1
|
Wang M, Yu F, Zhang Y. Present and future of cancer nano-immunotherapy: opportunities, obstacles and challenges. Mol Cancer 2025; 24:26. [PMID: 39827147 PMCID: PMC11748575 DOI: 10.1186/s12943-024-02214-5] [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: 09/24/2024] [Accepted: 12/25/2024] [Indexed: 01/22/2025] Open
Abstract
Clinically, multimodal therapies are adopted worldwide for the management of cancer, which continues to be a leading cause of death. In recent years, immunotherapy has firmly established itself as a new paradigm in cancer care that activates the body's immune defense to cope with cancer. Immunotherapy has resulted in significant breakthroughs in the treatment of stubborn tumors, dramatically improving the clinical outcome of cancer patients. Multiple forms of cancer immunotherapy, including immune checkpoint inhibitors (ICIs), adoptive cell therapy and cancer vaccines, have become widely available. However, the effectiveness of these immunotherapies is not much satisfying. Many cancer patients do not respond to immunotherapy, and disease recurrence appears to be unavoidable because of the rapidly evolving resistance. Moreover, immunotherapies can give rise to severe off-target immune-related adverse events. Strategies to remove these hindrances mainly focus on the development of combinatorial therapies or the exploitation of novel immunotherapeutic mediations. Nanomaterials carrying anticancer agents to the target site are considered as practical approaches for cancer treatment. Nanomedicine combined with immunotherapies offers the possibility to potentiate systemic antitumor immunity and to facilitate selective cytotoxicity against cancer cells in an effective and safe manner. A myriad of nano-enabled cancer immunotherapies are currently under clinical investigation. Owing to gaps between preclinical and clinical studies, nano-immunotherapy faces multiple challenges, including the biosafety of nanomaterials and clinical trial design. In this review, we provide an overview of cancer immunotherapy and summarize the evidence indicating how nanomedicine-based approaches increase the efficacy of immunotherapies. We also discuss the key challenges that have emerged in the era of nanotechnology-based cancer immunotherapy. Taken together, combination nano-immunotherapy is drawing increasing attention, and it is anticipated that the combined treatment will achieve the desired success in clinical cancer therapy.
Collapse
Affiliation(s)
- Man Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, 38 Dengzhou Road, Qingdao, 266021, China.
| | - Fei Yu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, 38 Dengzhou Road, Qingdao, 266021, China
| | - Yuan Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, 38 Dengzhou Road, Qingdao, 266021, China
| |
Collapse
|
2
|
Dong C, Tan D, Sun H, Li Z, Zhang L, Zheng Y, Liu S, Zhang Y, He Q. Interleukin-12 Delivery Strategies and Advances in Tumor Immunotherapy. Curr Issues Mol Biol 2024; 46:11548-11579. [PMID: 39451566 PMCID: PMC11506767 DOI: 10.3390/cimb46100686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/11/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
Interleukin-12 (IL-12) is considered to be a promising cytokine for enhancing an antitumor immune response; however, recombinant IL-12 has shown significant toxicity and limited efficacy in early clinical trials. Recently, many strategies for delivering IL-12 to tumor tissues have been developed, such as modifying IL-12, utilizing viral vectors, non-viral vectors, and cellular vectors. Previous studies have found that the fusion of IL-12 with extracellular matrix proteins, collagen, and immune factors is a way to enhance its therapeutic potential. In addition, studies have demonstrated that viral vectors are a good platform, and a variety of viruses such as oncolytic viruses, adenoviruses, and poxviruses have been used to deliver IL-12-with testing previously conducted in various cancer models. The local expression of IL-12 in tumors based on viral delivery avoids systemic toxicity while inducing effective antitumor immunity and acting synergistically with other therapies without compromising safety. In addition, lipid nanoparticles are currently considered to be the most mature drug delivery system. Moreover, cells are also considered to be drug carriers because they can effectively deliver therapeutic substances to tumors. In this article, we will systematically discuss the anti-tumor effects of IL-12 on its own or in combination with other therapies based on different delivery strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Qing He
- State Key Laboratory of Drug Regulatory Sciences, National Institutes for Food and Drug Control, Beijing 102629, China; (C.D.); (D.T.); (H.S.); (Z.L.); (L.Z.); (Y.Z.); (S.L.); (Y.Z.)
| |
Collapse
|
3
|
Chen P, Paraiso WKD, Cabral H. Revitalizing Cytokine-Based Cancer Immunotherapy through Advanced Delivery Systems. Macromol Biosci 2023; 23:e2300275. [PMID: 37565723 DOI: 10.1002/mabi.202300275] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/04/2023] [Indexed: 08/12/2023]
Abstract
Cytokines can coordinate robust immune responses, holding great promise as therapeutics against infections, autoimmune diseases, and cancers. In cancer treatment, numerous pro-inflammatory cytokines have displayed promising efficacy in preclinical studies. However, their clinical application is hindered by poor pharmacokinetics, significant toxicity and unsatisfactory anticancer efficacy. Thus, while IFN-α and IL-2 are approved for specific cancer treatments, other cytokines still remain subject of intense investigation. To accelerate the application of cytokines as cancer immunotherapeutics, strategies need to be directed to improve their safety and anticancer performance. In this regard, delivery systems could be used to generate innovative therapies by targeting the cytokines or nucleic acids, such as DNA and mRNA, encoding the cytokines to tumor tissues. This review centers on these innovative delivery strategies for cytokines, summarizing key approaches, such as gene delivery and protein delivery, and critically examining their potential and challenges for clinical translation.
