1
|
Kandasamy G, Maity D. Inorganic nanocarriers for siRNA delivery for cancer treatments. Biomed Mater 2024; 19:022001. [PMID: 38181441 DOI: 10.1088/1748-605x/ad1baf] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 01/05/2024] [Indexed: 01/07/2024]
Abstract
RNA interference is one of the emerging methodologies utilized in the treatment of a wide variety of diseases including cancer. This method specifically uses therapeutic RNAs (TpRNAs) like small interfering RNAs (siRNAs) to regulate/silence the cancer-linked genes, thereby minimizing the distinct activities of the cancer cells while aiding in their apoptosis. But, many complications arise during the transport/delivery of these TpRNAs that include poor systemic circulation, instability/degradation inside the body environment, no targeting capacity and also low cellular internalization. These difficulties can be overcome by using nanocarriers to deliver the TpRNAs inside the cancer cells. The following are the various categories of nanocarriers-viral vectors (e.g. lentivirus and adenovirus) and non-viral nanocarriers (self-assembling nanocarriers and inorganic nanocarriers). Viral vectors suffer from disadvantages like high immunogenicity compared to the non-viral nanocarriers. Among non-viral nanocarriers, inorganic nanocarriers gained significant attention as their inherent properties (like magnetic properties) can aid in the effective cellular delivery of the TpRNAs. Most of the prior reports have discussed about the delivery of TpRNAs through self-assembling nanocarriers; however very few have reviewed about their delivery using the inorganic nanoparticles. Therefore, in this review, we have mainly focussed on the delivery of TpRNAs-i.e. siRNA, especially programmed death ligand-1 (PD-L1), survivin, B-cell lymphoma-2 (Bcl-2), vascular endothelial growth factor and other siRNAs using the inorganic nanoparticles-mainly magnetic, metal and silica nanoparticles. Moreover, we have also discussed about the combined delivery of these TpRNAs along with chemotherapeutic drugs (mainly doxorubicin) andin vitroandin vivotherapeutic effectiveness.
Collapse
Affiliation(s)
- Ganeshlenin Kandasamy
- Department of Biomedical Engineering, School of Electrical and Communication, Vel Tech Rangarajan Dr. Sagunthala R&D Institute of Science and Technology, Avadi, Chennai, India
| | - Dipak Maity
- Department of Environmental and Occupational Health, School of Public Health, Texas A&M University, College Station, TX 77843, United States of America
| |
Collapse
|
2
|
de Santana WMOS, Surur AK, Momesso VM, Lopes PM, Santilli CV, Fontana CR. Nanocarriers for photodynamic-gene therapy. Photodiagnosis Photodyn Ther 2023; 43:103644. [PMID: 37270046 DOI: 10.1016/j.pdpdt.2023.103644] [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: 02/15/2023] [Revised: 05/30/2023] [Accepted: 05/31/2023] [Indexed: 06/05/2023]
Abstract
The use of nanotechnology in medicine has important potential applications, including in anticancer strategies. Nanomedicine has made it possible to overcome the limitations of conventional monotherapies, in addition to improving therapeutic results by means of synergistic or cumulative effects. A highlight is the combination of gene therapy (GT) and photodynamic therapy (PDT), which are alternative anticancer approaches that have attracted attention in the last decade. In this review, strategies involving the combination of PDT and GT will be discussed, together with the role of nanocarriers (nonviral vectors) in this synergistic therapeutic approach, including aspects related to the design of nanomaterials, responsiveness, the interaction of the nanomaterial with the biological environment, and anticancer performance in studies in vitro and in vivo.
Collapse
Affiliation(s)
| | - Amanda Koberstain Surur
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, São Paulo, 14800-903, Brazil
| | - Vinícius Medeiros Momesso
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, São Paulo, 14800-903, Brazil
| | - Pedro Monteiro Lopes
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, São Paulo, 14800-903, Brazil
| | - Celso V Santilli
- São Paulo State University (UNESP), Institute of Chemistry, Araraquara, São Paulo, 14800-900, Brazil
| | - Carla Raquel Fontana
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, São Paulo, 14800-903, Brazil.
| |
Collapse
|
3
|
Li Y, Zhang X, Xiang Z, Chen T, Hu Z, Yang K, Sun X, Wu Y, Wu J. Public Attitudes About the Use of Gene Therapy in Mainland China. JAMA Netw Open 2023; 6:e2328352. [PMID: 37566417 PMCID: PMC10422191 DOI: 10.1001/jamanetworkopen.2023.28352] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/30/2023] [Indexed: 08/12/2023] Open
Abstract
Importance In addition to technical barriers, public attitudes about the use of gene therapy have an important association with the clinical implementation of gene therapy. Objective To investigate the factors associated with public acceptance of gene therapy among individuals in China. Design, Setting, and Participants This cross-sectional study used data from a survey conducted among 21 880 individuals in mainland China from June 20 to August 31, 2022. Main Outcomes and Measures Stepwise linear regression was used to analyze factors associated with public acceptance of gene therapy in 5 key areas: basic personal information (gender, region, age, and educational level), family situation (marital status, children, and cousins), economic status (assets, debts, and insurance coverage), health knowledge (health literacy score and media use), and physical health status (chronic illness, cancer, European Quality of Life 5-Dimension 5-Level version [EQ-5D-5L] score, and Brief Illness Perception Questionnaire [BIPQ] score). Acceptance scores were calculated based on a visual analog scale (range, 0-100, with higher scores indicating higher acceptance of gene therapy). Further subgroup analysis was carried out in different age subgroups and populations with or without chronic diseases. Results A total of 21 880 participants (mean [SD] age, 39.4 [18.9] years; 10 947 female participants [50.0%]; 10 933 male participants [50.0%]) were analyzed in this study. The mean (SD) acceptance score of gene therapy in the survey was 60.56 (27.60). Compared with people aged 60 years or older, those aged 12 to 18 years had higher acceptance of gene therapy (β = 1.48 [95% CI, 0.09-2.88]), while groups aged 19 to 30 years (β = -3.43 [95% CI, -4.80 to -2.07]), 31 to 44 years (β = -1.44 [95% CI, -2.76 to -0.12]), and 45 to 59 years (β = -2.05 [95% CI, -3.27 to -0.83]) had lower acceptance. Compared with people living in Eastern China, those in Central China had lower acceptance of gene therapy (β = -1.58 [95% CI, -2.54 to -0.62]), while those in Western China had higher acceptance (β = 0.92 [95% CI, 0.09-1.76]). Higher educational level (undergraduate or above vs junior high or below) was associated with higher acceptance of gene therapy (β = 1.56 [95% CI, 0.49-2.63]). Number of properties owned was also associated with higher acceptance of gene therapy (2 vs 0: β = 2.38 [95% CI, 1.04-3.72]; ≥3 vs 0: β = 4.66 [95% CI, 2.92-6.39]). Diagnosis of chronic disease was associated with lower acceptance of gene therapy (β = -17.86 [95% CI, -20.49 to -15.24]), while diagnosis of cancer was associated with higher acceptance (β = 6.99 [95% CI, 1.84-12.14]). Higher BIPQ score (β = 0.40 [95% CI, 0.34-0.45]), higher health literacy score (β = 0.70 [95% CI, 0.62-0.78]), and media use (β = 0.49 [95% CI, 0.41-0.57]) were all associated with high acceptance of gene therapy, while a higher EQ-5D-5L score was associated with lower acceptance (β = -0.29 [95% CI, -0.47 to -0.11]). For older people, being in debt, not having health insurance, and the EQ-5D-5L score were uniquely relevant factors. For people with chronic disease, having an undergraduate degree or higher, a diagnosis of cancer, and the BIPQ score were uniquely relevant factors. Conclusions and Relevance These results suggest that basic personal information, economic status, health knowledge, and physical health status were the main factors associated with the acceptance of gene therapy. Improving the health literacy of the population and promoting trust in gene therapy may be effective ways to increase the acceptance of gene therapy. Poorer economic levels and worse disease states may reduce the public's willingness to accept gene therapy.
