequestration. The tumor-specific delivery of these complexes can be further improved upon by inclusion of tumor-targeting moieties on the surface. Therefore, modification of the Ad surface using polymers is viewed as a potential strategy to enhance the delivery of Ad via systemic administration. This review aims to provide a comprehensive overview of polymer-complexed Ads, their progress, and future challenges in cancer treatment.
Collapse
Affiliation(s)
- Thavasyappan Thambi
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro Seongdong-gu, Seoul, 04763, Korea
| | - JinWoo Hong
- GeneMedicine CO., Ltd., 222 Wangsimni-ro Seongdong-gu, Seoul, 04763, Seoul, Korea
| | - A-Rum Yoon
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro Seongdong-gu, Seoul, 04763, Korea
- Hanyang Institute of Bioscience and Biotechnology (HY-IBB), Hanyang University, Seoul, 04763, Korea
| | - Chae-Ok Yun
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro Seongdong-gu, Seoul, 04763, Korea.
- GeneMedicine CO., Ltd., 222 Wangsimni-ro Seongdong-gu, Seoul, 04763, Seoul, Korea.
- Hanyang Institute of Bioscience and Biotechnology (HY-IBB), Hanyang University, Seoul, 04763, Korea.
- Institute of Nano Science and Technology (INST), Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Korea.
| |
Collapse
|
7
|
Macchi S, Zubair M, Hill R, Alwan N, Khan Y, Ali N, Guisbiers G, Berry B, Siraj N. Improved Photophysical Properties of Ionic Material-Based Combination Chemo/PDT Nanomedicine. ACS APPLIED BIO MATERIALS 2021; 4:7708-7718. [PMID: 35006702 PMCID: PMC8900487 DOI: 10.1021/acsabm.1c00961] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Herein, a cost-effective and prompt approach to develop ionic material-based combination nanodrugs for cancer therapy is presented. A chemotherapeutic (phosphonium) cation and photodynamic therapeutic (porphyrin) anion are combined using a single step ion exchange reaction. Afterward, a nanomedicine is prepared from this ionic materials-based combination drug using a simplistic strategy of reprecipitation. Improved photophysical characteristics such as a slower nonradiative rate constant, an enhanced phosphorescence emission, a longer lifetime, and a bathochromic shift in absorbance spectra of porphyrin are observed in the presence of a chemotherapeutic countercation. The photodynamic therapeutic activity of nanomedicines is investigated by measuring the singlet oxygen quantum yield using two probes. As compared to the parent porphyrin compound, the synthesized combination material showed a 2-fold increase in the reactive oxygen species quantum yield, due to inhibition of face-to-face aggregation of porphyrin units in the presence of bulky chemotherapeutic ions. The dark cytotoxicity of combination therapy nanomedicines in the MCF-7 (cancerous breast) cell line is also increased as compared to their corresponding parent compounds in vitro. This is due to the high cellular uptake of the combination nanomedicines as compared to that of the free drug. Further, selective toxicity toward cancer cells was acquired by functionalizing nanomedicine with folic acid followed by incubation with MCF-7 and MCF-10A (noncancerous breast). Light toxicity experiments indicate that the synthesized ionic nanomedicine shows a greater cell death than either parent drug due to the improved photophysical properties and effective combination effect. This facile and economical strategy can easily be utilized in the future to develop many other combination ionic nanomedicines with improved photodynamics.
Collapse
Affiliation(s)
- Samantha Macchi
- Department of Chemistry, University of Arkansas at Little Rock, 2801 South University Avenue, Little Rock, Arkansas 72204, United States
| | - Mohd Zubair
- Department of Biology, University of Arkansas at Little Rock, 2801 South University Avenue, Little Rock, Arkansas 72204, United States
| | - Robert Hill
- Department of Chemistry, University of Arkansas at Little Rock, 2801 South University Avenue, Little Rock, Arkansas 72204, United States
| | - Nabeel Alwan
- Department of Chemistry, University of Arkansas at Little Rock, 2801 South University Avenue, Little Rock, Arkansas 72204, United States
| | - Yusuf Khan
- Department of Electrical and Computer Engineering, University of Texas at Austin, Austin, Texas 78712, United States
| | - Nawab Ali
- Department of Biology, University of Arkansas at Little Rock, 2801 South University Avenue, Little Rock, Arkansas 72204, United States
| | - Grégory Guisbiers
- Department of Physics and Astronomy, University of Arkansas at Little Rock, 2801 South University Avenue, Little Rock, Arkansas 72204, United States
| | - Brian Berry
- Department of Chemistry, University of Arkansas at Little Rock, 2801 South University Avenue, Little Rock, Arkansas 72204, United States
| | - Noureen Siraj
- Department of Chemistry, University of Arkansas at Little Rock, 2801 South University Avenue, Little Rock, Arkansas 72204, United States
| |
Collapse
|
8
|
Lan T, Chen L, Wei X. Inflammatory Cytokines in Cancer: Comprehensive Understanding and Clinical Progress in Gene Therapy. Cells 2021; 10:E100. [PMID: 33429846 PMCID: PMC7827947 DOI: 10.3390/cells10010100] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/30/2020] [Accepted: 01/05/2021] [Indexed: 02/06/2023] Open
Abstract
The relationship between chronic inflammation and neoplastic diseases is not fully understood. The inflammatory microenvironment of a tumor is an intricate network that consists of numerous types of cells, cytokines, enzymes and signaling pathways. Recent evidence shows that the crucial components of cancer-related inflammation are involved in a coordinated system to influence the development of cancer, which may shed light on the development of potential anticancer therapies. Since the last century, considerable effort has been devoted to developing gene therapies for life-threatening diseases. When it comes to modulating the inflammatory microenvironment for cancer therapy, inflammatory cytokines are the most efficient targets. In this manuscript, we provide a comprehensive review of the relationship between inflammation and cancer development, especially focusing on inflammatory cytokines. We also summarize the clinical trials for gene therapy targeting inflammatory cytokines for cancer treatment. Future perspectives concerned with new gene-editing technology and novel gene delivery systems are finally provided.
Collapse
Affiliation(s)
- Tianxia Lan
- Laboratory of Aging Research and Cancer Drug Target, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China; (T.L.); (L.C.)
- State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China
| | - Li Chen
- Laboratory of Aging Research and Cancer Drug Target, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China; (T.L.); (L.C.)
- State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China; (T.L.); (L.C.)
- State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China
| |
Collapse
|
9
|
Gao X, Ding J, Long Q, Zhan C. Virus-mimetic systems for cancer diagnosis and therapy. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 13:e1692. [PMID: 33354937 DOI: 10.1002/wnan.1692] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/13/2020] [Accepted: 11/30/2020] [Indexed: 01/02/2023]
Abstract
Over past decades, various strategies have been developed to enhance the delivery efficiency of therapeutics and imaging agents to tumor tissues. However, the therapeutic outcome of tumors to date have not been significantly improved, which can be partly attributed to the weak targeting ability, fast elimination, and low stability of conventional delivery systems. Viruses are the most efficient agents for gene transfer, serving as a valuable source of inspiration for designing nanoparticle-based delivery systems. Based on the properties of viruses, including well-defined geometry, precise composition, easy modification, stable construction, and specific infection, researchers attempt to design biocompatible delivery vectors by mimicking virus assembly and using the vector system to selectively concentrate drugs or imaging probes in tumors with mitigated toxicity and improved efficacy. In this review, we introduce common viruses features and provide an overview of various virus-mimetic strategies for cancer therapy and diagnosis. The challenges faced by virus-mimetic systems are also discussed. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Xihui Gao
- School of Basic Medical Sciences & Center of Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University, Shanghai, China
| | - Junqiang Ding
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, Shanghai, China
| | - Qianqian Long
- School of Basic Medical Sciences & Center of Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University, Shanghai, China
| | - Changyou Zhan
- School of Basic Medical Sciences & Center of Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University, Shanghai, China.,State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, China
| |
Collapse
|
10
|
Jamieson TR, Poutou J, Ilkow CS. Redirecting oncolytic viruses: Engineering opportunists to take control of the tumour microenvironment. Cytokine Growth Factor Rev 2020; 56:102-114. [DOI: 10.1016/j.cytogfr.2020.07.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 07/02/2020] [Indexed: 12/12/2022]
|
11
|
Abstract
Therapeutic viral gene delivery is an emerging technology which aims to correct genetic mutations by introducing new genetic information to cells either to correct a faulty gene or to initiate cell death in oncolytic treatments. In recent years, significant scientific progress has led to several clinical trials resulting in the approval of gene therapies for human treatment. However, successful therapies remain limited due to a number of challenges such as inefficient cell uptake, low transduction efficiency (TE), limited tropism, liver toxicity and immune response. To adress these issues and increase the number of available therapies, additives from a broad range of materials like polymers, peptides, lipids, nanoparticles, and small molecules have been applied so far. The scope of this review is to highlight these selected delivery systems from a materials perspective.