Collapse
Affiliation(s)
- Pengwen Chen
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | | | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| |
Collapse
|
4
|
Tran TAT, Kim YH, Kim GE, Jung S, Kim IY, Moon KS, Kim YJ, Lee TK, Yun H, Lee JJ, Lee HJ, Lee CW, Jung TY. The long multi-epitope peptide vaccine combined with adjuvants improved the therapeutic effects in a glioblastoma mouse model. Front Immunol 2022; 13:1007285. [DOI: 10.3389/fimmu.2022.1007285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 10/04/2022] [Indexed: 11/10/2022] Open
Abstract
Emerging data have suggested that single short peptides have limited success as a cancer vaccine; however, extending the short peptides into longer multi-epitope peptides overcame the immune tolerance and induced an immune response. Moreover, the combination of adjuvants such as lenalidomide and anti-programmed cell death protein 1 (PD1) with a peptide vaccine showed potential vaccine effects in previous studies. Therefore, the effects of a long multi-epitope peptide vaccine in combination with lenalidomide and anti-PD1 were analyzed in this study. Long multi-epitope peptides from two MHCI peptides (BIRC597-104 and EphA2682-689) and the pan-human leukocyte antigen-DR isotype (HLA-DR) binding epitope (PADRE) were synthesized. The therapeutic effects of long multi-epitope peptides in combination with lenalidomide and anti-PD1 were confirmed in the murine GL261 intracranial glioma model. Immune cells’ distribution and responses to the long multi-epitope peptides in combination with these adjuvants were also estimated in the spleens, lymph nodes, and tumor tissues. The difference between long multi-epitope peptides and a cocktail of multi-epitope peptides combined with lenalidomide and anti-PD1 was also clarified. As a result, long multi-epitope peptides combined with lenalidomide and anti-PD1 prolonged the survival of mice according to the suppression of tumor growth in an intracranial mouse model. While long multi-epitope peptides combined with these adjuvants enhanced the percentages of activated and memory effector CD8+ T cells, the increase in percentages of regulatory T cells (Tregs) was observed in a cocktail of multi-epitope peptides combined with lenalidomide and anti-PD1 group in the tumors. Long multi-epitope peptides combined with these adjuvants also enhanced the function of immune cells according to the enhanced pro-inflammatory cytokines and cytotoxicity against GL261 cells in ex vivo. In conclusion, long multi-epitope peptides composed of MHCI peptides, BIRC5 and EphA2, and the MHCII peptide, PADRE, in combination with lenalidomide and anti-PD1 has the potential to improve the therapeutic effects of a vaccine against GBM.
Collapse
|
5
|
Cao TQ, Wainwright DA, Lee-Chang C, Miska J, Sonabend AM, Heimberger AB, Lukas RV. Next Steps for Immunotherapy in Glioblastoma. Cancers (Basel) 2022; 14:4023. [PMID: 36011015 PMCID: PMC9406905 DOI: 10.3390/cancers14164023] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 08/12/2022] [Accepted: 08/17/2022] [Indexed: 11/16/2022] Open
Abstract
Outcomes for glioblastoma (GBM) patients undergoing standard of care treatment remain poor. Here we discuss the portfolio of previously investigated immunotherapies for glioblastoma, including vaccine therapy and checkpoint inhibitors, as well as novel emerging therapeutic approaches. In addition, we explore the factors that potentially influence response to immunotherapy, which should be considered in future research aimed at improving immunotherapy efficacy.
Collapse
Affiliation(s)
- Toni Q. Cao
- Department of Neurology, Northwestern University, Chicago, IL 60611, USA
| | - Derek A. Wainwright
- Department of Neurological Surgery, Northwestern University, Chicago, IL 60611, USA
- Lou & Jean Malnati Brain Tumor Institute, Chicago, IL 60611, USA
- Department of Medicine, Division of Hematology/Oncology, Northwestern University, Chicago, IL 60611, USA
- Department of Neuroscience, Northwestern University, Chicago, IL 60611, USA
- Department of Microbiology-Immunology, Northwestern University, Chicago, IL 60611, USA
| | - Catalina Lee-Chang
- Department of Neurological Surgery, Northwestern University, Chicago, IL 60611, USA
- Lou & Jean Malnati Brain Tumor Institute, Chicago, IL 60611, USA
| | - Jason Miska
- Department of Neurological Surgery, Northwestern University, Chicago, IL 60611, USA
- Lou & Jean Malnati Brain Tumor Institute, Chicago, IL 60611, USA
| | - Adam M. Sonabend
- Department of Neurological Surgery, Northwestern University, Chicago, IL 60611, USA
- Lou & Jean Malnati Brain Tumor Institute, Chicago, IL 60611, USA
| | - Amy B. Heimberger
- Department of Neurological Surgery, Northwestern University, Chicago, IL 60611, USA
- Lou & Jean Malnati Brain Tumor Institute, Chicago, IL 60611, USA
| | - Rimas V. Lukas
- Department of Neurology, Northwestern University, Chicago, IL 60611, USA
- Lou & Jean Malnati Brain Tumor Institute, Chicago, IL 60611, USA
| |
Collapse
|
6
|
Brain Cancer Treatment; A Systematic Review. INTERNATIONAL JOURNAL OF CANCER MANAGEMENT 2022. [DOI: 10.5812/ijcm-121473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Context: One of the most common aggressive and primary brain tumors is glioma. The majority of diagnoses are referred to high-grade malignant glioblastoma, which carries the worst prognosis. Still, treatment of brain tumors remains a big challenge for clinicians. This study was designed to investigate the efficacy of gene therapy in the treatment of brain cancer. Methods: Studies use genes as a therapeutic agent in brain cancer treatment even alone or in combination with other treatment methods. Full-text papers, which met the inclusion criteria, were independently assessed by two reviewers. Disagreements were resolved by consultation with a third reviewer. Results: Statistical analysis showed that 50% of the papers used a virus, 36% used polymers, and 14% used cells as carriers to transfect the genes as a therapeutic agent in brain tumor models. Data showed that the estimated size of the brain tumor was reduced by using co-treatment of the gene with one of the conventional therapies. Conclusions: According to the results, co-treatment of the gene with conventional therapies could be more effective than the monotherapy methods.