Collapse
Affiliation(s)
- Yiqi Li
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xinyue Zhang
- School of Health Policy and Management, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ze Xiang
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Tianle Chen
- School of Mathematical Sciences, Zhejiang University, Hangzhou, China
| | - Zihao Hu
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Kexin Yang
- School of Health Policy and Management, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xinying Sun
- School of Public Health, Peking University, Beijing, China
| | - Yibo Wu
- School of Public Health, Peking University, Beijing, China
| | - Jian Wu
- Department of Laboratory Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| |
Collapse
|
4
|
Sauvagère S, Siatka C. CRISPR-Cas: 'The Multipurpose Molecular Tool' for Gene Therapy and Diagnosis. Genes (Basel) 2023; 14:1542. [PMID: 37628594 PMCID: PMC10454384 DOI: 10.3390/genes14081542] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/20/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023] Open
Abstract
Since the discovery of the CRISPR-Cas engineering system in 2012, several approaches for using this innovative molecular tool in therapeutic strategies and even diagnosis have been investigated. The use of this tool requires a global approach to DNA damage processes and repair systems in cells. The diversity in the functions of various Cas proteins allows for the use of this technology in clinical applications and trials. Wide variants of Cas12 and Cas13 are exploited using the collateral effect in many diagnostic applications. Even though this tool is well known, its use still raises real-world ethical and regulatory questions.
Collapse
|
5
|
Ben Yacoub T, Wohlschlegel J, Sahel JA, Zeitz C, Audo I. [CRISPR/Cas9: From research to therapeutic application]. J Fr Ophtalmol 2023; 46:398-407. [PMID: 36759244 DOI: 10.1016/j.jfo.2022.10.008] [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: 10/03/2022] [Revised: 10/12/2022] [Accepted: 10/21/2022] [Indexed: 02/10/2023]
Abstract
For several decades, genome engineering has raised interest among many researchers and physicians in the study of genetic disorders and their treatments. Compared to its predecessors, zinc-finger nucleases (ZFN) and transcription activator-like effectors (TALEN), clustered regularly interspaced short palindromic repeats (CRISPR/Cas9) is currently the most efficient molecular tool for genome editing. This system, originally identified as a bacterial adaptive immune system, is capable of cutting and modifying any gene of a large number of living organisms. Numerous trials using this technology are being developed to provide effective treatment for several diseases, such as cancer, cardiovascular and ophthalmic disorders. In research, this technology is increasingly used for genetic disease modelling, providing meaningful models of relevant studies as well as a better understanding of underlying pathological mechanisms. Many molecular tools are now available to put this technique into practice in laboratories, and despite the technical and ethical issues raised by manipulation of the genome, CRIPSR/Cas9 offers a new breath of hope for therapeutic research around the world.
Collapse
Affiliation(s)
- T Ben Yacoub
- Sorbonne université, Inserm, CNRS, institut de la Vision, 75012 Paris, France.
| | - J Wohlschlegel
- Sorbonne université, Inserm, CNRS, institut de la Vision, 75012 Paris, France
| | - J-A Sahel
- Sorbonne université, Inserm, CNRS, institut de la Vision, 75012 Paris, France; CHNO des Quinze-Vingts, Inserm-DGOS CIC 1423, 75012 Paris, France; Department of ophthalmology, fondation ophtalmologique Adolphe De Rothschild, 75019 Paris, France; Department of ophthalmology, the university of Pittsburgh School of Medicine, Pittsburgh PA 15213, United States; Académie des sciences, institut de France, 75006 Paris, France
| | - C Zeitz
- Sorbonne université, Inserm, CNRS, institut de la Vision, 75012 Paris, France
| | - I Audo
- Sorbonne université, Inserm, CNRS, institut de la Vision, 75012 Paris, France; CHNO des Quinze-Vingts, Inserm-DGOS CIC 1423, 75012 Paris, France; Institute of ophthalmology, university College of London, London EC1V 9EL, United Kingdom
| |
Collapse
|
6
|
Cui Y, Yin S, Qin X, Jiao W, Ren A, Wang F, Zhao B. Advances in the treatment of intraocular malignancies: A literature review. Front Med (Lausanne) 2022; 9:975565. [PMID: 36330064 PMCID: PMC9624174 DOI: 10.3389/fmed.2022.975565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/17/2022] [Indexed: 11/29/2022] Open
Abstract
Intraocular malignant tumors including primary and metastatic tumors, are mainly found in Retina and uvea, and very few cases originate from the sclera and optic nerve. Intraocular tumors can endanger the patient's vision and even life, and proper treatment is vital. There have been several traditional treatments for intraocular tumors, such as radiotherapy, chemotherapy and surgery. In recent years, new methods have been developed in clinical applications including anti-VEGF and gene therapy. This paper aims to provide a timely review about recent progress in the treatment of intraocular malignant tumor.
Collapse
Affiliation(s)
- Yanyan Cui
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shan Yin
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xuejiao Qin
- The Second Hospital of Shandong University, Jinan, China
| | - Wanzhen Jiao
- Department of Ophthalmology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Anqi Ren
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Fei Wang
- Shengli Oilfield Central Hospital, Dongying, China
| | - Bojun Zhao
- Department of Ophthalmology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- *Correspondence: Bojun Zhao
| |
Collapse
|
7
|
Zhang J, Zhang T, Gao J. Biocompatible Iron Oxide Nanoparticles for Targeted Cancer Gene Therapy: A Review. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:nano12193323. [PMID: 36234452 PMCID: PMC9565336 DOI: 10.3390/nano12193323] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/12/2022] [Accepted: 09/20/2022] [Indexed: 05/14/2023]
Abstract
In recent years, gene therapy has made remarkable achievements in tumor treatment. In a successfully cancer gene therapy, a smart gene delivery system is necessary for both protecting the therapeutic genes in circulation and enabling high gene expression in tumor sites. Magnetic iron oxide nanoparticles (IONPs) have demonstrated their bright promise for highly efficient gene delivery target to tumor tissues, partly due to their good biocompatibility, magnetic responsiveness, and extensive functional surface modification. In this review, the latest progress in targeting cancer gene therapy is introduced, and the unique properties of IONPs contributing to the efficient delivery of therapeutic genes are summarized with detailed examples. Furthermore, the diagnosis potentials and synergistic tumor treatment capacity of IONPs are highlighted. In addition, aiming at potential risks during the gene delivery process, several strategies to improve the efficiency or reduce the potential risks of using IONPs for cancer gene therapy are introduced and addressed. The strategies and applications summarized in this review provide a general understanding for the potential applications of IONPs in cancer gene therapy.
Collapse
Affiliation(s)
- Jinsong Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Tianyuan Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Correspondence: (T.Z.); (J.G.)
| | - Jianqing Gao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Department of Pharmacy, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
- Correspondence: (T.Z.); (J.G.)
| |
Collapse
|
8
|
Dahl E, Villwock S, Habenberger P, Choidas A, Rose M, Klebl BM. White Paper: Mimetics of Class 2 Tumor Suppressor Proteins as Novel Drug Candidates for Personalized Cancer Therapy. Cancers (Basel) 2022; 14:cancers14184386. [PMID: 36139547 PMCID: PMC9496810 DOI: 10.3390/cancers14184386] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/01/2022] [Accepted: 09/07/2022] [Indexed: 11/21/2022] Open
Abstract
Simple Summary A concept is presented for a new therapeutic approach, still in its early stages, which focuses on the phenotypic mimicry (“mimesis”) of proteins encoded by highly disease-relevant class 2 tumor suppressor genes that are silenced by DNA promoter methylation. Proteins derived from tumor suppressor genes are usually considered control systems of cells against oncogenic properties. Thus they represent the brakes in the “car-of-life.” Restoring this “brake function” in tumors by administering mimetic drugs may have a significant therapeutic effect. The proposed approach could thus open up a new, hitherto unexploited area of research for the development of anticancer drugs for difficult-to-treat cancers. Abstract The aim of our proposed concept is to find new target structures for combating cancers with unmet medical needs. This, unfortunately, still applies to the majority of the clinically most relevant tumor entities such as, for example, liver cancer, pancreatic cancer, and many others. Current target structures almost all belong to the class of oncogenic proteins caused by tumor-specific genetic alterations, such as activating mutations, gene fusions, or gene amplifications, often referred to as cancer “driver alterations” or just “drivers.” However, restoring the lost function of tumor suppressor genes (TSGs) could also be a valid approach to treating cancer. TSG-derived proteins are usually considered as control systems of cells against oncogenic properties; thus, they represent the brakes in the “car-of-life.” Restoring these tumor-defective brakes by gene therapy has not been successful so far, with a few exceptions. It can be assumed that most TSGs are not being inactivated by genetic alteration (class 1 TSGs) but rather by epigenetic silencing (class 2 TSGs or short “C2TSGs”). Reactivation of C2TSGs in cancer therapy is being addressed by the use of DNA demethylating agents and histone deacetylase inhibitors which act on the whole cancer cell genome. These epigenetic therapies have neither been particularly successful, probably because they are “shotgun” approaches that, although acting on C2TSGs, may also reactivate epigenetically silenced oncogenic sequences in the genome. Thus, new strategies are needed to exploit the therapeutic potential of C2TSGs, which have also been named DNA methylation cancer driver genes or “DNAme drivers” recently. Here we present a concept for a new translational and therapeutic approach that focuses on the phenotypic imitation (“mimesis”) of proteins encoded by highly disease-relevant C2TSGs/DNAme drivers. Molecular knowledge on C2TSGs is used in two complementary approaches having the translational concept of defining mimetic drugs in common: First, a concept is presented how truncated and/or genetically engineered C2TSG proteins, consisting solely of domains with defined tumor suppressive function can be developed as biologicals. Second, a method is described for identifying small molecules that can mimic the effect of the C2TSG protein lost in the cancer cell. Both approaches should open up a new, previously untapped discovery space for anticancer drugs.