Collapse
Affiliation(s)
- Kübra Kaygisiz
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | | |
Collapse
|
12
|
Surface Modification of Adenovirus Vector to Improve Immunogenicity and Tropism. Methods Mol Biol 2020. [PMID: 32959253 DOI: 10.1007/978-1-0716-0795-4_18] [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: 09/29/2023]
Abstract
Although adenovirus is a popular vector for delivering genes, there are several drawbacks that limit its effectiveness, including tropism and both the innate and adaptive immune responses. One approach that has been used to ameliorate these drawbacks is PEGylation of the virus with subsequent modification to add functional moieties for the purpose of cell targeting or enhancing infection. Here, we describe a general approach for PEGylating adenovirus and conjugating cell-penetrating peptides to the surface of the virus to impart the ability to transduce CAR-negative cells.
Collapse
|
13
|
Ye ZQ, Zou CL, Chen HB, Lv QY, Wu RQ, Gu DN. Folate-conjugated herpes simplex virus for retargeting to tumor cells. J Gene Med 2020; 22:e3177. [PMID: 32096291 DOI: 10.1002/jgm.3177] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 02/13/2020] [Accepted: 02/19/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Herpes simplex virus type 1 (HSV-1)-mediated oncolytic therapy is a promising cancer treatment modality. However, viral tropism is considered to be one of the major stumbling blocks to the development of HSV-1 as an anticancer agent. METHODS The surface of oncolytic HSV-1 G207 was covalently modified with folate-poly (ethylene glycol) conjugate (FA-PEG). The specificities and tumor targeting efficiencies of modified or unmodified G207 particles were analyzed by a real-time polymerase chain reaction at the level of cell attachment and entry. Immune responses were assessed by an interleukin-6 release assay from RAW264.7 macrophages. Biodistribution and in vivo antitumoral activity after intravenous delivery was evaluated in BALB/c nude mice bearing subcutaneous KB xenograft tumors. RESULTS FA-PEG-HSV exhibited enhanced targeting specificity for folate receptor over-expressing tumor cells and had lower immunogenicity than the unmodified HSV. In vivo, the FA-PEG-HSV group revealed an increased anti-tumor efficiency and tumor targeting specificity compared to the naked HSV. CONCLUSIONS These results indicate that folate-conjugated HSV G207 presents a folate receptor-targeted oncolytic virus with a potential therapeutic value via retargeting to tumor cells.
Collapse
Affiliation(s)
- Zhi-Qiang Ye
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Chang-Lin Zou
- Department of Chemoradiotherapy, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Han-Bin Chen
- Department of Chemoradiotherapy, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Qi-Yuan Lv
- Department of Chemoradiotherapy, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Ruo-Qi Wu
- Department of Chemoradiotherapy, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Dian-Na Gu
- Department of Chemoradiotherapy, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| |
Collapse
|
14
|
Wu J, Wu H, Nakagawa S, Gao J. Virus-derived materials: bury the hatchet with old foes. Biomater Sci 2020; 8:1058-1072. [DOI: 10.1039/c9bm01383k] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Viruses, with special architecture and unique biological nature, can be utilized for various biomedical applications.
Collapse
Affiliation(s)
- Jiahe Wu
- Institute of Pharmaceutics
- College of Pharmaceutical Sciences
- Zhejiang University
- Hangzhou 310058
- China
| | - Honghui Wu
- Institute of Pharmaceutics
- College of Pharmaceutical Sciences
- Zhejiang University
- Hangzhou 310058
- China
| | - Shinsaku Nakagawa
- Department of Pharmaceutics
- Graduate School of Pharmaceutical Sciences
- Osaka University
- Suita
- Japan
| | - Jianqing Gao
- Institute of Pharmaceutics
- College of Pharmaceutical Sciences
- Zhejiang University
- Hangzhou 310058
- China
| |
Collapse
|
15
|
Sun Y, Lv X, Ding P, Wang L, Sun Y, Li S, Zhang H, Gao Z. Exploring the functions of polymers in adenovirus-mediated gene delivery: Evading immune response and redirecting tropism. Acta Biomater 2019; 97:93-104. [PMID: 31386928 DOI: 10.1016/j.actbio.2019.06.059] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 05/31/2019] [Accepted: 06/24/2019] [Indexed: 02/06/2023]
Abstract
Adenovirus (Ad) is a promising viral carrier in gene therapy because of its unique attribution. However, clinical applications of Ad vectors are currently restricted by their immunogenicity and broad native tropism. To address these obstacles, a variety of nonimmunogenic polymers are utilized to modify Ad vectors chemically or physically. In this review, we systemically discuss the functions of polymers in Ad-mediated gene delivery from two aspects: evading the host immune responses to Ads and redirecting Ad tropism. With polyethylene glycol (PEG) first in order, a variety of polymers have been developed to shield the surface of Ad vectors and well accomplished to evade the host immune response, block CAR-dependant cellular uptake, and reduce accumulation in the liver. In addition, shielding Ad vectors with targeted polymers (including targeting ligand-conjugated polymers and bio-responsive polymers) can also efficiently retarget Ad vectors to tumor tissues and reduce their distribution in nontargeted tissues. With its potential to evade the immune response and retarget Ad vectors, modification with polymers has been generally regarded as a promising strategy to facilitate the clinical applications of Ad vectors for virotherapy. STATEMENT OF SIGNIFICANCE: There is no doubt that Adenovirus (Ads) are attractive vectors for gene therapy, with high sophistication and effectiveness in overcoming both extra- and intracellular barriers, which cannot be exceeded by any other nonviral gene vectors. Unfortunately, their clinical applications are still restricted by some critical hurdles, including immunogenicity and native broad tropism. Therefore, a variety of elegant strategies have been developed from various angles to address these hurdles. Among these various strategies, coating Ads with nonimmunogenic polymers has attracted much attention. In this review, we systemically discuss the functions of polymers in Ad-mediated gene delivery from two aspects: evading the host immune responses to Ads and redirecting Ad tropism. In addition, the key factors in Ad modification with polymers have been highlighted and summarized to provide guiding theory for the design of more effective and safer polymer-Ad hybrid gene vectors.
Collapse
Affiliation(s)
- Yanping Sun
- Department of Pharmacy, Hebei University of Science and Technology, Shijiazhuang 050018, China; State Key Laboratory Breeding Base - Hebei Province Key Laboratory of Molecular Chemistry for Drugs, Hebei University of Science and Technology, Shijiazhuang 050018, China; Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang 050018, China
| | - Xiaoqian Lv
- Department of Pharmacy, Hebei University of Science and Technology, Shijiazhuang 050018, China
| | - Pingtian Ding
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Long Wang
- State Key Laboratory Breeding Base - Hebei Province Key Laboratory of Molecular Chemistry for Drugs, Hebei University of Science and Technology, Shijiazhuang 050018, China; Department of Family and Consumer Sciences, California State University, Long Beach, CA 90840, USA
| | - Yongjun Sun
- Department of Pharmacy, Hebei University of Science and Technology, Shijiazhuang 050018, China; Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang 050018, China
| | - Shuo Li
- State Key Laboratory Breeding Base - Hebei Province Key Laboratory of Molecular Chemistry for Drugs, Hebei University of Science and Technology, Shijiazhuang 050018, China
| | - Huimin Zhang
- State Key Laboratory Breeding Base - Hebei Province Key Laboratory of Molecular Chemistry for Drugs, Hebei University of Science and Technology, Shijiazhuang 050018, China
| | - Zibin Gao
- Department of Pharmacy, Hebei University of Science and Technology, Shijiazhuang 050018, China; State Key Laboratory Breeding Base - Hebei Province Key Laboratory of Molecular Chemistry for Drugs, Hebei University of Science and Technology, Shijiazhuang 050018, China; Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang 050018, China.