Collapse
|
7
|
Hasbum A, Quintanilla J, Jr JA, Ding MH, Levy A, Chew SA. Strategies to better treat glioblastoma: antiangiogenic agents and endothelial cell targeting agents. Future Med Chem 2021; 13:393-418. [PMID: 33399488 PMCID: PMC7888526 DOI: 10.4155/fmc-2020-0289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 11/26/2020] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most prevalent and aggressive form of glioma, with poor prognosis and high mortality rates. As GBM is a highly vascularized cancer, antiangiogenic therapies to halt or minimize the rate of tumor growth are critical to improving treatment. In this review, antiangiogenic therapies, including small-molecule drugs, nucleic acids and proteins and peptides, are discussed. The authors further explore biomaterials that have been utilized to increase the bioavailability and bioactivity of antiangiogenic factors for better antitumor responses in GBM. Finally, the authors summarize the current status of biomaterial-based targeting moieties that target endothelial cells in GBM to more efficiently deliver therapeutics to these cells and avoid off-target cell or organ side effects.
Collapse
Affiliation(s)
- Asbiel Hasbum
- School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX 78541, USA
| | - Jaqueline Quintanilla
- Department of Health & Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78526, USA
| | - Juan A Amieva Jr
- Department of Health & Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78526, USA
| | - May-Hui Ding
- Department of Health & Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78526, USA
| | - Arkene Levy
- Dr Kiran C Patel College of Allopathic Medicine, Nova Southeastern University, FL 33314, USA
| | - Sue Anne Chew
- Department of Health & Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78526, USA
| |
Collapse
|
8
|
Nguyen KG, Vrabel MR, Mantooth SM, Hopkins JJ, Wagner ES, Gabaldon TA, Zaharoff DA. Localized Interleukin-12 for Cancer Immunotherapy. Front Immunol 2020; 11:575597. [PMID: 33178203 PMCID: PMC7593768 DOI: 10.3389/fimmu.2020.575597] [Citation(s) in RCA: 268] [Impact Index Per Article: 53.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 09/08/2020] [Indexed: 12/30/2022] Open
Abstract
Interleukin-12 (IL-12) is a potent, pro-inflammatory type 1 cytokine that has long been studied as a potential immunotherapy for cancer. Unfortunately, IL-12's remarkable antitumor efficacy in preclinical models has yet to be replicated in humans. Early clinical trials in the mid-1990's showed that systemic delivery of IL-12 incurred dose-limiting toxicities. Nevertheless, IL-12's pleiotropic activity, i.e., its ability to engage multiple effector mechanisms and reverse tumor-induced immunosuppression, continues to entice cancer researchers. The development of strategies which maximize IL-12 delivery to the tumor microenvironment while minimizing systemic exposure are of increasing interest. Diverse IL-12 delivery systems, from immunocytokine fusions to polymeric nanoparticles, have demonstrated robust antitumor immunity with reduced adverse events in preclinical studies. Several localized IL-12 delivery approaches have recently reached the clinical stage with several more at the precipice of translation. Taken together, localized delivery systems are supporting an IL-12 renaissance which may finally allow this potent cytokine to fulfill its considerable clinical potential. This review begins with a brief historical account of cytokine monotherapies and describes how IL-12 went from promising new cure to ostracized black sheep following multiple on-study deaths. The bulk of this comprehensive review focuses on developments in diverse localized delivery strategies for IL-12-based cancer immunotherapies. Advantages and limitations of different delivery technologies are highlighted. Finally, perspectives on how IL-12-based immunotherapies may be utilized for widespread clinical application in the very near future are offered.