Collapse
Affiliation(s)
- Edgar Dahl
- Institute of Pathology, Medical Faculty, RWTH Aachen University, D-52074 Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), D-52074 Aachen, Germany
- Correspondence:
| | - Sophia Villwock
- Institute of Pathology, Medical Faculty, RWTH Aachen University, D-52074 Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), D-52074 Aachen, Germany
| | - Peter Habenberger
- Lead Discovery Center GmbH (LDC), Otto-Hahn-Straße 15, D-44227 Dortmund, Germany
| | - Axel Choidas
- Lead Discovery Center GmbH (LDC), Otto-Hahn-Straße 15, D-44227 Dortmund, Germany
| | - Michael Rose
- Institute of Pathology, Medical Faculty, RWTH Aachen University, D-52074 Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), D-52074 Aachen, Germany
| | - Bert M. Klebl
- Lead Discovery Center GmbH (LDC), Otto-Hahn-Straße 15, D-44227 Dortmund, Germany
| |
Collapse
|
9
|
Li Y, Tang K, Zhang X, Pan W, Li N, Tang B. Tumor microenvironment responsive nanocarriers for gene therapy. Chem Commun (Camb) 2022; 58:8754-8765. [PMID: 35880654 DOI: 10.1039/d2cc02759c] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Stimuli responsive nanocarriers are important non-viral gene carriers for gene therapy. We discuss the stimulus conditions and then highlight various stimuli responsive nanocarriers in the tumor microenvironment for cancer gene therapy. We hope that this review will inspire readers to develop more effective stimuli responsive nanocarriers for delivering genes.
Collapse
Affiliation(s)
- Yanhua Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Kun Tang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Xia Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Wei Pan
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Na Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| |
Collapse
|
10
|
Deng L, Liang P, Cui H. Pseudotyped lentiviral vectors: Ready for translation into targeted cancer gene therapy? Genes Dis 2022. [PMID: 37492721 PMCID: PMC10363566 DOI: 10.1016/j.gendis.2022.03.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Gene therapy holds great promise for curing cancer by editing the deleterious genes of tumor cells, but the lack of vector systems for efficient delivery of genetic material into specific tumor sites in vivo has limited its full therapeutic potential in cancer gene therapy. Over the past two decades, increasing studies have shown that lentiviral vectors (LVs) modified with different glycoproteins from a donating virus, a process referred to as pseudotyping, have altered tropism and display cell-type specificity in transduction, leading to selective tumor cell killing. This feature of LVs together with their ability to enable high efficient gene delivery in dividing and non-dividing mammalian cells in vivo make them to be attractive tools in future cancer gene therapy. This review is intended to summarize the status quo of some typical pseudotypings of LVs and their applications in basic anti-cancer studies across many malignancies. The opportunities of translating pseudotyped LVs into clinic use in cancer therapy have also been discussed.
Collapse
|
11
|
Advances in Allogeneic Cancer Cell Therapy and Future Perspectives on “Off-the-Shelf” T Cell Therapy Using iPSC Technology and Gene Editing. Cells 2022; 11:cells11020269. [PMID: 35053386 PMCID: PMC8773622 DOI: 10.3390/cells11020269] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/07/2022] [Accepted: 01/10/2022] [Indexed: 12/03/2022] Open
Abstract
The concept of allogeneic cell therapy was first presented over 60 years ago with hematopoietic stem cell transplantation. However, complications such as graft versus host disease (GVHD) and regimen-related toxicities remained as major obstacles. To maximize the effect of graft versus leukemia, while minimizing the effect of GVHD, donor lymphocyte infusion was utilized. This idea, which was used against viral infections, postulated that adoptive transfer of virus-specific cytotoxic T lymphocytes could reconstitute specific immunity and eliminate virus infected cells and led to the idea of banking third party cytotoxic T cells (CTLs). T cell exhaustion sometimes became a problem and difficulty arose in creating robust CTLs. However, the introduction of induced pluripotent stem cells (iPSCs) lessens such problems, and by using iPSC technology, unlimited numbers of allogeneic rejuvenated CTLs with robust and proliferative cytotoxic activity can be created. Despite this revolutionary concept, several concerns still exist, such as immunorejection by recipient cells and safety issues of gene editing. In this review, we describe approaches to a feasible “off-the-shelf” therapy that can be distributed rapidly worldwide. We also offer perspectives on the future of allogeneic cell cancer immunotherapy.
Collapse
|
12
|
Xia Y, Li X, Sun W. Applications of Recombinant Adenovirus-p53 Gene Therapy for Cancers in the Clinic in China. Curr Gene Ther 2021; 20:127-141. [PMID: 32951572 DOI: 10.2174/1566523220999200731003206] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/30/2020] [Accepted: 07/10/2020] [Indexed: 01/30/2023]
Abstract
Suppression of TP53 function is nearly ubiquitous in human cancers, and a significant fraction of cancers have mutations in the TP53 gene itself. Therefore, the wild-type TP53 gene has become an important target gene for transformation research of cancer gene therapy. In 2003, the first anti-tumor gene therapy drug rAd-p53 (recombinant human p53 adenovirus), trade name Gendicine™, was approved by the China Food and Drug Administration (CFDA) for treatment of head and neck squamous cell carcinoma (HNSCC) in combination with radiotherapy. The recombinant human TP53 gene is delivered into cancer cells by an adenovirus vector constructed to express the functional p53 protein. Although the only currently approved used of Gendicine is in combination with radiotherapy for treatment of HNSCC, clinical studies have been carried out for more than 20 other applications of Gendicine in treating cancer, including treatment of advanced lung cancer, advanced liver cancer, malignant gynecological tumors, and soft tissue sarcomas. Currently more than 30,000 patients have been treated with Gendicine. This review provides an overview of the clinical applications of Gendicine in China. We summarize a total of 48 studies with 2,561 patients with solid tumors, including 34 controlled clinical studies and 14 open clinical studies, i.e., clinical studies without a control group. There are 11 studies for head and neck cancer, 10 for liver cancer, 6 for malignant gynecological tumors, 4 for non-small cell lung cancer, 4 for soft tissue sarcoma, 4 for malignant effusion, 2 for gastrointestinal tumors, and 7 for other types of cancer. In all the reported clinical studies, the most common side effect was self-limited fever. Intratumoral injection and intra-arterial infusion were the most common routes of administration. Overall, Gendicine combined with chemotherapy, radiotherapy, or other conventional treatment regimens demonstrated significantly higher response rates compared to standard therapies alone. Some of the published studies also showed that Gendicine combination regimens demonstrated longer progression-free survival times than conventional treatments alone. To date, Gendicine has been clinically used in China for treatment of cancers other than HNSCC for more than ten years, mainly for patients with advanced or unresectable malignant tumors. However, the establishment of standard treatment regimens using TP53 gene therapy is still needed in order to advance its use in clinical practice.
Collapse
Affiliation(s)
- Yu Xia
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, Liaoning, 110004, China
| | - Xiuqin Li
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, Liaoning, 110004, China
| | - Wei Sun
- Radiology Department, Shengjing Hospital of China Medical University, Sanhao, China
| |
Collapse
|
13
|
Bhoopathi P, Mannangatti P, Emdad L, Das SK, Fisher PB. The quest to develop an effective therapy for neuroblastoma. J Cell Physiol 2021; 236:7775-7791. [PMID: 33834508 DOI: 10.1002/jcp.30384] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/27/2021] [Accepted: 03/22/2021] [Indexed: 12/18/2022]
Abstract
Neuroblastoma (NB) is a common solid extracranial tumor developing in pediatric populations. NB can spontaneously regress or grow and metastasize displaying resistance to therapy. This tumor is derived from primitive cells, mainly those of the neural crest, in the sympathetic nervous system and usually develops in the adrenal medulla and paraspinal ganglia. Our understanding of the molecular characteristics of human NBs continues to advance documenting abnormalities at the genome, epigenome, and transcriptome levels. The high-risk tumors have MYCN oncogene amplification, and the MYCN transcriptional regulator encoded by the MYCN oncogene is highly expressed in the neural crest. Studies on the biology of NB has enabled a more precise risk stratification strategy and a concomitant reduction in the required treatment in an expanding number of cases worldwide. However, newer treatment strategies are mandated to improve outcomes in pediatric patients who are at high-risk and display relapse. To improve outcomes and survival rates in such high-risk patients, it is necessary to use a multicomponent therapeutic approach. Accuracy in clinical staging of the disease and assessment of the associated risks based on biological, clinical, surgical, and pathological criteria are of paramount importance for prognosis and to effectively plan therapeutic approaches. This review discusses the staging of NB and the biological and genetic features of the disease and several current therapies including targeted delivery of chemotherapy, novel radiation therapy, and immunotherapy for NB.