| |
Collapse
|
16
|
Marofi F, Vahedi G, hasanzadeh A, Salarinasab S, Arzhanga P, Khademi B, Farshdousti Hagh M. Mesenchymal stem cells as the game‐changing tools in the treatment of various organs disorders: Mirage or reality? J Cell Physiol 2018; 234:1268-1288. [DOI: 10.1002/jcp.27152] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 07/05/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Faroogh Marofi
- Department of Hematology Faculty of Medicine, Tabriz University of Medical Sciences Tabriz Iran
| | - Ghasem Vahedi
- Faculty of Veterinary Medicine, University of Tehran Tehran Iran
| | - Ali hasanzadeh
- Department of Hematology Faculty of Medicine, Tabriz University of Medical Sciences Tabriz Iran
| | - Sadegh Salarinasab
- Department of Biochemistry and Clinical Laboratories Faculty of Medicine, Tabriz University of Medical Science Tabriz Iran
| | - Pishva Arzhanga
- Department of Biochemistry and Diet Therapy Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences Tabriz Iran
| | - Bahareh Khademi
- Department of Medical Genetic Faculty of Medicine, Tabriz University of Medical Sciences Tabriz Iran
| | | |
Collapse
|
17
|
Machitani M, Sakurai F, Wakabayashi K, Nakatani K, Tachibana M, Kato N, Fujiwara T, Mizuguchi H. Suppression of Oncolytic Adenovirus-Mediated Hepatotoxicity by Liver-Specific Inhibition of NF-κB. MOLECULAR THERAPY-ONCOLYTICS 2017; 7:76-85. [PMID: 29202008 PMCID: PMC5704103 DOI: 10.1016/j.omto.2017.10.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 10/22/2017] [Indexed: 11/28/2022]
Abstract
Telomerase-specific replication-competent adenoviruses (Ads), i.e., TRADs, which possess an E1 gene expression cassette driven by the human telomerase reverse transcriptase promoter, are promising agents for cancer treatment. However, even though oncolytic Ads, including TRAD, are intratumorally administered, they are disseminated from the tumor to systemic circulation, causing concern about oncolytic Ad-mediated hepatotoxicity (due mainly to leaky expression of Ad genes in liver). We reported that inhibition of nuclear factor-κB (NF-κB) leads to the suppression of replication-incompetent Ad vector-mediated hepatotoxicity via reduction of the leaky expression of Ad genes in liver. Here, to develop a TRAD with an improved safety profile, we designed a TRAD that carries a liver-specific promoter-driven dominant-negative IκBα (DNIκBα) expression cassette (TRAD-DNIκBα). Compared with a conventional TRAD, TRAD-DNIκBα showed hepatocyte-specific inhibition of NF-κB signaling and significantly reduced Ad gene expression and replication in the normal human hepatocyte cell line. TRAD-induced hepatotoxicity was largely suppressed in mice following intravenous administration of TRAD-DNIκBα. However, the replication profiles and oncolytic activities of TRAD-DNIκBα were comparable with those of the conventional TRAD in human non-hepatic tumor cells. These results indicate that oncolytic Ads containing the liver-specific DNIκBα expression cassette have improved safety profiles without inhibiting oncolytic activities.
Collapse
Affiliation(s)
- Mitsuhiro Machitani
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan.,Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Fuminori Sakurai
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan.,Laboratory of Regulatory Sciences for Oligonucleotide Therapeutics, Clinical Drug Development Unit, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Keisaku Wakabayashi
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Kosuke Nakatani
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Masashi Tachibana
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan.,Global Center for Medical Engineering and Informatics, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Nobuyuki Kato
- Department of Tumor Virology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Toshiyoshi Fujiwara
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Hiroyuki Mizuguchi
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan.,Global Center for Medical Engineering and Informatics, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.,Laboratory of Hepatocyte Regulation, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito, Asagi, Ibaraki, Osaka 567-0085, Japan.,iPS Cell-Based Research Project on Hepatic Toxicity and Metabolism, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan.,Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
18
|
|
19
|
Wan L, Yao X, Faiola F, Liu B, Zhang T, Tabata Y, Mizuguchi H, Nakagawa S, Gao JQ, Zhao RC. Coating with spermine-pullulan polymer enhances adenoviral transduction of mesenchymal stem cells. Int J Nanomedicine 2016; 11:6763-6769. [PMID: 28008251 PMCID: PMC5167484 DOI: 10.2147/ijn.s109897] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are adult stem cells with multilineage potential, which makes them attractive tools for regenerative medicine applications. Efficient gene transfer into MSCs is essential not only for basic research in developmental biology but also for therapeutic applications involving gene-modification in regenerative medicine. Adenovirus vectors (Advs) can efficiently and transiently introduce an exogenous gene into many cell types via their primary receptors, the coxsackievirus and adenovirus receptors, but not into MSCs, which are deficient in coxsackievirus and adenovirus receptors expression. To overcome this problem, we developed an Adv coated with a spermine-pullulan (SP) cationic polymer and investigated its physicochemical properties and internalization mechanisms. We demonstrated that the SP coating could enhance adenoviral transduction of MSCs without detectable cytotoxicity or effects on differentiation. Our results argue in favor of the potentiality of the SP-coated Adv as a prototype vector for efficient and safe transduction of MSCs.
Collapse
Affiliation(s)
- Li Wan
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing
| | - Xinglei Yao
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing; Institute of Pharmaceutics, Zhejiang University, Hangzhou; State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences
| | - Francesco Faiola
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences
| | - Bojun Liu
- YouAn Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Tianyuan Zhang
- Institute of Pharmaceutics, Zhejiang University, Hangzhou
| | - Yasuhiko Tabata
- Department of Biomaterials, Field of Tissue Engineering, Institute for Frontier Medical Sciences, Kyoto University, Kyoto
| | - Hiroyuki Mizuguchi
- Department of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka
| | - Shinsaku Nakagawa
- Department of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Jian-Qing Gao
- Institute of Pharmaceutics, Zhejiang University, Hangzhou
| | - Robert Chunhua Zhao
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Beijing
| |
Collapse
|
20
|
Fan G, Fan M, Wang Q, Jiang J, Wan Y, Gong T, Zhang Z, Sun X. Bio-inspired polymer envelopes around adenoviral vectors to reduce immunogenicity and improve in vivo kinetics. Acta Biomater 2016; 30:94-105. [PMID: 26546972 DOI: 10.1016/j.actbio.2015.11.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Revised: 10/27/2015] [Accepted: 11/03/2015] [Indexed: 10/22/2022]
Abstract
Adenoviral vectors have attracted substantial interest for systemic tumor gene therapy, but further work is needed to reduce their immunogenicity and alter their biodistribution before they can be used in the clinic. Here we describe a bio-inspired, cleavable PEGylated β-cyclodextrin-polyethyleneimine conjugate (CDPCP) that spontaneously coats adenovirus in solution. This cleavable PEG coating reduces the innate and adaptive immunogenicity of adenovirus particles, as well as improves their biodistribution away from the liver and into the tumor. Insertion of a matrix metalloproteinase substrate sequence into the conjugate allows PEG cleavage at the tumor site, simultaneously reducing liver biodistribution and increasing transgene expression in tumors, thereby avoiding the "PEG dilemma". Cationic β-cyclodextrin-PEI not only provides electrostatic attraction to promote envelope attachment to the viral capsid, but it also improves vector internalization and transduction after PEG cleavage. These results suggest that CDPCP may help expand the use of adenoviral vectors in cancer gene therapy. STATEMENT OF SIGNIFICANCE The synthesized β-cyclodextrin-PEI-MMP-cleavable-PEG polymer (CDPCP), held great potential for gene therapy when applied for adenovirus coating. The β-cyclodextrin-PEI provided a powerful electrostatic attraction to attach the whole polymer onto the viral capsid, while the MMPs-cleavable PEG reduced innate and adaptive immunogenicity and improved the biodistribution of adenovirus vectors due to the tumor-specific enzyme triggered PEG cleavage. More importantly, an ingenious cooperation between the two components could solve the PEG dilemma. The CDPCP/Ad complexes exhibited a comprehensive and valued profile to be a candidate vector for future tumor gene therapy, we believe the current investigation on this kind of biomaterial may be of particular interest to the readership of Acta biomaterialia.