Collapse
Affiliation(s)
- Khue G Nguyen
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, NC, United States
| | - Maura R Vrabel
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, NC, United States
| | - Siena M Mantooth
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, NC, United States
| | - Jared J Hopkins
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, NC, United States
| | - Ethan S Wagner
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, NC, United States
| | - Taylor A Gabaldon
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, NC, United States
| | - David A Zaharoff
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, NC, United States
| |
Collapse
|
9
|
Tran TAT, Kim YH, Duong THO, Jung S, Kim IY, Moon KS, Jang WY, Lee HJ, Lee JJ, Jung TY. Peptide Vaccine Combined Adjuvants Modulate Anti-tumor Effects of Radiation in Glioblastoma Mouse Model. Front Immunol 2020; 11:1165. [PMID: 32733437 PMCID: PMC7358653 DOI: 10.3389/fimmu.2020.01165] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 05/12/2020] [Indexed: 11/13/2022] Open
Abstract
Glioblastoma, the most common aggressive cancer, has a poor prognosis. Among the current standard treatment strategies, radiation therapy is the most commonly recommended. However, it is often unsuccessful at completely eliminating the cancer from the brain. A combination of radiation with other treatment methods should therefore be considered. It has been reported that radiotherapy in combination with immunotherapy might show a synergistic effect; however, this still needs to be investigated. In the current study, a “branched multipeptide and peptide adjuvants [such as pan DR epitope (PADRE) and polyinosinic-polycytidylic acid—stabilized with polylysine and carboxymethylcellulose—(poly-ICLC)],” namely vaccine and anti-PD1, were used as components of immunotherapy to assist in the anti-tumor effects of radiotherapy against glioblastomas. With regard to experimental design, immunological characterization of GL261 cells was performed and the effects of radiation on this cell line were also evaluated. An intracranial GL261 mouse glioma model was established, and therapeutic effects were observed based on tumor size and survival time. The distribution of effector immune cells in the spleen, based on cytotoxic T lymphocyte (CTL) and natural killer (NK) cell function, was determined. The pro-inflammatory and anti-inflammatory cytokine production from re-stimulated splenocytes and single tumor cells were also evaluated. As GL261 cells demonstrated both immunological characteristics and radiation sensitivity, they were found to be promising candidates for testing this combination treatment. Combinatorial treatment with radiation, vaccine, and anti-PD1 prolonged mouse survival by delaying tumor growth. Although this combination treatment led to an increase in the functional activity of both CTLs and NK cells, as evidenced by the increased percentage of these cells in the spleen, there was a greater shift toward CTL rather than NK cell activity. Moreover, the released cytokines from re-stimulated splenocytes and single tumor cells also showed a shift toward the pro-inflammatory response. This study suggests that immunotherapy comprising a branched multipeptide plus PADRE, poly-ICLC, and anti-PD1 could potentially enhance the anti-tumor effects of radiotherapy in a glioblastoma mouse model.
Collapse
Affiliation(s)
- Thi-Anh-Thuy Tran
- Brain Tumor Research Laboratory, Chonnam National University Medical School and Hwasun Hospital, Chonnam National University, Hwasun, South Korea
| | - Young-Hee Kim
- Brain Tumor Research Laboratory, Chonnam National University Medical School and Hwasun Hospital, Chonnam National University, Hwasun, South Korea
| | - Thi-Hoang-Oanh Duong
- Brain Tumor Research Laboratory, Chonnam National University Medical School and Hwasun Hospital, Chonnam National University, Hwasun, South Korea
| | - Shin Jung
- Brain Tumor Research Laboratory, Chonnam National University Medical School and Hwasun Hospital, Chonnam National University, Hwasun, South Korea.,Department of Neurosurgery, Chonnam National University Medical School and Hwasun Hospital, Chonnam National University, Hwasun, South Korea
| | - In-Young Kim
- Brain Tumor Research Laboratory, Chonnam National University Medical School and Hwasun Hospital, Chonnam National University, Hwasun, South Korea.,Department of Neurosurgery, Chonnam National University Medical School and Hwasun Hospital, Chonnam National University, Hwasun, South Korea
| | - Kyung-Sub Moon
- Brain Tumor Research Laboratory, Chonnam National University Medical School and Hwasun Hospital, Chonnam National University, Hwasun, South Korea.,Department of Neurosurgery, Chonnam National University Medical School and Hwasun Hospital, Chonnam National University, Hwasun, South Korea
| | - Woo-Youl Jang
- Brain Tumor Research Laboratory, Chonnam National University Medical School and Hwasun Hospital, Chonnam National University, Hwasun, South Korea.,Department of Neurosurgery, Chonnam National University Medical School and Hwasun Hospital, Chonnam National University, Hwasun, South Korea
| | - Hyun-Ju Lee
- Research Center for Cancer Immunotherapy, Chonnam National University Medical School and Hwasun Hospital, Chonnam National University, Hwasun, South Korea
| | - Je-Jung Lee
- Research Center for Cancer Immunotherapy, Chonnam National University Medical School and Hwasun Hospital, Chonnam National University, Hwasun, South Korea.,Department of Internal Medicine, Chonnam National University Medical School and Hwasun Hospital, Chonnam National University, Hwasun, South Korea
| | - Tae-Young Jung
- Brain Tumor Research Laboratory, Chonnam National University Medical School and Hwasun Hospital, Chonnam National University, Hwasun, South Korea.,Department of Neurosurgery, Chonnam National University Medical School and Hwasun Hospital, Chonnam National University, Hwasun, South Korea
| |
Collapse
|
10
|
Clavreul A, Pourbaghi-Masouleh M, Roger E, Menei P. Nanocarriers and nonviral methods for delivering antiangiogenic factors for glioblastoma therapy: the story so far. Int J Nanomedicine 2019; 14:2497-2513. [PMID: 31040671 PMCID: PMC6461002 DOI: 10.2147/ijn.s194858] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Angiogenesis, the formation of new blood vessels, is an essential component of glioblastoma (GB) progression. The development of angiogenesis inhibitor therapy, including treatments targeting vascular endothelial growth factor (VEGF) in particular, raised new hopes for the treatment of GB, but no Phase III clinical trial to date has reported survival benefits relative to standard treatment. There are several possible reasons for this limited efficacy, including VEGF-independent angiogenesis, induction of tumor invasion, and inefficient antiangiogenic factor delivery to the tumor. Efforts have been made to overcome these limitations by identifying new angiogenesis inhibitors that target angiogenesis through different mechanisms of action without inducing tumor invasion, and through the development of viral and nonviral delivery methods to improve antiangiogenic activity. Herein, we describe the nonviral methods, including convection-enhanced delivery devices, implantable polymer devices, nanocarriers, and cellular vehicles, to deliver antiangiogenic factors. We focus on those evaluated in intracranial (orthotopic) animal models of GB, the most relevant models of this disease, as they reproduce the clinical scenario of tumor progression and therapy response encountered in GB patients.