Collapse
Affiliation(s)
- Praveen Bhoopathi
- Department of Human and Molecular Genetics, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA.,VCU Institute of Molecular Medicine, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Padmanabhan Mannangatti
- Department of Human and Molecular Genetics, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Luni Emdad
- Department of Human and Molecular Genetics, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA.,VCU Institute of Molecular Medicine, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA.,VCU Massey Cancer Center, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Swadesh K Das
- Department of Human and Molecular Genetics, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA.,VCU Institute of Molecular Medicine, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA.,VCU Massey Cancer Center, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Paul B Fisher
- Department of Human and Molecular Genetics, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA.,VCU Institute of Molecular Medicine, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA.,VCU Massey Cancer Center, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
14
|
Mashima H, Zhang R, Kobayashi T, Tsukamoto H, Liu T, Iwama T, Hagiya Y, Yamamoto M, Fukushima S, Okada S, Idiris A, Kaneko S, Nakatsura T, Ohdan H, Uemura Y. Improved safety of induced pluripotent stem cell-derived antigen-presenting cell-based cancer immunotherapy. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 21:171-179. [PMID: 33816647 PMCID: PMC7994724 DOI: 10.1016/j.omtm.2021.03.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 03/02/2021] [Indexed: 11/02/2022]
Abstract
The tumorigenicity and toxicity of induced pluripotent stem cells (iPSCs) and their derivatives are major safety concerns in their clinical application. Recently, we developed granulocyte-macrophage colony-stimulating factor (GM-CSF)-producing proliferating myeloid cells (GM-pMCs) from mouse iPSCs as a source of unlimited antigen-presenting cells for use in cancer immunotherapy. As GM-pMCs are generated by introducing c-Myc and Csf2 into iPSC-derived MCs and are dependent on self-produced GM-CSF for proliferation, methods to control their proliferation after administration should be introduced to improve safety. In this study, we compared the efficacy of two promising suicide gene systems, herpes simplex virus-thymidine kinase (HSV-TK)/ganciclovir (GCV) and inducible caspase-9 (iCasp9)/AP1903, for safeguarding GM-pMCs in cancer immunotherapy. The expression of HSV-TK or iCasp9 did not impair the fundamental properties of GM-pMCs. Both of these suicide gene-expressing cells selectively underwent apoptosis after treatment with the corresponding apoptosis-inducing drug, and they were promptly eliminated in vivo. iCasp9/AP1903 induced apoptosis more efficiently than HSV-TK/GCV. Furthermore, high concentrations of GCV were toxic to cells not expressing HSV-TK, whereas AP1903 was bioinert. These results suggest that iCasp9/AP1903 is superior to HSV-TK/GCV in terms of both safety and efficacy when controlling the fate of GM-pMCs after priming antitumor immunity.
Collapse
Affiliation(s)
- Hiroaki Mashima
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan.,Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical & Health Science, Hiroshima University, Hiroshima 734-8551, Japan
| | - Rong Zhang
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan
| | - Tsuyoshi Kobayashi
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical & Health Science, Hiroshima University, Hiroshima 734-8551, Japan
| | - Hirotake Tsukamoto
- Department of Immunology, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Tianyi Liu
- Key Laboratory of Cancer Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Tatsuaki Iwama
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan
| | - Yuichiro Hagiya
- Biochemistry Team, Bio Science Division, Technology General Division, Materials Integration Laboratories, AGC, Inc., Yokohama 221-8755, Japan
| | - Masateru Yamamoto
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan.,Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical & Health Science, Hiroshima University, Hiroshima 734-8551, Japan
| | - Satoshi Fukushima
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Seiji Okada
- Division of Hematopoiesis, Center for AIDS Research, Kumamoto University, Kumamoto 860-8556, Japan
| | - Alimjan Idiris
- Biochemistry Team, Bio Science Division, Technology General Division, Materials Integration Laboratories, AGC, Inc., Yokohama 221-8755, Japan
| | - Shin Kaneko
- Shin Kaneko Laboratory, Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan
| | - Tetsuya Nakatsura
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan
| | - Hideki Ohdan
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical & Health Science, Hiroshima University, Hiroshima 734-8551, Japan
| | - Yasushi Uemura
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan
| |
Collapse
|
15
|
He Y, Wang M, Li X, Yu T, Gao X. Targeted MIP-3β plasmid nanoparticles induce dendritic cell maturation and inhibit M2 macrophage polarisation to suppress cancer growth. Biomaterials 2020; 249:120046. [PMID: 32325346 DOI: 10.1016/j.biomaterials.2020.120046] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 04/05/2020] [Accepted: 04/09/2020] [Indexed: 02/05/2023]
Abstract
In recent decades, cancer immunotherapy has demonstrated considerable clinical advantages in cancer therapy. Particularly, the use of immunological gene therapy continues to grow in this field. Macrophage Inflammatory Protein 3 Beta (MIP-3β) has emerged as a potential immunomodulator for anti-cancer treatments by enhancing the interaction among immune responses. In this study, we demonstrate an innovative targeted gene delivery system based on a self-assembly technique with 1,2-Dioleoyl-3-trimethylammonium-propane (DOTAP), Methoxy poly(ethylene glycol)-poly(lactide) (MPEG-PLA), and folic acid modified poly(ethylene glycol)-poly(ε-caprolactone) (FA-PEG-PCL) (FDMCA). Results showed that the expression of MIP-3β was up-regulated in cancer cells following the transfection with FDMCA-pMIP-3β, in comparison with cells transfected with DMCA-pMIP-3β. The supernatants collected from cancer cells transfected with FDMCA-pMIP-3β and DMCA-pMIP-3β both instigate dendritic cell maturation, M1 polarisation of macrophages, activation and presentation of cytotoxicity in lymphocytes. Moreover, tumor growth and metastasis were markedly inhibited following the administration of the FDMCA-pMIP-3β complex in both subcutaneous and pulmonary metastasis mice models, which is attributed to reduced angiogenesis, enhanced cancer cell apoptosis, and suppressed proliferation by activation of the immune system. Our study suggests that the MIP-3β plasmid and FDMCA complex provide a new approach for the treatment of breast cancer.
Collapse
Affiliation(s)
- Yihong He
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, PR China
| | - Manni Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, PR China
| | - Xiaoling Li
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, PR China
| | - Ting Yu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, PR China; Department of Pathology and Laboratory of Pathology, State Key Laboratory of Biotherapy, West China Hospital, West China School of Medicine, Sichuan University, PR China
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, PR China.
| |
Collapse
|
16
|
Iskrov G, Vasilev G, Stefanov R. What could gene therapies learn from orphan drugs’ post-regulatory approval access in the EU? Expert Opin Orphan Drugs 2019. [DOI: 10.1080/21678707.2019.1663171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Georgi Iskrov
- Department of Social Medicine and Public Health, Faculty of Public Health, Medical University of Plovdiv, Plovdiv, Bulgaria
- Institute for Rare Diseases, Plovdiv, Bulgaria
| | - Georgi Vasilev
- Faculty of Medicine, Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Rumen Stefanov
- Department of Social Medicine and Public Health, Faculty of Public Health, Medical University of Plovdiv, Plovdiv, Bulgaria
- Institute for Rare Diseases, Plovdiv, Bulgaria
| |
Collapse
|
17
|
Lin Q, Wang DG, Zhang ZQ, Liu DP. Applications of Virus Vector-Mediated Gene Therapy in China. Hum Gene Ther 2019; 29:98-109. [PMID: 29284296 DOI: 10.1089/hum.2017.238] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Due to the increased safety and efficiency of virus vectors, virus vector-mediated gene therapy is now widely used for various diseases, including monogenic diseases, complex disorders, and infectious diseases. Recent gene therapy trials have shown significant therapeutic benefits, and Chinese researchers have contributed significantly to this progress. This review highlights disease applications and strategies for virus vector-mediated gene therapy in preclinical studies and clinical trials in China.