Collapse
|
21
|
Amphiphilic block copolymer of SN38 prodrugs by atom transfer radical polymerization: Synthesis, kinetic studies and self-assembly. J Control Release 2015; 213:e124. [DOI: 10.1016/j.jconrel.2015.05.209] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
22
|
Peng LH, Zhang YH, Han LJ, Zhang CZ, Wu JH, Wang XR, Gao JQ, Mao ZW. Cell Membrane Capsules for Encapsulation of Chemotherapeutic and Cancer Cell Targeting in Vivo. ACS APPLIED MATERIALS & INTERFACES 2015; 7:18628-37. [PMID: 26262951 DOI: 10.1021/acsami.5b05065] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Systemic administration of chemotherapeutic agents can cause indiscriminate drug distribution and severe toxicity. Until now, encapsulation and targeting of drugs have typically relied on synthetic vehicles, which cannot minimize the clearance by the renal system and may also increase the risk of chemical side effects. Cell membrane capsules (CMCs) provide a generic and far more natural approach to the challenges of drug encapsulation and delivery in vivo. Here aptamer AS1411, which can recognize and bind overexpressed nucleolin on a cancer cell membrane, was chemically conjugated onto CMCs. As a result, AS1411 modified CMCs showed enhanced ingestion in certain cancer cells in vitro and accumulation in mouse cancer xenografts in vivo. Chemotherapeutics and contrast agents with therapeutically significant concentrations can be packaged into CMCs by reversible permeating their plasma membranes. The systematic administration of cancer targeting CMCs loaded with doxorubicin hydrochloride can significantly inhibit tumor growth in mouse xenografts, with significantly reduced toxicity compared to free drug. These findings suggest that cancer targeting CMCs may have considerable benefits in drug delivery and cancer treatment.
Collapse
Affiliation(s)
- Li-Hua Peng
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou 310027, People's Republic of China
| | - Yuan-Hong Zhang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University , Hangzhou 310027, People's Republic of China
| | - Li-Jie Han
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University , Hangzhou 310027, People's Republic of China
| | - Chen-Zhen Zhang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou 310027, People's Republic of China
| | - Jia-He Wu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou 310027, People's Republic of China
| | - Xia-Rong Wang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou 310027, People's Republic of China
| | - Jian-Qing Gao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou 310027, People's Republic of China
| | - Zheng-Wei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University , Hangzhou 310027, People's Republic of China
| |
Collapse
|
23
|
Dubey P, Gidwani B, Pandey R, Shukla SS. In vitro and in vivo evaluation of PEGylated nanoparticles of bendamustine for treatment of lung cancer. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2015; 44:1491-7. [DOI: 10.3109/21691401.2015.1052466] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Pooja Dubey
- Columbia Institute of Pharmacy, Raipur, Tekari, Chhattisgarh, India
| | - Bina Gidwani
- University Institute of Pharmacy, Pandit Ravishankar Shukla University, Raipur, Chhattisgarh, India
| | - Ravindra Pandey
- Columbia Institute of Pharmacy, Raipur, Tekari, Chhattisgarh, India
| | | |
Collapse
|
24
|
Choi JW, Jung SJ, Kasala D, Hwang JK, Hu J, Bae YH, Yun CO. pH-sensitive oncolytic adenovirus hybrid targeting acidic tumor microenvironment and angiogenesis. J Control Release 2015; 205:134-43. [PMID: 25575865 DOI: 10.1016/j.jconrel.2015.01.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 12/24/2014] [Accepted: 01/04/2015] [Indexed: 12/14/2022]
Abstract
Although oncolytic adenoviruses (Ads) are an attractive option for cancer gene therapy, the intravenous administration of naked Ad still encounters unfavorable host responses, non-specific interactions, and heterogeneity in targeted cancer cells. To overcome these obstacles and achieve specific targeting of the tumor microenvironment, Ad was coated with the pH-sensitive block copolymer, methoxy poly(ethylene glycol)-b-poly(l-histidine-co-l-phenylalanine) (PEGbPHF). The physicochemical properties of the generated nanocomplex, Ad/PEGbPHF, were assessed. At pH6.4, GFP-expressing Ad/PEGbPHF induced significantly higher GFP expression than naked Ad in both coxsackie and adenovirus receptor (CAR)-positive and -negative cells. To assess the therapeutic efficacy of the Ad/PEGbPHF complex platform, an oncolytic Ad expressing VEGF promoter-targeting transcriptional repressor (KOX) was used to form complexes. At pH6.4, KOX/PEGbPHF significantly suppressed VEGF gene expression, cancer cell migration, vessel sprouting, and cancer cell killing effect compared to naked KOX or KOX/PEGbPHF at pH7.4, demonstrating that KOX/PEGbPHF can overcome the lack of CAR that is frequently observed in tumor tissues. The antitumor activity of KOX/PEGbPHF systemically administered to a tumor xenograft model was significantly higher than that of naked KOX. Furthermore, KOX/PEGbPHF showed lower hepatic toxicity and did not induce an innate immune response against Ad. Altogether, these results demonstrate that pH-sensitive polymer-coated Ad complex significantly increases net positive charge upon exposure to hypoxic tumor microenvironment, allowing passive targeting to the tumor tissue. It may offer superior potential for systemic therapy, due to its improved tumor selectivity, increased therapeutic efficacy, and lower toxicity compared to naked KOX.
Collapse
Affiliation(s)
- Joung-Woo Choi
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsinmi-ro, Seongdong-gu, Seoul, Republic of Korea
| | - Soo-Jung Jung
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsinmi-ro, Seongdong-gu, Seoul, Republic of Korea
| | - Dayananda Kasala
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsinmi-ro, Seongdong-gu, Seoul, Republic of Korea
| | - June Kyu Hwang
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsinmi-ro, Seongdong-gu, Seoul, Republic of Korea
| | - Jun Hu
- Department of Pharmaceutics and Pharmaceutical Chemistry, The University of Utah, 30S 2000 E, Room 2972, Salt Lake City, UT 84112, USA
| | - You Han Bae
- Department of Pharmaceutics and Pharmaceutical Chemistry, The University of Utah, 30S 2000 E, Room 2972, Salt Lake City, UT 84112, USA; Utah-Inha Drug Delivery Systems (DDS) and Advanced Therapeutics Research Center, 7-50 Songdo-dong, Yeonsu-gu, Incheon 406-840, Republic of Korea.
| | - Chae-Ok Yun
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsinmi-ro, Seongdong-gu, Seoul, Republic of Korea.
| |
Collapse
|
25
|
Li Q, Yang S, Zhu L, Kang H, Qu X, Liu R, Huang Y. Dual-stimuli sensitive keratin graft PHPMA as physiological trigger responsive drug carriers. Polym Chem 2015. [DOI: 10.1039/c4py01750a] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Keratin graft PHPMA copolymers were successfully synthesized and can be used as drug carriers with physiological stimuli responsive properties.