Collapse
Affiliation(s)
- Anne Clavreul
- Department of Neurosurgery, CHU, Angers, France, .,CRCINA, INSERM, University of Nantes, University of Angers, Angers, France,
| | - Milad Pourbaghi-Masouleh
- CRCINA, INSERM, University of Nantes, University of Angers, Angers, France, .,Division of Drug Delivery and Tissue Engineering, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Emilie Roger
- MINT, INSERM 1066, CNRS 6021, University of Angers, Angers, France
| | - Philippe Menei
- Department of Neurosurgery, CHU, Angers, France, .,CRCINA, INSERM, University of Nantes, University of Angers, Angers, France,
| |
Collapse
|
11
|
Li S, Feng X, Wang J, He L, Wang C, Ding J, Chen X. Polymer nanoparticles as adjuvants in cancer immunotherapy. NANO RESEARCH 2018; 11:5769-5786. [DOI: 10.1007/s12274-018-2124-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 06/03/2018] [Accepted: 06/06/2018] [Indexed: 10/14/2024]
|
12
|
H1/pHGFK1 nanoparticles exert anti-tumoural and radiosensitising effects by inhibition of MET in glioblastoma. Br J Cancer 2018; 118:522-533. [PMID: 29348487 PMCID: PMC5830599 DOI: 10.1038/bjc.2017.461] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Revised: 11/23/2017] [Accepted: 11/23/2017] [Indexed: 12/20/2022] Open
Abstract
Background: The therapeutic resistance to ionising radiation (IR) and anti-angiogenesis mainly impair the prognosis of patients with glioblastoma. The primary and secondary MET aberrant activation is one crucial factor for these resistances. The kringle 1 domain of hepatocyte growth factor (HGFK1), an angiogenic inhibitor, contains a high-affinity binding domain of MET; however, its effects on glioblastoma remain elusive. Methods: We formed the nanoparticles consisting of a folate receptor-targeted nanoparticle-mediated HGFK1 gene (H1/pHGFK1) and studied its anti-tumoural and radiosensitive activities in both subcutaneous and orthotopic human glioma cell-xenografted mouse models. We then elucidated its molecular mechanisms in human glioblastoma cell lines in vitro. Results: We demonstrated for the first time that peritumoural injection of H1/pHGFK1 nanoparticles significantly inhibited tumour growth and prolonged survival in tumour-bearing mice, as well as enhanced the anti-tumoural efficacies of IR in vivo by reducing Ki-67 expression, enhancing TUNEL staining-indicated apoptotic indexes, reducing microvascular intensity and reversing IR-induced MET overexpression in tumour tissues. Furthermore, we showed that HGFK1 suppressed the proliferation and induced cell apoptosis and enhanced sensitivity to IR in glioblastoma cell lines, mainly by suppressing the activities of MET receptor, down-regulating ATM-Chk2 axis but up-regulating Chk1. Conclusions: H1/pHGFK1 exerts anti-tumoural and radiosensitive activities mainly through the inhibition and reversal of IR-induced MET and ATM–Chk2 axis activities in glioblastoma. H1/pHGFK1 nanoparticles are a potential radiosensitiser and angiogenic inhibitor for glioblastoma treatment.
Collapse
|
13
|
Dendritic cell immunotherapy for brain tumors. J Neurooncol 2015; 123:425-32. [PMID: 26037466 DOI: 10.1007/s11060-015-1830-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 05/25/2015] [Indexed: 12/15/2022]
Abstract
Glioblastomas are characterized by immunosuppression, rapid proliferation, angiogenesis, and invasion into the surrounding brain parenchyma. Limitations in current therapeutic approaches have spurred the development of personalized, patient-specific treatments. Among these, active immunotherapy has emerged as a viable option for glioma treatment. The ability to generate an immune response utilizing patient-derived dendritic cells (DCs) (professional antigen-presenting cells) is especially attractive. This approach to glioma treatment allows for the immunologic targeting and destruction of malignant cells. Data acquired in multiple pre-clinical models and clinical trials have shown significant responses and prolonged survival. Here we provide an overview of the current status of DC vaccination for the treatment of gliomas.
Collapse
|
14
|
New insights into IL-12-mediated tumor suppression. Cell Death Differ 2014; 22:237-46. [PMID: 25190142 DOI: 10.1038/cdd.2014.134] [Citation(s) in RCA: 396] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2014] [Revised: 07/17/2014] [Accepted: 07/21/2014] [Indexed: 12/14/2022] Open
Abstract
During the past two decades, interleukin-12 (IL-12) has emerged as one of the most potent cytokines in mediating antitumor activity in a variety of preclinical models. Through pleiotropic effects on different immune cells that form the tumor microenvironment, IL-12 establishes a link between innate and adaptive immunity that involves different immune effector cells and cytokines depending on the type of tumor or the affected tissue. The robust antitumor response exerted by IL-12, however, has not yet been successfully translated into the clinics. The majority of clinical trials involving treatment with IL-12 failed to show sustained antitumor responses and were associated to toxic side effects. Here we discuss the therapeutic effects of IL-12 from preclinical to clinical studies, and will highlight promising strategies to take advantage of the antitumor activity of IL-12 while limiting adverse effects.