Collapse
Affiliation(s)
- Qiong Lin
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences , Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Deng-Gao Wang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences , Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhu-Qin Zhang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences , Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - De-Pei Liu
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences , Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
18
|
Zheng D, Li B, Xu L, Zhang QL, Fan JX, Li CX, Zhang XZ. Normalizing Tumor Microenvironment Based on Photosynthetic Abiotic/Biotic Nanoparticles. ACS NANO 2018; 12:6218-6227. [PMID: 29791792 DOI: 10.1021/acsnano.8b02977] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Tumor hypoxia has attained the status of a core hallmark of cancer that globally affects the entire tumor phenotype. Reversing tumor hypoxia might offer alternative therapeutic opportunities for current anticancer therapies. In this research, a photosynthetic leaf-inspired abiotic/biotic nano-thylakoid (PLANT) system was designed by fusing the thylakoid membrane with synthetic nanoparticles for efficient O2 generation in vivo. Under 660 nm laser irradiation, the PLANT system exhibited intracellular O2 generation and the anaerobic respiration of the multicellular tumor spheroid was suppressed by PLANT as well. In vivo, it was found that PLANT could not only normalize the entire metabolic network but also adjust the abnormal structure and function of the tumor vasculature. It was demonstrated that PLANT could significantly enhance the efficacy of phototherapy or antiangiogenesis therapy. This facile approach for normalizing the tumor microenvironment will find great potential in tumor therapy.
Collapse
Affiliation(s)
- Diwei Zheng
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry , Wuhan University , Wuhan 430072 , People's Republic of China
| | - Bin Li
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry , Wuhan University , Wuhan 430072 , People's Republic of China
| | - Lu Xu
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry , Wuhan University , Wuhan 430072 , People's Republic of China
| | - Qiu-Ling Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry , Wuhan University , Wuhan 430072 , People's Republic of China
| | - Jin-Xuan Fan
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry , Wuhan University , Wuhan 430072 , People's Republic of China
| | - Chu-Xin Li
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry , Wuhan University , Wuhan 430072 , People's Republic of China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry , Wuhan University , Wuhan 430072 , People's Republic of China
| |
Collapse
|
19
|
Zhao H, To KKW, Chu H, Ding Q, Zhao X, Li C, Shuai H, Yuan S, Zhou J, Kok KH, Jiang S, Yuen KY. Dual-functional peptide with defective interfering genes effectively protects mice against avian and seasonal influenza. Nat Commun 2018; 9:2358. [PMID: 29907765 PMCID: PMC6004018 DOI: 10.1038/s41467-018-04792-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 04/25/2018] [Indexed: 11/09/2022] Open
Abstract
Limited efficacy of current antivirals and antiviral-resistant mutations impairs anti-influenza treatment. Here, we evaluate the in vitro and in vivo antiviral effect of three defective interfering genes (DIG-3) of influenza virus. Viral replication is significantly reduced in cell lines transfected with DIG-3. Mice treated with DIG-3 encoded by jetPEI-vector, as prophylaxis and therapeutics against A(H7N7) virus, respectively, have significantly better survivals (80% and 50%) than control mice (0%). We further develop a dual-functional peptide TAT-P1, which delivers DIG-3 with high efficiency and concomitantly exerts antiviral activity by preventing endosomal acidification. TAT-P1/DIG-3 is more effective than jetPEI/DIG-3 in treating A(H7N7) or A(H1N1)pdm09-infected mice and shows potent prophylactic protection on A(H7N7) or A(H1N1)pdm09-infected mice. The addition of P1 peptide, which prevents endosomal acidification, can enhance the protection of TAT-P1/DIG-3 on A(H1N1)pdm09-infected mice. Dual-functional TAT-P1 with DIG-3 can effectively protect or treat mice infected by avian and seasonal influenza virus.
Collapse
Affiliation(s)
- Hanjun Zhao
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong.,Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Kelvin K W To
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong.,Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong.,Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Hin Chu
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong.,Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Qiulu Ding
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Xiaoyu Zhao
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Cun Li
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Huiping Shuai
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Shuofeng Yuan
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong.,Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Jie Zhou
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong.,Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Kin-Hang Kok
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong.,Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Shanghai Medical College and Institute of Medical Microbiology, Fudan University, Shanghai, 200032, China.,Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, 10065, USA
| | - Kwok-Yung Yuen
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong. .,Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong. .,Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong. .,The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong.
| |
Collapse
|
20
|
Mainini F, Larsen DS, Webster GA, Young SL, Eccles MR. MIS416 as a siRNA Delivery System with the Ability to Target Antigen-Presenting Cells. Nucleic Acid Ther 2018; 28:225-232. [PMID: 29893623 PMCID: PMC6080116 DOI: 10.1089/nat.2017.0695] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
MIS416 is a microparticulate formulation derived from propionibacterium acnes cell wall skeletons with intrinsic adjuvant activity. Conjugates of MIS416-SS-peptide containing a disulfide linkage facilitate the cytoplasmic delivery and release of peptides in antigen-presenting cells (APCs). We hypothesized that MIS416-siRNA (small interfering RNA) conjugates, containing a disulfide linkage between MIS416 and the siRNA, would allow cytoplasmic release of siRNA in APCs. MIS416-SS-siStat3 conjugates added to cell culture medium of monolayers of DCs in culture flasks successfully targeted Stat3 mRNA in DCs in vitro without transfection, downregulating Stat3 mRNA and protein levels. These results suggest that MIS416-SS-siRNA conjugates can be used as a novel siRNA delivery system for the knockdown of mRNA levels in APCs.
Collapse
Affiliation(s)
- Francesco Mainini
- 1 Department of Pathology, University of Otago , Dunedin, New Zealand
| | - David S Larsen
- 2 Department of Chemistry, University of Otago , Dunedin, New Zealand
| | | | - Sarah L Young
- 1 Department of Pathology, University of Otago , Dunedin, New Zealand .,4 Maurice Wilkins Center for Molecular Biodiscovery , Auckland, New Zealand
| | - Michael R Eccles
- 1 Department of Pathology, University of Otago , Dunedin, New Zealand .,4 Maurice Wilkins Center for Molecular Biodiscovery , Auckland, New Zealand
| |
Collapse
|
21
|
Therapeutic activity of retroviral replicating vector-mediated prodrug activator gene therapy for pancreatic cancer. Cancer Gene Ther 2018; 25:184-195. [PMID: 29735994 DOI: 10.1038/s41417-018-0020-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 02/17/2018] [Indexed: 01/20/2023]
Abstract
Toca 511, a retroviral replicating vector (RRV) encoding the yeast cytosine deaminase (yCD) prodrug activator gene, which mediates conversion of the prodrug 5-fluorocytosine (5-FC) to the anticancer drug 5-fluorouracil (5-FU), is currently being evaluated in Phase II/III clinical trials for glioma, and showing highly promising evidence of therapeutic activity. Here we evaluated RRV-mediated prodrug activator gene therapy as a new therapeutic approach for pancreatic ductal adenocarcinoma (PDAC). RRV spread rapidly and conferred significant cytotoxicity with prodrug in a panel of PDAC cells. Efficient intratumoral replication and complete inhibition of tumor growth upon 5-FC administration were observed in both immunodeficient and immunocompetent subcutaneous PDAC models. Biodistribution of RRV was highly restricted in normal tissues, especially in immunocompetent hosts. Tumor growth inhibition by Toca 511 followed by 5-FC was also confirmed in the orthotopic PDAC model. This study provides the first proof-of-concept for application of Toca 511 and Toca FC (extended release 5-FC) to the treatment of human PDAC, and provided support for inclusion of PDAC in a Phase I study evaluating Toca 511 in various systemic malignancies, (NCT02576665), which has recently been initiated.