Collapse
Affiliation(s)
- Qinmei Li
- Sate Key Laboratory of Polymer Physics and Chemistry
- Beijing National Laboratory of Molecular Sciences
- Institute of Chemistry
- Chinese Academy of Sciences
- Beijing 100190
| | - Saina Yang
- Sate Key Laboratory of Polymer Physics and Chemistry
- Beijing National Laboratory of Molecular Sciences
- Institute of Chemistry
- Chinese Academy of Sciences
- Beijing 100190
| | - Lijun Zhu
- Sate Key Laboratory of Polymer Physics and Chemistry
- Beijing National Laboratory of Molecular Sciences
- Institute of Chemistry
- Chinese Academy of Sciences
- Beijing 100190
| | - Hongliang Kang
- Sate Key Laboratory of Polymer Physics and Chemistry
- Beijing National Laboratory of Molecular Sciences
- Institute of Chemistry
- Chinese Academy of Sciences
- Beijing 100190
| | - Xiaozhong Qu
- University of Chinese Academy of Science
- Beijing 100049
- China
| | - Ruigang Liu
- Sate Key Laboratory of Polymer Physics and Chemistry
- Beijing National Laboratory of Molecular Sciences
- Institute of Chemistry
- Chinese Academy of Sciences
- Beijing 100190
| | - Yong Huang
- Sate Key Laboratory of Polymer Physics and Chemistry
- Beijing National Laboratory of Molecular Sciences
- Institute of Chemistry
- Chinese Academy of Sciences
- Beijing 100190
| |
Collapse
|
26
|
Bagheri M, Shateri S, Niknejad H, Entezami AA. Thermosensitive biotinylated hydroxypropyl cellulose-based polymer micelles as a nano-carrier for cancer-targeted drug delivery. JOURNAL OF POLYMER RESEARCH 2014. [DOI: 10.1007/s10965-014-0567-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
27
|
Yao X, Zhou N, Wan L, Su X, Sun Z, Mizuguchi H, Yoshioka Y, Nakagawa S, Zhao RC, Gao JQ. Polyethyleneimine-coating enhances adenoviral transduction of mesenchymal stem cells. Biochem Biophys Res Commun 2014; 447:383-7. [PMID: 24727452 DOI: 10.1016/j.bbrc.2014.03.142] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 03/25/2014] [Indexed: 12/18/2022]
Abstract
Mesenchymal stem cells (MSCs) are non-hematopoietic cells with multi-lineage potential, which makes them attractive targets for regenerative medicine applications. Efficient gene transfer into MSCs is essential for basic research in developmental biology and for therapeutic applications involving gene-modification in regenerative medicine. Adenovirus vectors (Advs) can efficiently and transiently introduce an exogenous gene into many cell types via their primary receptors, the coxsackievirus and adenovirus receptors (CARs), but not into MSCs, which lack CAR expression. To overcome this problem, an Adv coated with cationic polymer polyethyleneimine (PEI) was developed. In this study, we demonstrated that PEI coating with an optimal ratio can enhance adenoviral transduction of MSCs without cytotoxicity. We also investigated the physicochemical properties and internalization mechanisms of the PEI-coated Adv. These results could help to evaluate the potentiality of the PEI-coated Adv as a prototype vector for efficient and safe transduction into MSCs.
Collapse
Affiliation(s)
- Xinglei Yao
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, 5# Dongdansantiao, 100005 Beijing, PR China; Institute of Pharmaceutics, Zhejiang University(2), 866 Yuhangtang Road, Hangzhou 310058, PR China
| | - Na Zhou
- Department of Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, 1# Shuaifuyuan, 100730 Beijing, PR China
| | - Li Wan
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, 5# Dongdansantiao, 100005 Beijing, PR China
| | - Xiaodong Su
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, 5# Dongdansantiao, 100005 Beijing, PR China
| | - Zhao Sun
- Department of Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, 1# Shuaifuyuan, 100730 Beijing, PR China
| | - Hiroyuki Mizuguchi
- Department of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yasuo Yoshioka
- Department of Toxicology and Safety Science, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Shinsaku Nakagawa
- Department of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Robert Chunhua Zhao
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Peking Union Medical College Hospital, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, 5# Dongdansantiao, 100005 Beijing, PR China.
| | - Jian-Qing Gao
- Institute of Pharmaceutics, Zhejiang University(2), 866 Yuhangtang Road, Hangzhou 310058, PR China.
| |
Collapse
|
28
|
Peptide-based technologies to alter adenoviral vector tropism: ways and means for systemic treatment of cancer. Viruses 2014; 6:1540-63. [PMID: 24699364 PMCID: PMC4014709 DOI: 10.3390/v6041540] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 03/15/2014] [Accepted: 03/20/2014] [Indexed: 12/11/2022] Open
Abstract
Due to the fundamental progress in elucidating the molecular mechanisms of human diseases and the arrival of the post-genomic era, increasing numbers of therapeutic genes and cellular targets are available for gene therapy. Meanwhile, the most important challenge is to develop gene delivery vectors with high efficiency through target cell selectivity, in particular under in situ conditions. The most widely used vector system to transduce cells is based on adenovirus (Ad). Recent endeavors in the development of selective Ad vectors that target cells or tissues of interest and spare the alteration of all others have focused on the modification of the virus broad natural tropism. A popular way of Ad targeting is achieved by directing the vector towards distinct cellular receptors. Redirecting can be accomplished by linking custom-made peptides with specific affinity to cellular surface proteins via genetic integration, chemical coupling or bridging with dual-specific adapter molecules. Ideally, targeted vectors are incapable of entering cells via their native receptors. Such altered vectors offer new opportunities to delineate functional genomics in a natural environment and may enable efficient systemic therapeutic approaches. This review provides a summary of current state-of-the-art techniques to specifically target adenovirus-based gene delivery vectors.
Collapse
|
29
|
Efficient lung orthotopic tumor-growth suppression of oncolytic adenovirus complexed with RGD-targeted bioreducible polymer. Gene Ther 2014; 21:476-83. [PMID: 24598892 DOI: 10.1038/gt.2014.18] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 12/11/2013] [Accepted: 01/21/2014] [Indexed: 01/24/2023]
Abstract
Oncolytic adenoviruses (Ad) have been developed for the eradication of tumors. Although they hold much promise as a cancer therapy, they have a short blood circulation time and high liver toxicity. An effective strategy to overcome these problems has been complexing Ad with shielding materials. However, the therapeutic efficacy of the Ad complexes has also been an issue because passive accumulation does not allow for sufficient delivery of Ad to the cancer cells. To enhance the therapeutic efficacy of the polymer-coated Ads, the attachment of a targeting moiety to polymer-coated Ad vectors is inescapable. Our lab has previously reported the potential use of Arg-Gly-Asp (RGD)-targeted bioreducible polymers with a polyethylene glycol (PEG) linker for delivering oncolytic Ads. We have shown the enhanced in vitro transduction efficiency and increased cancer-killing effect with producing progeny oncolytic Ad particles. In addition, we have shown significant tumor-growth inhibition of the polymer-shielded Ad in an in vivo lung orthotopic tumor model. The shielding effect of the Ad surface with the polymers allowed evasion of host immune responses and reduction of liver toxicity. This data demonstrates that the RGD-conjugated bioreducible polymer for delivering the oncolytic Ad vectors could be utilized for cancer therapy via systemic administration.
Collapse
|
30
|
Chen H, Zheng X, Di B, Wang D, Zhang Y, Xia H, Mao Q. Aptamer modification improves the adenoviral transduction of malignant glioma cells. J Biotechnol 2013. [DOI: 10.1016/j.jbiotec.2013.10.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
31
|
Park JS, Yang HN, Woo DG, Jeon SY, Park KH. Poly(N-isopropylacrylamide-co-acrylic acid) nanogels for tracing and delivering genes to human mesenchymal stem cells. Biomaterials 2013; 34:8819-34. [PMID: 23937912 DOI: 10.1016/j.biomaterials.2013.07.082] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 07/23/2013] [Indexed: 12/24/2022]
Abstract
Drugs, proteins, and cells can be macro- and micro-encapsulated by unique materials that respond to specific stimuli. The phases and hydrophobic interactions of these materials are reversibly altered by environmental stimuli such as pH and temperature. These changes can lead to self-assembly of the materials, which enables controlled drug release and safe gene delivery into cells and tissues. The fate of stem cells delivered by such methods is of great interest. The formation of transgenic tissues requires genes to be delivered safely into stem cells. A cell tracing vehicle and a gene delivery carrier were simultaneously introduced into human mesenchymal stem cells (hMSCs). A thermo-sensitive hydrogel, poly(N-isopropylacrylamide-co-acrylic acid) (p(NiPAAm-co-AAc)), was created to generate self-assembled nanoparticles with nanogel characteristics. Hydrophobic interactions mediated the binding of the carboxyl group on the outside of p(NiPAAm-co-AAc) with the amine group of iron oxide. Nanogels carrying iron oxide and a fluorescent dye were complexed with specific genes. These nanogels could be internalized by hMSCs, and the transplantation of these cells into mice was monitored by in vivo imaging. Self-assembled p(NiPAAm-co-dAAc) nanogels complexed with green fluorescent protein were highly expressed in hMSCs and are a potential material for gene delivery.