Collapse
|
15
|
Dehshahri A, Alhashemi SH, Jamshidzadeh A, Sabahi Z, Samani SM, Sadeghpour H, Mohazabieh E, Fadaei M. Comparison of the effectiveness of polyethylenimine, polyamidoamine and chitosan in transferring plasmid encoding interleukin-12 gene into hepatocytes. Macromol Res 2013. [DOI: 10.1007/s13233-013-1180-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
16
|
Anwer K, Kelly FJ, Chu C, Fewell JG, Lewis D, Alvarez RD. Phase I trial of a formulated IL-12 plasmid in combination with carboplatin and docetaxel chemotherapy in the treatment of platinum-sensitive recurrent ovarian cancer. Gynecol Oncol 2013; 131:169-73. [PMID: 23863356 DOI: 10.1016/j.ygyno.2013.07.081] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 07/01/2013] [Accepted: 07/07/2013] [Indexed: 01/01/2023]
Abstract
OBJECTIVES The primary objective of this study was to evaluate the safety and tolerability of a formulated IL-12 plasmid administered intraperitoneally (IP) in conjunction with intravenous (IV) carboplatin/docetaxel in platinum-sensitive ovarian cancer patients. METHODS Escalating doses of IL-12 plasmid (phIL-12) formulated with the lipopolymer PEG-PEI-Cholesterol (PPC) were administered IP every 10-11 days for a total of four treatments and the highest dose was expanded to eight treatments. Patients also received IV carboplatin (AUC 5) and docetaxel (75 mg/m(2)) every 21 days. Patients were followed for safety, biological activity and antitumor activity after phIL-12/PPC treatment. RESULTS All 13 patients enrolled in the study received both phIL-12/PPC and chemotherapy treatment. There were 49 plasmid-associated adverse events (AEs). The most common AEs were abdominal pain, transient hypotension, low grade fever, catheter site pain, chills, dysgeusia, infusion-related reaction, and nausea. These AEs appeared to be plasmid dose related. Grade 3 AEs included manageable abdominal pain and cytokine release syndrome. There were no dose limiting toxicities and the plasmid treatment did not augment the chemotherapy-associated AEs. The best overall antitumor response (17% CR, 33% PR, 42% SD and 8% PD) was typical of the patient population enrolled for the study. Translational studies showed rise in IFN-γ and TNF-α concentrations in a dose dependent manner. CONCLUSIONS The escalating doses and cycles of intraperitoneal phIL-12/PPC when combined with carboplatin/docetaxel chemotherapy in recurrent ovarian cancer patients were well tolerated and did not appear to exacerbate the side effects or attenuate the efficacy of the chemotherapy treatment.
Collapse
|
17
|
Anti-PD-1 blockade and stereotactic radiation produce long-term survival in mice with intracranial gliomas. Int J Radiat Oncol Biol Phys 2013; 86:343-9. [PMID: 23462419 DOI: 10.1016/j.ijrobp.2012.12.025] [Citation(s) in RCA: 696] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Revised: 12/20/2012] [Accepted: 12/23/2012] [Indexed: 02/08/2023]
Abstract
PURPOSE Glioblastoma multiforme (GBM) is the most common primary brain tumor in adults, and radiation is one of the main treatment modalities. However, cure rates remain low despite best available therapies. Immunotherapy is a promising modality that could work synergistically with radiation, which has been shown to increase antigen presentation and promote a proinflammatory tumor microenvironment. Programmed-death-1 (PD-1) is a surface receptor expressed on activated and exhausted T cells, which mediate T cell inhibition upon binding with its ligand PD-L1, expressed on many tumor types including human GBMs. We tested the combination of anti-PD-1 immunotherapy with stereotactic radiosurgery in a mouse orthotopic GBM model. METHODS AND MATERIALS We performed intracranial implantation of mouse glioma cell line GL261 transfected with luciferase into C57BL/6 mice. Mice were stratified into 4 treatment groups: (1) control; (2) radiation only; (3) anti-PD-1 antibody only; and (4) radiation plus anti-PD-1 antibody. Overall survival was quantified. The mice were killed on day 21 after implantation to assess immunologic parameters in the brain/tumor, cervical lymph nodes, and spleen. RESULTS Improved survival was demonstrated with combination anti-PD-1 therapy plus radiation compared with either modality alone: median survival was 25 days in the control arm, 27 days in the anti-PD-1 antibody arm, 28 days in the radiation arm, and 53 days in the radiation plus anti-PD-1 therapy arm (P<.05 by log-rank Mantle-Cox). Long-term survival was seen only in the combined treatment arm, with a fraction (15%-40%) of animals alive at day 180+ after treatment. Immunologic data on day 21 after implantation showed increased tumor infiltration by cytotoxic T cells (CD8+/interferon-γ+/tumor necrosis factor-α+) and decreased regulatory T cells (CD4+/FOXP3) in the combined treatment group compared with the single modality arms. CONCLUSIONS The combination of PD-1 blockade and localized radiation therapy results in long-term survival in mice with orthotopic brain tumors. These studies provide strong preclinical evidence to support combination trials in patients with GBM.