Collapse
|
22
|
Survivin-Based Treatment Strategies for Squamous Cell Carcinoma. Int J Mol Sci 2018; 19:ijms19040971. [PMID: 29587347 PMCID: PMC5979467 DOI: 10.3390/ijms19040971] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 03/20/2018] [Accepted: 03/21/2018] [Indexed: 02/07/2023] Open
Abstract
Survivin, an anti-apoptotic molecule abundantly expressed in most human neoplasms, has been reported to contribute to cancer initiation and drug resistance in a wide variety of human tumors. Efficient downregulation of survivin can sensitize tumor cells to various therapeutic interventions, generating considerable efforts in its validation as a new target in cancer therapy. This review thoroughly analyzes up-to-date information on the potential of survivin as a therapeutic target for new anticancer treatments. The literature dealing with the therapeutic targeting of survivin will be reviewed, discussing specifically squamous cell carcinomas (SCCs), and with emphasis on the last clinical trials. This review gives insight into the recent developments undertaken in validating various treatment strategies that target survivin in SCCs and analyze the translational possibility, identifying those strategies that seem to be the closest to being incorporated into clinical practice. The most recent developments, such as dominant-negative survivin mutants, RNA interference, anti-sense oligonucleotides, small-molecule inhibitors, and peptide-based immunotherapy, seem to be helpful for effectively downregulating survivin expression and reducing tumor growth potential, increasing the apoptotic rate, and sensitizing tumor cells to chemo- and radiotherapy. However, selective and efficient targeting of survivin in clinical trials still poses a major challenge.
Collapse
|
23
|
van Zandwijk N, McDiarmid J, Brahmbhatt H, Reid G. Response to "An innovative mesothelioma treatment based on mir-16 mimic loaded EGFR targeted minicells (TargomiRs)". Transl Lung Cancer Res 2018. [PMID: 29531907 DOI: 10.21037/tlcr.2018.01.11] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Nico van Zandwijk
- Concord Repatriation General Hospital, Concord, NSW, Australia.,Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | | | | | - Glen Reid
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia.,Asbestos Diseases Research Institute, Concord, NSW, Australia
| |
Collapse
|
24
|
Carvalho M, Sepodes B, Martins AP. Regulatory and Scientific Advancements in Gene Therapy: State-of-the-Art of Clinical Applications and of the Supporting European Regulatory Framework. Front Med (Lausanne) 2017; 4:182. [PMID: 29124055 PMCID: PMC5662580 DOI: 10.3389/fmed.2017.00182] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 10/11/2017] [Indexed: 12/21/2022] Open
Abstract
Advanced therapy medicinal products (ATMPs) have a massive potential to address existing unmet medical needs. Specifically, gene therapy medicinal products (GTMPs) may potentially provide cure for several genetic diseases. In Europe, the ATMP regulation was fully implemented in 2009 and, at this point, the Committee for Advanced Therapies was created as a dedicated group of specialists to evaluate medicinal products requiring specific expertise in this area. To date, there are three authorized GTMPs, and the first one was approved in 2012. Broad research has been conducted in this field over the last few decades and different clinical applications are being investigated worldwide, using different strategies that range from direct gene replacement or addition to more complex pathways such as specific gene editing or RNA targeting. Important safety risks, limited efficacy, manufacturing hurdles, or ethical conflicts may represent challenges in the success of a candidate GTMP. During the development process, it is fundamental to take such aspects into account and establish overcoming strategies. This article reviews the current European legal framework of ATMPs, provides an overview of the clinical applications for approved and investigational GTMPs, and discusses critical challenges in the development of GTMPs.
Collapse
Affiliation(s)
- Marta Carvalho
- Faculdade de Farmácia, Research Institute for Medicines and Pharmaceutical Sciences (iMed.ULisboa), Universidade de Lisboa, Lisboa, Portugal
| | - Bruno Sepodes
- Faculdade de Farmácia, Research Institute for Medicines and Pharmaceutical Sciences (iMed.ULisboa), Universidade de Lisboa, Lisboa, Portugal
| | - Ana Paula Martins
- Faculdade de Farmácia, Research Institute for Medicines and Pharmaceutical Sciences (iMed.ULisboa), Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
25
|
Liu X, Gao X, Zheng S, Wang B, Li Y, Zhao C, Muftuoglu Y, Chen S, Li Y, Yao H, Sun H, Mao Q, You C, Guo G, Wei Y. Modified nanoparticle mediated IL-12 immunogene therapy for colon cancer. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:1993-2004. [DOI: 10.1016/j.nano.2017.04.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 04/04/2017] [Accepted: 04/10/2017] [Indexed: 12/18/2022]
|
26
|
Villadangos JA. Antigen-specific impairment of adoptive T-cell therapy against cancer: players, mechanisms, solutions and a hypothesis. Immunol Rev 2017; 272:169-82. [PMID: 27319350 DOI: 10.1111/imr.12433] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Adoptive cell therapy (ACT) destroys tumors with infused cytotoxic T lymphocytes (CTLs). Although successful in some settings, ACT is compromised due to impaired survival or functional inactivation of the CTL. To better understand the mechanisms involved, we have exploited a mouse model of leukemia expressing ovalbumin as a tumor neoantigen to address these questions: (i) Is CTL impairment during ACT antigen specific? (ii) If yes, which are the antigen-presenting cells responsible? (iii) Can this information assist the development of complementary therapies to improve ACT? Our results indicate that the target (tumor) cells, not cross-presenting cells, are the main culprits of antigen-specific CTL inactivation. We find that the affinity/avidity of the CTL-tumor cell interaction has little influence on ACT outcomes, while tumor density is a major determinant. Reduction of tumor burden with mild non-lymphoablative and non-inflammatory chemotherapy can dramatically improve the efficacy of ACT and may minimize side-effects. We propose a general mechanism for the inactivation of anti-self CTL in the same tissues where the activity of anti-foreign CTL is preserved, based on the density of target cells. This mechanism, which we tentatively call stunning, may have evolved to protect infected sites from self-destruction and is exploited by tumors to inactivate CTL.
Collapse
Affiliation(s)
- Jose A Villadangos
- Department of Microbiology and Immunology, Doherty Institute of Infection and Immunity, The University of Melbourne, Melbourne, Vic., Australia.,Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Vic., Australia
| |
Collapse
|
27
|
Folic acid-decorated polyamidoamine dendrimer exhibits high tumor uptake and sustained highly localized retention in solid tumors: Its utility for local siRNA delivery. Acta Biomater 2017; 57:251-261. [PMID: 28438704 DOI: 10.1016/j.actbio.2017.04.023] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 04/06/2017] [Accepted: 04/20/2017] [Indexed: 12/13/2022]
Abstract
The utility of folic acid (FA)-decorated polyamidoamine dendrimer G4 (G4-FA) as a vector was investigated for local delivery of siRNA. In a xenograft HN12 (or HN12-YFP) tumor mouse model of head and neck squamous cell carcinomas (HNSCC), intratumorally (i.t.) injected G4-FA exhibited high tumor uptake and sustained highly localized retention in the tumors according to near infrared (NIR) imaging assessment. siRNA against vascular endothelial growth factor A (siVEGFA) was chosen as a therapeutic modality. Compared to the nontherapeutic treatment groups (PBS solution or dendrimer complexed with nontherapeutic siRNA against green fluorescent protein (siGFP)), G4-FA/siVEGFA showed tumor inhibition effects in single-dose and two-dose regimen studies. In particular, two doses of G4-FA/siVEGFA i.t. administered eight days apart resulted in a more profound inhibition of tumor growth, accompanied with significant reduction in angiogenesis, as judged by CD31 staining and microvessel counts. Tumor size reduction in the two-dose regimen study was ascertained semi-quantitatively by live fluorescence imaging of YFP tumors and independently supported antitumor effects of G4-FA/siVEGFA. Taken together, G4-FA shows high tumor uptake and sustained retention properties, making it a suitable platform for local delivery of siRNAs to treat cancers that are readily accessible such as HNSCC. STATEMENT OF SIGNIFICANCE Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer worldwide and is difficult to transfect for gene therapy. We developed folate receptor (FR)-targeted polyamidoamine (PAMAM) dendrimer for enhanced delivery of genes to HNSCC and gained in-depth understanding of how gene delivery and transfection in head and neck squamous cancer cells can be enhanced via FR-targeted PAMAM dendrimers. The results we report here are encouraging and present latest advances in using dendrimers for cancer therapies, in particular for HNSCC. Our work has demonstrated that localized delivery of FR-targeted PAMAM dendrimer G4 complexed with siVEGFA resulted in pronounced tumor suppression in an HN12 xenograft tumor model. Tumor suppression was attributed to enhanced tumor uptake of siRNA and prolonged nanoparticle retention in the tumor. Taken together, G4-FA shows high tumor uptake and sustained highly localized retention properties, making it a suitable platform for local delivery of siRNAs to treat cancers that are readily accessible such as HNSCC.