Collapse
Affiliation(s)
- Ji Sun Park
- Department of Biomedical Science, College of Life Science, CHA University, 3F, Yatap Acecore, 502 Yatap-dong Bundang-gu, Seongnam-si, Republic of Korea
| | | | | | | | | |
Collapse
|
32
|
Hayashi Y, Higashi T, Motoyama K, Mori Y, Jono H, Ando Y, Arima H. Design and evaluation of polyamidoamine dendrimer conjugate with PEG,α-cyclodextrin and lactose as a novel hepatocyte-selective gene carrierin vitroandin vivo. J Drug Target 2013; 21:487-96. [DOI: 10.3109/1061186x.2013.769105] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
33
|
Wan Y, Han J, Fan G, Zhang Z, Gong T, Sun X. Enzyme-responsive liposomes modified adenoviral vectors for enhanced tumor cell transduction and reduced immunogenicity. Biomaterials 2013; 34:3020-30. [PMID: 23360783 DOI: 10.1016/j.biomaterials.2012.12.051] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Accepted: 12/14/2012] [Indexed: 10/27/2022]
Abstract
Limitations of adenoviral (Ad) vectors for cancer gene therapy could be overcome by their combination with pharmaceutical technologies. Here we show that an enzyme-responsive liposomal formulation could significantly enhance the tumor cell transduction abilities and reduce the immunogenicity of Ad vectors. In the current research, the enzymatically cleavable PEG-lipids composed of a PEG/matrix metalloproteinase (MMP)-substrate peptide/cholesterol (PPC) were synthesized and characterized by (1)H NMR and TOF MS ES(+). The obtained MMP-cleavable lipids were inserted into the anionic liposomal Ad vectors (AL-Ad) by the post-insertion method. The results of in vitro infection assays indicated that the enzymatically cleavable formulation (PPC-AL-Ad) displayed a much higher gene expression than naked Ad5 and the non-cleavable PEG-lipid modified Ad vectors in tumor cells. More importantly, PPC-AL-Ad induces a lower production of neutralizing antibody and lower innate immune response, as well as significantly reduced liver toxicity in vivo. These findings suggest that PPC-AL-Ad is a promising system for gene delivery in tumor therapy.
Collapse
Affiliation(s)
- Yu Wan
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, Sichuan, PR China
| | | | | | | | | | | |
Collapse
|
34
|
Reetz J, Herchenröder O, Schmidt A, Pützer BM. Vector Technology and Cell Targeting: Peptide-Tagged Adenoviral Vectors as a Powerful Tool for Cell Specific Targeting. Regen Med 2013. [DOI: 10.1007/978-94-007-5690-8_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
35
|
Singarapu K, Pal I, Ramsey JD. Polyethylene glycol–grafted polyethylenimine used to enhance adenovirus gene delivery. J Biomed Mater Res A 2012; 101:1857-64. [DOI: 10.1002/jbm.a.34483] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Revised: 10/05/2012] [Accepted: 10/09/2012] [Indexed: 11/08/2022]
Affiliation(s)
- Kumar Singarapu
- School of Chemical Engineering, Oklahoma State University, Stillwater, Oklahoma
| | - Ivy Pal
- School of Chemical Engineering, Oklahoma State University, Stillwater, Oklahoma
| | - Joshua D. Ramsey
- School of Chemical Engineering, Oklahoma State University, Stillwater, Oklahoma
| |
Collapse
|
36
|
Jie LY, Cai LL, Wang LJ, Ying XY, Yu RS, Zhang MM, Du YZ. Actively-targeted LTVSPWY peptide-modified magnetic nanoparticles for tumor imaging. Int J Nanomedicine 2012; 7:3981-9. [PMID: 22866005 PMCID: PMC3410692 DOI: 10.2147/ijn.s33593] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Magnetic resonance imaging (MRI) is widely used in modern clinical medicine as a diagnostic tool, and provides noninvasive and three-dimensional visualization of biological phenomena in living organisms with high spatial and temporal resolution. Therefore, considerable attention has been paid to magnetic nanoparticles as MRI contrast agents with efficient targeting ability and cellular internalization ability, which make it possible to offer higher contrast and information-rich images for detection of disease. METHODS LTVSPWY peptide-modified PEGylated chitosan (LTVSPWY-PEG-CS) was synthesized by chemical reaction, and the chemical structure was confirmed by (1)H-NMR. LTVSPWY-PEG-CS-modified magnetic nanoparticles were prepared successfully using the solvent diffusion method. Their particle size, size distribution, and zeta potential were measured by dynamic light scattering and electrophoretic mobility, and their surface morphology was investigated by transmission electron microscopy. To investigate their selective targeting ability, the cellular uptake of the LTVSPWY-PEG-CS-modified magnetic nanoparticles was observed in a cocultured system of SKOV-3 cells which overexpress HER2 and A549 cells which are HER2-negative. The in vitro cytotoxicity of these nanoparticles in SKOV-3 and A549 cells was measured using the MTT method. The SKOV-3-bearing nude mouse model was used to investigate the tumor targeting ability of the magnetic nanoparticles in vivo. RESULTS The average diameter and zeta potential of the LTVSPWY-PEG-CS-modified magnetic nanoparticles was 267.3 ± 23.4 nm and 30.5 ± 7.0 mV, respectively, with a narrow size distribution and spherical morphology. In vitro cytotoxicity tests demonstrated that these magnetic nanoparticles were carriers suitable for use in cancer diagnostics with low toxicity. With modification of the LTVSPWY homing peptide, magnetic nanoparticles could be selectively taken up by SKOV-3 cells overexpressing HER2 when cocultured with HER2-negative A549 cells. In vivo biodistribution results suggest that treatment with LTVSPWY-PEG-CS-modified magnetic nanoparticles/DiR enabled tumors to be identified and diagnosed more rapidly and efficiently in vivo. CONCLUSION LTVSPWY-PEG-CS-modified magnetic nanoparticles are a promising contrast agent for early detection of tumors overexpressing HER2 and further diagnostic application.
Collapse
Affiliation(s)
- Li-Yong Jie
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
37
|
Yao XL, Yoshioka Y, Ruan GX, Chen YZ, Mizuguchi H, Mukai Y, Okada N, Gao JQ, Nakagawa S. Optimization and internalization mechanisms of PEGylated adenovirus vector with targeting peptide for cancer gene therapy. Biomacromolecules 2012; 13:2402-9. [PMID: 22746837 DOI: 10.1021/bm300665u] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
We have previously developed a novel adenovirus vector (Adv) that targeted tumor tissues/vasculatures after systemic administration. The surface of this Adv is conjugated with CGKRK tumor homing peptide by the cross-linking reaction of polyethyleneglycol (PEG). In this study, we showed that the condition of PEG modification was important to minimize the gene expression in normal tissues after systemic treatment. When Adv was modified only with PEG-linked CGKRK, its luciferase expression was enhanced even in the liver tissue, as well as the tumor tissue. However, in the reaction with the mixture of non-cross-linking PEG and PEG-linked CGKRK, we found out that the best modification could suppress its gene expression in the liver, without losing that in the tumor. We also studied the internalization mechanisms of CGKRK-conjugated Adv. Results suggested that there is a specific interaction of the CGKRK peptide with a receptor at the cell surface enabling efficient internalization of CGKRK-conjugated Adv. The presence of cell-surface heparan sulfate is important receptor for the cellular binding and uptake of CGKRK-conjugated Adv. Moreover, macropinocytosis-mediated endocytosis is also important in endocytosis of CGKRK-conjugated Adv, aside from clathrin-mediated and caveolae-mediated endocytosis. These results could help evaluate the potentiality of CGKRK-conjugated Adv as a prototype vector with suitable efficacy and safety for systemic cancer gene therapy.