Collapse
|
18
|
Abstract
INTRODUCTION Despite extensive research, cancer continues to be a leading cause of death worldwide and is expected to continue to rise as a result of an aging population. Therefore, new therapies are constantly being developed. Par-4 is a naturally occurring tumor suppressor protein that is capable of inducing apoptosis in cancer, but not normal cells. For this reason, Par-4 offers an attractive target for development of cancer therapy, particularly of difficult to treat cancers. AREAS COVERED The mechanisms by which Par-4 induces cell death are summarized. The ways that Par-4 is controlled in cancer cells are discussed. We discuss how different research groups have developed ways to overexpress and/or activate Par-4 in vitro and in vivo. The studies described demonstrate that when Par-4 levels and/or activity are increased, susceptibility to apoptosis is enhanced and tumor growth is inhibited. EXPERT OPINION Par-4 is a promising therapeutic protein that can be overexpressed and/or activated to induce apoptosis in a cancer-selective manner. This cancer selectivity is important given that the side-effects of chemotherapeutics can be as debilitating as cancer itself. However, there are key issues that need to be addressed to optimize the effects of Par-4 in patients.
Collapse
Affiliation(s)
- Rosalyn B Irby
- Penn State Hershey Cancer Institute, 500 University Drive, Hershey, PA 17033, USA.
| | | |
Collapse
|
19
|
Castro MG, Candolfi M, Kroeger K, King GD, Curtin JF, Yagiz K, Mineharu Y, Assi H, Wibowo M, Ghulam Muhammad AKM, Foulad D, Puntel M, Lowenstein PR. Gene therapy and targeted toxins for glioma. Curr Gene Ther 2011; 11:155-80. [PMID: 21453286 DOI: 10.2174/156652311795684722] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Accepted: 03/08/2011] [Indexed: 12/12/2022]
Abstract
The most common primary brain tumor in adults is glioblastoma. These tumors are highly invasive and aggressive with a mean survival time of 15-18 months from diagnosis to death. Current treatment modalities are unable to significantly prolong survival in patients diagnosed with glioblastoma. As such, glioma is an attractive target for developing novel therapeutic approaches utilizing gene therapy. This review will examine the available preclinical models for glioma including xenographs, syngeneic and genetic models. Several promising therapeutic targets are currently being pursued in pre-clinical investigations. These targets will be reviewed by mechanism of action, i.e., conditional cytotoxic, targeted toxins, oncolytic viruses, tumor suppressors/oncogenes, and immune stimulatory approaches. Preclinical gene therapy paradigms aim to determine which strategies will provide rapid tumor regression and long-term protection from recurrence. While a wide range of potential targets are being investigated preclinically, only the most efficacious are further transitioned into clinical trial paradigms. Clinical trials reported to date are summarized including results from conditionally cytotoxic, targeted toxins, oncolytic viruses and oncogene targeting approaches. Clinical trial results have not been as robust as preclinical models predicted; this could be due to the limitations of the GBM models employed. Once this is addressed, and we develop effective gene therapies in models that better replicate the clinical scenario, gene therapy will provide a powerful approach to treat and manage brain tumors.
Collapse
Affiliation(s)
- Maria G Castro
- Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Maes W, Van Gool SW. Experimental immunotherapy for malignant glioma: lessons from two decades of research in the GL261 model. Cancer Immunol Immunother 2011; 60:153-60. [PMID: 21120655 PMCID: PMC11028904 DOI: 10.1007/s00262-010-0946-6] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2010] [Accepted: 11/05/2010] [Indexed: 02/04/2023]
Abstract
Nearly twenty years of experimental immunotherapy for malignant glioma yielded important insights in the mechanisms governing glioma immunology. Still considered promising, it is clear that immunotherapy does not on its own represent the magic bullet in glioma therapy. In this review, we summarize the major immunotherapeutic achievements in the mouse GL261 glioma model, which has emerged as the gold standard syngeneic model for experimental glioma therapy. Gene therapy, monoclonal antibody treatment, cytokine therapy, cell transfer strategies and dendritic cell therapy were hereby considered. Apart from the considerable progress made in understanding glioma immunology in this model, we also addressed its most pertinent issues and shortcomings. Despite these, the GL261 model will remain indispensable in glioma research since it is a fast, highly reproducible and easy-to-establish model system.
Collapse
Affiliation(s)
- Wim Maes
- Laboratory of Experimental Immunology (WM and SWVG), Pediatric Hemato-oncology (SWVG), University Hospital Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Stefaan W. Van Gool
- Laboratory of Experimental Immunology (WM and SWVG), Pediatric Hemato-oncology (SWVG), University Hospital Leuven, Herestraat 49, 3000 Leuven, Belgium
| |
Collapse
|
21
|
Anwer K, Barnes MN, Fewell J, Lewis DH, Alvarez RD. Phase-I clinical trial of IL-12 plasmid/lipopolymer complexes for the treatment of recurrent ovarian cancer. Gene Ther 2009; 17:360-9. [PMID: 20033066 DOI: 10.1038/gt.2009.159] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
A phase-I trial to assess the safety and tolerability of human interleukin-12 (IL-12) plasmid (phIL-12) formulated with a synthetic lipopolymer, polyethyleneglycol-polyethyleneimine-cholesterol (PPC), was conducted on women with chemotherapy-resistant recurrent ovarian cancer. A total of 13 patients were enrolled in four dose-escalating cohorts and treated with 0.6, 3, 12 or 24 mg m(-2) of the formulated plasmid once every week for 4 weeks. Administration of phIL-12/PPC was generally safe and well-tolerated. Common side effects included low-grade fever and abdominal pain. Stable disease and reduction in serum CA-125 levels were clinically observed in some patients. Measurable levels of IL-12 plasmid were detectable in PF samples collected throughout the course of phIL-12/PPC treatment. In comparison, serum samples either did not contain detectable amounts of plasmid DNA or contained <1% of the amount found in the corresponding PF samples. Treatment-related increases in IFN-gamma levels were observed in PF but not in serum. These data demonstrate that IL-12 gene delivery with a synthetic delivery system is feasible for ovarian cancer patients.