Collapse
|
28
|
Sun D, Zhao L, Lin J, Zhao Y, Zheng Y. Cationic liposome co-encapsulation of SMAC mimetic and zVAD using a novel lipid bilayer fusion loaded with MLKL-pDNA for tumour inhibition in vivo. J Drug Target 2017. [PMID: 28649853 DOI: 10.1080/1061186x.2017.1339192] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Dan Sun
- College of Life Sciences, Sichuan University, Chengdu, Sichuan, PR China
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Linshu Zhao
- Division of Biosciences, Faculty of Life Sciences, University College London, London, UK
| | - Junzhong Lin
- Department of Colorectal Surgery,Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yun Zhao
- College of Life Sciences, Sichuan University, Chengdu, Sichuan, PR China
| | - Yu Zheng
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| |
Collapse
|
29
|
Fail-Safe System against Potential Tumorigenicity after Transplantation of iPSC Derivatives. Stem Cell Reports 2017; 8:673-684. [PMID: 28262544 PMCID: PMC5355810 DOI: 10.1016/j.stemcr.2017.02.003] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 02/02/2017] [Accepted: 02/02/2017] [Indexed: 01/10/2023] Open
Abstract
Human induced pluripotent stem cells (iPSCs) are promising in regenerative medicine. However, the risks of teratoma formation and the overgrowth of the transplanted cells continue to be major hurdles that must be overcome. Here, we examined the efficacy of the inducible caspase-9 (iCaspase9) gene as a fail-safe against undesired tumorigenic transformation of iPSC-derived somatic cells. We used a lentiviral vector to transduce iCaspase9 into two iPSC lines and assessed its efficacy in vitro and in vivo. In vitro, the iCaspase9 system induced apoptosis in approximately 95% of both iPSCs and iPSC-derived neural stem/progenitor cells (iPSC-NS/PCs). To determine in vivo function, we transplanted iPSC-NS/PCs into the injured spinal cord of NOD/SCID mice. All transplanted cells whose mass effect was hindering motor function recovery were ablated upon transduction of iCaspase9. Our results suggest that the iCaspase9 system may serve as an important countermeasure against post-transplantation adverse events in stem cell transplant therapies.
Collapse
|
30
|
McErlean EM, McCrudden CM, McCarthy HO. Delivery of nucleic acids for cancer gene therapy: overcoming extra- and intra-cellular barriers. Ther Deliv 2016; 7:619-37. [PMID: 27582234 DOI: 10.4155/tde-2016-0049] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The therapeutic potential of cancer gene therapy has been limited by the difficulty of delivering genetic material to target sites. Various biological and molecular barriers exist which need to be overcome before effective nonviral delivery systems can be applied successfully in oncology. Herein, various barriers are described and strategies to circumvent such obstacles are discussed, considering both the extracellular and intracellular setting. Development of multifunctional delivery systems holds much promise for the progression of gene delivery, and a growing body of evidence supports this approach involving rational design of vectors, with a unique molecular architecture. In addition, the potential application of composite gene delivery platforms is highlighted which may provide an alternative delivery strategy to traditional systemic administration.
Collapse
|
31
|
Hanoun N, Gayral M, Pointreau A, Buscail L, Cordelier P. Initial Characterization of Integrase-Defective Lentiviral Vectors for Pancreatic Cancer Gene Therapy. Hum Gene Ther 2016; 27:184-92. [PMID: 26731312 PMCID: PMC4779299 DOI: 10.1089/hum.2015.151] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 12/23/2015] [Indexed: 12/11/2022] Open
Abstract
The vast majority (85%) of pancreatic ductal adenocarcinomas (PDACs) are discovered at too of a late stage to allow curative surgery. In addition, PDAC is highly resistant to conventional methods of chemotherapy and radiotherapy, which only offer a marginal clinical benefit. Consequently, the prognosis of this cancer is devastating, with a 5-year survival rate of less than 5%. In this dismal context, we recently demonstrated that PDAC gene therapy using nonviral vectors is safe and feasible, with early signs of efficacy in selected patients. Our next step is to transfer to the clinic HIV-1-based lentiviral vectors (LVs) that outshine other therapeutic vectors to treat experimental models of PDAC. However, a primary safety issue presented by LVs that may delay their use in patients is the risk of oncogenesis after vector integration in the host's cell DNA. Thus, we developed a novel anticancerous approach based on integrase-defective lentiviral vectors (IDLVs) and demonstrated that IDLVs can be successfully engineered to transiently deliver therapeutic genes to inhibit pancreatic cancer cells proliferation. This work stems for the use of therapeutic IDLVs for the management of PDAC, in forthcoming early phase gene therapy clinical trial for this disease with no cure.
Collapse
Affiliation(s)
- Naima Hanoun
- Inserm, UMR1037 CRCT, Toulouse, France
- Université Toulouse III-Paul Sabatier, UMR1037 CRCT, Toulouse, France
| | - Marion Gayral
- Inserm, UMR1037 CRCT, Toulouse, France
- Université Toulouse III-Paul Sabatier, UMR1037 CRCT, Toulouse, France
| | - Adeline Pointreau
- Inserm, UMR1037 CRCT, Toulouse, France
- Université Toulouse III-Paul Sabatier, UMR1037 CRCT, Toulouse, France
- Department of Gastroenterology, CHU Toulouse-Rangueil, Toulouse, France
| | - Louis Buscail
- Inserm, UMR1037 CRCT, Toulouse, France
- Université Toulouse III-Paul Sabatier, UMR1037 CRCT, Toulouse, France
- Department of Gastroenterology, CHU Toulouse-Rangueil, Toulouse, France
| | - Pierre Cordelier
- Inserm, UMR1037 CRCT, Toulouse, France
- Université Toulouse III-Paul Sabatier, UMR1037 CRCT, Toulouse, France
| |
Collapse
|
32
|
Zhang H, Zhou X, Xu C, Yang J, Xiang J, Tao M, Xie Y. Synergistic tumor suppression by adenovirus-mediated ING4/PTEN double gene therapy for gastric cancer. Cancer Gene Ther 2015; 23:13-23. [PMID: 26564429 DOI: 10.1038/cgt.2015.59] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 10/12/2015] [Accepted: 10/16/2015] [Indexed: 12/17/2022]
Abstract
Both inhibitor of growth 4 (ING4) and phosphatase and tensin homolog (PTEN) have been shown to be strong candidate tumor suppressors. However, the combined efficacy of ING4 and PTEN for human gastric cancer remains to be determined. In this report, we constructed a multiple promoter expression cassette-based recombinant adenovirus coexpressing ING4 and PTEN (AdVING4/PTEN), assessed the combined effects of AdVING4/PTEN on gastric cancer using wild-type p53 AGS and SNU-1 human gastric cancer cell lines, and elucidated its underlying mechanisms. We found that AdVING4/PTEN-induced synergistic growth inhibition and apoptosis in vitro AGS or SNU-1 tumor cells and in vivo AGS xenografted tumors subcutaneously inoculated in athymic BALB/c nude mice. Mechanistically, AdVING4/PTEN exhibited an enhanced effect on upregulation of p53, Ac-p53 (K382), P21, Bax, PUMA, Noxa, cleaved Caspase-9, cleaved Caspase-3 and cleaved PARP as well as downregulation of Bcl-2 in vitro and in vivo. In addition, AdVING4/PTEN synergistically downregulated tumor vessel CD34 expression and reduced microvessel density, and additively inhibited vascular endothelial growth factor (VEGF) expression in vivo. The synergistic tumor suppression elicited by AdVING4/PTEN was closely associated with the synergistic induction of apoptosis possibly via enhancement of endogenous p53 responses through cooperatively facilitating p53's stability and acetylation, and the synergistic inhibition of tumor angiogenesis probably via overlapping reduction of VEGF through cooperatively downregulating hypoxia inducible factor-1α's level and transcription activity. Thus, our results indicate that cancer gene therapy combining ING4 and PTEN may constitute a novel and effective therapeutic modality for human gastric cancer and other cancers.