Collapse
Affiliation(s)
- Xing-Lei Yao
- Institute of Pharmaceutics, Zhejiang University, Hangzhou 310058, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Evolution of oncolytic adenovirus for cancer treatment. Adv Drug Deliv Rev 2012; 64:720-9. [PMID: 22212901 DOI: 10.1016/j.addr.2011.12.011] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Revised: 12/14/2011] [Accepted: 12/16/2011] [Indexed: 12/18/2022]
Abstract
Oncolytic adenovirus (Ad) has been used in cancer gene therapy largely due to its ability to selectively infect and replicate in tumor cells. However, because the oncolytic antitumor activity is insufficient to effectively eliminate tumors, various strategies have been devised to improve the therapeutic efficacy. Single-vector Ads "armed" with short hairpin RNA, cytokines, or matrix-modulating proteins have been developed. Two clear advantages are viral amplification of the therapeutic gene, and the additive effects of oncolytic and therapeutic gene-mediated antitumor activities. To develop systemically injectable Ad carriers, strategies to modify the Ad surface with polymers, liposomes, or nanoparticles have been shown to extend circulation time, reduce immunogenicity, and result in increased antitumor effect as well as lower accumulation and toxicity in liver. Specific targeting platforms for tumor-selective oncolytic therapies against both primary and metastatic cancers have been developed. This review will focus on updated strategies to develop potent oncolytic Ads for use in cancer treatment.
Collapse
|
39
|
Matsui H, Sakurai F, Katayama K, Yamaguchi T, Okamoto S, Takahira K, Tachibana M, Nakagawa S, Mizuguchi H. A hexon-specific PEGylated adenovirus vector utilizing blood coagulation factor X. Biomaterials 2012; 33:3743-55. [DOI: 10.1016/j.biomaterials.2012.02.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Accepted: 01/12/2012] [Indexed: 12/01/2022]
|
40
|
In vivo delivery of cell-permeable antisense hypoxia-inducible factor 1α oligonucleotide to adipose tissue reduces adiposity in obese mice. J Control Release 2012; 161:1-9. [PMID: 22546680 DOI: 10.1016/j.jconrel.2012.04.026] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Revised: 04/13/2012] [Accepted: 04/18/2012] [Indexed: 01/06/2023]
Abstract
Ongoing research has gradually recognized and understood the importance of adipose tissue (AT) angiogenesis as a key modulating factor of adipogenesis in the development of obesity. Previously, we carried out the first in vitro demonstration of the down-regulation of hypoxic angiogenesis during adipogenesis using cell-permeable chemical conjugates composed of antisense hypoxia-inducible factor 1α (HIF1α) oligonucleotide (ASO) and low-molecular weight protamine (LMWP). To further confirm the in vivo feasibility, we administered ASO-LMWP conjugates (AL) to diet-induced obese (DIO) mice by intraperitoneal injection (IP). Results showed that the AL conjugates significantly reduced the body weight, total fat tissue weight, and plasma lipid concentrations in the mice. Moreover, the AL conjugates not only decreased liver weight and hepatic triglyceride concentration but also significantly attenuated subcutaneous adipocyte cell size, which was conversely increased in the AL-untreated high-fat diet (HFD) group. Interestingly, more blood vessels were observed in the HFD group than in the lean group, indicating that blood vessel development could induce growth of the fat mass. This pattern was reversed in the AL-treated groups, which displayed a decrease in blood vessel density compared to the AL-untreated HFD group. This study presents the first in vivo evidence, in an obese mouse model, of the feasibility of achieving a biological treatment modality for obesity by blocking the angiogenic transcriptional factor HIF1α, thereby limiting angiogenesis, via the use of an adipose tissue-permeable ASO-LMWP.
Collapse
|
41
|
Li BH, Kim SM, Yoo SB, Kim MJ, Jahng JW, Lee JH. Recombinant human nerve growth factor (rhNGF-β) gene transfer promotes regeneration of crush-injured mental nerve in rats. Oral Surg Oral Med Oral Pathol Oral Radiol 2012; 113:e26-34. [DOI: 10.1016/j.tripleo.2011.07.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Revised: 06/22/2011] [Accepted: 07/11/2011] [Indexed: 10/14/2022]
|
42
|
Polymer coatings for delivery of nucleic acid therapeutics. J Control Release 2012; 161:537-53. [PMID: 22366547 DOI: 10.1016/j.jconrel.2012.02.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Revised: 02/08/2012] [Accepted: 02/11/2012] [Indexed: 12/15/2022]
Abstract
Gene delivery remains the greatest challenge in applying nucleic acid therapeutic for a broad range of diseases. Combining stability during the delivery phase with activation and transgene expression following arrival at the target site requires sophisticated vectors that can discriminate between cell types and respond to target-associated conditions to trigger expression. Efficient intravenous delivery is the greatest single hurdle, with synthetic vectors frequently found to be unstable in the harsh conditions of the bloodstream, and viral vectors often recognized avidly by both the innate and the adaptive immune system. Both types of vectors benefit from coating with hydrophilic polymers. Self-assembling polyelectrolyte non-viral vectors can achieve both steric and lateral stabilization following surface coating, endowing them with much improved systemic circulation properties and better access to disseminated targets; similarly viral vectors can be 'stealthed' and their physical properties modulated by surface coating. Both types of vectors may also have their tropism changed following chemical linkage of novel ligands to the polymer coating. These families of vectors go some way towards realizing the goal of efficient systemic delivery of genes and should find a range of important uses in bringing this still-emerging field to fruition.
Collapse
|
43
|
Duffy MR, Parker AL, Bradshaw AC, Baker AH. Manipulation of adenovirus interactions with host factors for gene therapy applications. Nanomedicine (Lond) 2012; 7:271-88. [DOI: 10.2217/nnm.11.186] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Nanomedicine based on the use of adenovirus vectors for therapeutic gene delivery shows broad potential. Specific targeting for many gene therapy applications, such as metastatic cancers or cardiovascular diseases requires intravascular delivery of the vector. However, a major barrier to successful adenovirus vector targeting follows systemic delivery, as upon contact with the bloodstream the virus interacts with a variety of host proteins, in particular coagulation factor X, which mediates profound liver gene transfer. This inherent hepatic tropism combined with macrophage scavenging minimizes the efficacy of the virus at the desired sites and induces toxic side effects. Understanding the complex, multifaceted interactions of adenovirus with host factors is of vital importance to the design of safer vectors with improved efficacy and pharmacokinetic profiles. Increased knowledge of adenovirus biology provides the opportunity to develop innovative strategies to detarget the virus from the liver following intravascular delivery and redirect the vector to disease areas.