Collapse
Affiliation(s)
- K Anwer
- EGEN Inc, Huntsville, AL 35806, USA
| | | | | | | | | |
Collapse
|
22
|
Intracerebral interleukin 12 induces glioma rejection in the brain predominantly by CD8+ T cells and independently of interferon-gamma. J Neuropathol Exp Neurol 2009; 68:525-34. [PMID: 19525900 DOI: 10.1097/nen.0b013e3181a2afa0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The prognosis of gliomas is generally poor since these tumors elude established therapeutic approaches. Immunotherapy might present an effective therapy in particular because the glioma cells are diffusely dispersed in the infiltration zone of the tumor and show a strong propensity to invade the surrounding brain along white matter tracts. Although various immune therapies for brain tumors are successful in rodents, there is currently no effective therapy in humans. In the present study, we investigated the mechanisms by which intracerebral IL-12 mediates rejection of GL261 cells in a syngenic mouse glioma model. Wild type mice revealed smaller tumors as compared to mice lacking functional T and B cells indicating that considerable immune dependent tumor rejection occurs physiologically in this model. However, glioma rejection was significantly enhanced in mice expressing IL-12 in the CNS and was predominantly dependent on the presence of CD8+ T cells while CD4+ T cells had less impact. Interestingly, the rejection of tumors was independent of IFN-gamma. Our findings contrast results obtained after in vitro or systemic stimulation with IL-12 and demonstrate that successful IL-12 induced glioma rejection critically depends on the localization, duration and time of IL-12 expression.
Collapse
|
23
|
Benny O, Pakneshan P. Novel technologies for antiangiogenic drug delivery in the brain. Cell Adh Migr 2009; 3:224-9. [PMID: 19262168 DOI: 10.4161/cam.3.2.7766] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Antiangiogenic therapies aimed at inhibiting the formation of tumor vasculature hold great promise for cancer therapy, with multiple compounds currently undergoing clinical trials. As with many forms of chemotherapy, antiangiogenic drugs face numerous hurdles in their translation to clinical use. Many such promising agents exhibit a short half-life, low solubility, poor bioavailability and multiple toxic side effects. Furthermore, when targeting malignant brain tumors the blood-brain barrier represents a formidable obstacle, preventing drugs from penetrating into the central nervous system (CNS). In this review, we discuss several preclinical antiangiogenic therapies and describe issues related to the unique conditions in the brain with regard to cancer treatment and neurotoxicity. We focus on the limitations of antiangiogenic drugs in the brain, along with numerous solutions that involve novel biomaterials and nanotechnological approaches. We also discuss an example in which modifying the properties of an antiangiogenic compound enhanced its clinical efficacy in treating tumors while simultaneously mitigating undesirable neurological side-effects.
Collapse
Affiliation(s)
- Ofra Benny
- Department of Surgery, Children's Hospital Boston, Harvard Medical School, MA, USA.
| | | |
Collapse
|
24
|
Li Z, Xiang J, Zhang W, Fan S, Wu M, Li X, Li G. Nanoparticle delivery of anti-metastatic NM23-H1 gene improves chemotherapy in a mouse tumor model. Cancer Gene Ther 2008; 16:423-9. [PMID: 19096443 DOI: 10.1038/cgt.2008.97] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Gene therapy provides a promising approach for cancer treatment. Earlier studies suggested that poly-L-lysine-modified iron oxide nanoparticles (IONP-PLL) might be a promising gene delivery system that can transfect DNA efficiently in vitro and in vivo. In this study we used IONP-PLL as gene carriers to deliver the NM23-H1 gene, the first suppressor gene of cancer metastasis, to tumor cells in vivo. The intravenous injection of IONP-PLL carrying NM23-H1-GFP plasmid DNA significantly extended the survival time of an experimental pulmonary metastasis mouse model. In the IONP-PLL/NM23-H1-GFP-treated group, metastasis was clearly suppressed compared with the group treated with free NM23-H1-GFP plasmid. Furthermore, this gene therapy combined with cyclophosphamide treatment resulted in longer survival times and greater suppression of metastasis growth. In conclusion, treatment with IONP-PLL nanoparticles incorporating the NM23-H1gene is an efficient gene therapy method, and it is even more effective in combination with chemotherapy. This approach appears to be a promising strategy for treatment of metastatic tumors.
Collapse
Affiliation(s)
- Z Li
- Cancer Research Institute, Central South University, Changsha, Hunan, The People's Republic of China
| | | | | | | | | | | | | |
Collapse
|