Collapse
Affiliation(s)
- Margaret R Duffy
- British Heart Foundation Glasgow Cardiovascular Research Center, College of Medical, Veterinary & Life Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Alan L Parker
- British Heart Foundation Glasgow Cardiovascular Research Center, College of Medical, Veterinary & Life Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Angela C Bradshaw
- British Heart Foundation Glasgow Cardiovascular Research Center, College of Medical, Veterinary & Life Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Andrew H Baker
- British Heart Foundation Glasgow Cardiovascular Research Center, College of Medical, Veterinary & Life Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| |
Collapse
|
44
|
Li Q, Zhu L, Liu R, Huang D, Jin X, Che N, Li Z, Qu X, Kang H, Huang Y. Biological stimuli responsive drug carriers based on keratin for triggerable drug delivery. ACTA ACUST UNITED AC 2012. [DOI: 10.1039/c2jm34136k] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
45
|
Shimizu T, Ichihara M, Yoshioka Y, Ishida T, Nakagawa S, Kiwada H. Intravenous Administration of Polyethylene Glycol-Coated (PEGylated) Proteins and PEGylated Adenovirus Elicits an Anti-PEG Immunoglobulin M Response. Biol Pharm Bull 2012; 35:1336-42. [DOI: 10.1248/bpb.b12-00276] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Taro Shimizu
- Department of Pharmacokinetics and Biopharmaceutics, Subdivision of Biopharmaceutical Sciences, Institute of Health Biosciences, The University of Tokushima
| | - Masako Ichihara
- Department of Pharmacokinetics and Biopharmaceutics, Subdivision of Biopharmaceutical Sciences, Institute of Health Biosciences, The University of Tokushima
| | - Yasuo Yoshioka
- Department of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Tatsuhiro Ishida
- Department of Pharmacokinetics and Biopharmaceutics, Subdivision of Biopharmaceutical Sciences, Institute of Health Biosciences, The University of Tokushima
| | - Shinsaku Nakagawa
- Department of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Hiroshi Kiwada
- Department of Pharmacokinetics and Biopharmaceutics, Subdivision of Biopharmaceutical Sciences, Institute of Health Biosciences, The University of Tokushima
| |
Collapse
|
46
|
Cai LL, Liu P, Li X, Huang X, Ye YQ, Chen FY, Yuan H, Hu FQ, Du YZ. RGD peptide-mediated chitosan-based polymeric micelles targeting delivery for integrin-overexpressing tumor cells. Int J Nanomedicine 2011; 6:3499-508. [PMID: 22282676 PMCID: PMC3265222 DOI: 10.2147/ijn.s26670] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Solid tumors need new blood vessels to feed and nourish them as well as to allow tumor cells to escape into the circulation and lodge in other organs, which is termed "angiogenesis." Some tumor cells within solid tumors can overexpress integrins α(v)β(3) and α(v)β(5), which can specifically recognize the peptide motif Arg-Gly-Asp (RGD). Thus, the targeting of RGD-modified micelles to tumor vasculature is a promising strategy for tumor-targeting treatment. METHODS RGD peptide (GSSSGRGDSPA) was coupled to poly(ethylene glycol)-modified stearic acid-grafted chitosan (PEG-CS-SA) micelles via chemical reaction in the presence of N,N'-Disuccinimidyl carbonate. The critical micelle concentration of the polymeric micelles was determined by measuring the fluorescence intensity of pyrene as a fluorescent probe. The micelle size, size distribution, and zeta potential were measured by light scattering and electrophoretic mobility. Doxorubicin (DOX) was chosen as a model anticancer drug to investigate the drug entrapment efficiency, in vitro drug-release profile, and in vitro antitumor activities of drug-loaded RGD-PEG-CS-SA micelles in cells that overexpress integrins (α(ν)β(3) and α(ν)β(5)) and integrin-deficient cells. RESULTS Using DOX as a model drug, the drug encapsulation efficiency could reach 90%, and the in vitro drug-release profiles suggested that the micelles could be used as a controlled-release carrier for the hydrophobic drug. Qualitative and quantitative analysis of cellular uptake indicated that RGD-modified micelles could significantly increase the DOX concentration in integrin-overexpressing human hepatocellular carcinoma cell line (BEL-7402), but not in human epithelial carcinoma cell line (Hela). The competitive cellular-uptake test showed that the cellular uptake of RGD-modified micelles in BEL-7402 cells was significantly inhibited in the presence of excess free RGD peptides. In vitro cytotoxicity tests demonstrated DOX-loaded RGD-modified micelles could specifically enhance the cytotoxicity against BEL-7402 compared with DOX-loaded PEG-CS-SA and doxorubicin hydrochlorate. CONCLUSION This study suggests that RGD-modified PEG-CS-SA micelles are promising drug carriers for integrin-overexpressing tumor active targeting therapy.
Collapse
Affiliation(s)
- Li-Li Cai
- College of Pharmaceutical Science, Zhejiang University, Hangzhou, People’s Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Kim J, Kim PH, Kim SW, Yun CO. Enhancing the therapeutic efficacy of adenovirus in combination with biomaterials. Biomaterials 2011; 33:1838-50. [PMID: 22142769 DOI: 10.1016/j.biomaterials.2011.11.020] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Accepted: 11/10/2011] [Indexed: 12/18/2022]
Abstract
With the reason that systemically administered adenovirus (Ad) is rapidly extinguished by innate/adaptive immune responses and accumulation in liver, in vivo application of the Ad vector is strictly restricted. For achieving to develop successful Ad vector systems for cancer therapy, the chemical or physical modification of Ad vectors with polymers has been generally used as a promising strategy to overcome the obstacles. With polyethylene glycol (PEG) first in order, a variety of polymers have been developed to shield the surface of therapeutic Ad vectors and well accomplished to extend circulation time in blood and reduce liver toxicity. However, although polymer-coated Ads can successfully evacuate from a series of guarding systems in vivo and locate within tumors by enhanced permeability and retention (EPR) effect, the possibility to entering into the target cell is few and far between. To endow targeting moiety to polymer-coated Ad vectors, a diversity of ligands such as tumor-homing peptides, growth factors or antibodies, have been introduced with avoiding unwanted transduction and enhancing therapeutic efficacy. Here, we will describe and classify the characteristics of the published polymers with respect to Ad vectors. Furthermore, we will also compare the properties of variable targeting ligands, which are being utilized for addressing polymer-coated Ad vectors actively.
Collapse
Affiliation(s)
- Jaesung Kim
- Center for Controlled Chemical Delivery, Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, USA
| | | | | | | |
Collapse
|
48
|
Kim PH, Kim J, Kim TI, Nam HY, Yockman JW, Kim M, Kim SW, Yun CO. Bioreducible polymer-conjugated oncolytic adenovirus for hepatoma-specific therapy via systemic administration. Biomaterials 2011; 32:9328-42. [DOI: 10.1016/j.biomaterials.2011.08.066] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2011] [Accepted: 08/20/2011] [Indexed: 12/18/2022]
|
49
|
Retargeting of viruses to generate oncolytic agents. Adv Virol 2011; 2012:798526. [PMID: 22312365 PMCID: PMC3265223 DOI: 10.1155/2012/798526] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 08/25/2011] [Accepted: 08/26/2011] [Indexed: 12/16/2022] Open
Abstract
Oncolytic virus therapy is based on the ability of viruses to effectively infect and kill tumor cells without destroying the normal tissues. While some viruses seem to have a natural preference for tumor cells, most viruses require the modification of their tropism to specifically enter and replicate in such cells. This review aims to describe the transductional targeting strategies currently employed to specifically redirect viruses towards surface receptors on tumor cells. Three major strategies can be distinguished; they involve (i) the incorporation of new targeting specificity into a viral surface protein, (ii) the incorporation of a scaffold into a viral surface protein to allow the attachment of targeting moieties, and (iii) the use of bispecific adapters to mediate targeting of a virus to a specified moiety on a tumor cell. Of each strategy key features, advantages and limitations are discussed and examples are given. Because of their potential to cause sustained, multiround infection—a desirable characteristic for eradicating tumors—particular attention is given to viruses engineered to become self-targeted by the genomic expression of a bispecific adapter protein.
Collapse
|
50
|
Eto Y, Yoshioka Y, Ishida T, Yao X, Morishige T, Narimatsu S, Mizuguchi H, Mukai Y, Okada N, Kiwada H, Nakagawa S. Optimized PEGylated adenovirus vector reduces the anti-vector humoral immune response against adenovirus and induces a therapeutic effect against metastatic lung cancer. Biol Pharm Bull 2011; 33:1540-4. [PMID: 20823571 DOI: 10.1248/bpb.33.1540] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Application of adenovirus vectors (Adv) in metastatic cancer treatment is limited. We previously demonstrated that covalent conjugation of polyethleneglycol (PEG) to Adv enhances therapeutic effects and decreases toxic side-effects after systemic administration, but the level of immune response to PEGylated Adv (PEG-Ad) was not examined. Here, we examined the effect of PEGylation of Adv on the production of anti-Adv antibodies and antitumor response. We constructed a set of PEG-Ad using 5-kDa PEG, with modification rates of 30%, 45% and 90%. After systemic administration of Advs to rats, we examined the level of anti-Adv immunoglobulin (Ig)G and IgM in serum. The levels of anti-Adv IgG and anti-Adv IgM in rats treated with unmodified Adv were higher than those in control group. Rats treated with PEG-Ad that had a 90% modification rate showed lower level of anti-Adv IgG and anti-Adv IgM than those treated with unmodified Adv, whereas rats treated with PEG-Ad that had a 30% or 45% modification rate showed a similar level of anti-Adv IgG and IgM to those treated with unmodified Adv. Systemic administration of PEG-Ad that had a 90% modification rate, and expressed tumor necrosis factor-alpha, significantly reduced the number of metastatic colonies in the lung compared to unmodified Adv, with negligible side effects. These results suggest that systemic administration of PEG-Ad with an appropriate PEG modification rate has the potential to reduce the production of antibodies against Adv and increase the therapeutic response against metastatic cancer.
Collapse
Affiliation(s)
- Yusuke Eto
- Department of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|