Collapse
Affiliation(s)
- H Zhang
- Department of General Surgery, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - X Zhou
- Department of Oncology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - C Xu
- Department of Oncology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - J Yang
- Department of Cell Biology, College of Medicine, Soochow University, Suzhou, China
| | - J Xiang
- Cancer Research Unit, Saskatchewan Cancer Agency, Saskatoon, Saskatchewan, Canada
| | - M Tao
- Department of Oncology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Y Xie
- Department of Oncology, the First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
33
|
Wu J, Zhu Y, Xu C, Xu H, Zhou X, Yang J, Xie Y, Tao M. Adenovirus-mediated p53 and ING4 gene co-transfer elicits synergistic antitumor effects through enhancement of p53 acetylation in breast cancer. Oncol Rep 2015; 35:243-52. [PMID: 26530780 DOI: 10.3892/or.2015.4385] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Accepted: 09/17/2015] [Indexed: 11/06/2022] Open
Abstract
Multigene-based combination therapy may be an effective practice in cancer gene therapy. Substantial studies have demonstrated that tumor suppressor p53 acetylation is indispensable for p53 activation. Inhibitor of growth 4 (ING4), as a novel tumor suppressor, is capable of remarkably enhancing p53 acetylation and its transcriptional activity. Hence, we assumed that combined treatment of p53 and ING4 double tumor suppressors would exhibit enhanced antitumor effects. The combined therapeutic efficacy of p53 and ING4 for human cancers has not been previously reported. We thus generated multiple promoter expression cassette-based recombinant adenovirus-co-expressing ING4 and p53 double tumor suppressor genes (AdVING4/p53), evaluated the combined effects of AdVING4/p53 on breast cancer using the MDA-MB-231 (mutant p53) human breast cancer cell line, and also elucidated its underlying molecular mechanisms. We demonstrated that AdVING4/p53-mediated p53 and ING4 co-expression induced synergistic growth inhibition and apoptosis as well as enhanced effects on upregulation of acetylated p53, P21, Bax, PUMA, Noxa, cleaved caspase-9, cleaved caspase-3 and cleaved PARP, and downregulation of Bcl-2, CD31 and microvessel density (MVD) in MDA-MB-231 breast cancer in vitro and/or in vivo subcutaneous (s.c.) xenografted tumors. The synergistic antitumor activity elicited by AdVING4/p53 was closely associated with the enhanced activation of the intrinsic apoptotic pathway and synergistic inhibition of tumor angiogenesis, very possibly via ING4-mediated enhancement of p53 acetylation and activity. Thus, our results indicate that cancer gene therapy combining two or more tumor suppressors such as p53 and ING4 may constitute a novel and effective therapeutic modality for human breast cancer and other cancers.
Collapse
Affiliation(s)
- Jie Wu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Yanbo Zhu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Chun Xu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Hong Xu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Xiumin Zhou
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Jicheng Yang
- Department of Cell Biology, College of Medicine, Soochow University, Suzhou, Jiangsu, P.R. China
| | - Yufeng Xie
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Min Tao
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| |
Collapse
|
34
|
Ando M, Nishimura T, Yamazaki S, Yamaguchi T, Kawana-Tachikawa A, Hayama T, Nakauchi Y, Ando J, Ota Y, Takahashi S, Nishimura K, Ohtaka M, Nakanishi M, Miles JJ, Burrows SR, Brenner MK, Nakauchi H. A Safeguard System for Induced Pluripotent Stem Cell-Derived Rejuvenated T Cell Therapy. Stem Cell Reports 2015; 5:597-608. [PMID: 26321144 PMCID: PMC4624898 DOI: 10.1016/j.stemcr.2015.07.011] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 07/28/2015] [Accepted: 07/30/2015] [Indexed: 12/21/2022] Open
Abstract
The discovery of induced pluripotent stem cells (iPSCs) has created promising new avenues for therapies in regenerative medicine. However, the tumorigenic potential of undifferentiated iPSCs is a major safety concern for clinical translation. To address this issue, we demonstrated the efficacy of suicide gene therapy by introducing inducible caspase-9 (iC9) into iPSCs. Activation of iC9 with a specific chemical inducer of dimerization (CID) initiates a caspase cascade that eliminates iPSCs and tumors originated from iPSCs. We introduced this iC9/CID safeguard system into a previously reported iPSC-derived, rejuvenated cytotoxic T lymphocyte (rejCTL) therapy model and confirmed that we can generate rejCTLs from iPSCs expressing high levels of iC9 without disturbing antigen-specific killing activity. iC9-expressing rejCTLs exert antitumor effects in vivo. The system efficiently and safely induces apoptosis in these rejCTLs. These results unite to suggest that the iC9/CID safeguard system is a promising tool for future iPSC-mediated approaches to clinical therapy.
Collapse
Affiliation(s)
- Miki Ando
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Toshinobu Nishimura
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | - Satoshi Yamazaki
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Tomoyuki Yamaguchi
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Ai Kawana-Tachikawa
- Division of Infectious Diseases, Advanced Clinical Research Center, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan; AIDS Research Center, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Tomonari Hayama
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Yusuke Nakauchi
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Jun Ando
- Department of Hematology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Yasunori Ota
- Department of Pathology, Research Hospital, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Satoshi Takahashi
- Division of Molecular Therapy, Advanced Clinical Research Center, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Ken Nishimura
- Laboratory of Gene Regulation, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Manami Ohtaka
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki 305-8562, Japan
| | - Mahito Nakanishi
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki 305-8562, Japan
| | - John J Miles
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Brisbane, QLD 4006, Australia
| | - Scott R Burrows
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Brisbane, QLD 4006, Australia
| | - Malcolm K Brenner
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital, Feigin Center, 1102 Bates Avenue, Houston, TX 77030, USA
| | - Hiromitsu Nakauchi
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA.
| |
Collapse
|
35
|
Fang CY, Tsai YD, Lin MC, Wang M, Chen PL, Chao CN, Huang YL, Chang D, Shen CH. Inhibition of Human Bladder Cancer Growth by a Suicide Gene Delivered by JC Polyomavirus Virus-like Particles in a Mouse Model. J Urol 2015; 193:2100-6. [PMID: 25623749 DOI: 10.1016/j.juro.2015.01.084] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2015] [Indexed: 12/23/2022]
Affiliation(s)
- Chiung-Yao Fang
- Department of Medical Research, Chiayi Christian Hospital, Chiayi, Taiwan, Republic of China
| | - Yi-Da Tsai
- Institute of Molecular Biology, National Chung Cheng University, Chiayi, Taiwan, Republic of China
| | - Mien-Chun Lin
- Department of Urology, Chiayi Christian Hospital, Chiayi, Taiwan, Republic of China
- Institute of Molecular Biology, National Chung Cheng University, Chiayi, Taiwan, Republic of China
| | - Meilin Wang
- Department of Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan, Republic of China
| | - Pei-Lain Chen
- Department of Medical Laboratory Science and Biotechnology, Central Taiwan University of Science and Technology, Taichung, Taiwan, Republic of China
| | - Chun-Nun Chao
- Department of Pediatrics, Chiayi Christian Hospital, Chiayi, Taiwan, Republic of China
| | - Yih-Leh Huang
- Department of Medical Research, Buddhist Dalin Tzu Chi General Hospital, Chiayi, Taiwan, Republic of China
| | - Deching Chang
- Institute of Molecular Biology, National Chung Cheng University, Chiayi, Taiwan, Republic of China
| | - Cheng-Huang Shen
- Department of Urology, Chiayi Christian Hospital, Chiayi, Taiwan, Republic of China
| |
Collapse
|
36
|
Xiao-Jie L, Hui-Ying X, Zun-Ping K, Jin-Lian C, Li-Juan J. CRISPR-Cas9: a new and promising player in gene therapy. J Med Genet 2015; 52:289-96. [PMID: 25713109 DOI: 10.1136/jmedgenet-2014-102968] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 02/02/2015] [Indexed: 12/14/2022]
Abstract
First introduced into mammalian organisms in 2013, the RNA-guided genome editing tool CRISPR-Cas9 (clustered regularly interspaced short palindromic repeats/CRISPR-associated nuclease 9) offers several advantages over conventional ones, such as simple-to-design, easy-to-use and multiplexing (capable of editing multiple genes simultaneously). Consequently, it has become a cost-effective and convenient tool for various genome editing purposes including gene therapy studies. In cell lines or animal models, CRISPR-Cas9 can be applied for therapeutic purposes in several ways. It can correct the causal mutations in monogenic disorders and thus rescue the disease phenotypes, which currently represents the most translatable field in CRISPR-Cas9-mediated gene therapy. CRISPR-Cas9 can also engineer pathogen genome such as HIV for therapeutic purposes, or induce protective or therapeutic mutations in host tissues. Moreover, CRISPR-Cas9 has shown potentials in cancer gene therapy such as deactivating oncogenic virus and inducing oncosuppressor expressions. Herein, we review the research on CRISPR-mediated gene therapy, discuss its advantages, limitations and possible solutions, and propose directions for future research, with an emphasis on the opportunities and challenges of CRISPR-Cas9 in cancer gene therapy.
Collapse
Affiliation(s)
- Lu Xiao-Jie
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xue Hui-Ying
- The Reproductive Center, Jiangsu Huai'an Maternity and Children Hospital, Huai'an, China
| | - Ke Zun-Ping
- Department of Cardiology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Chen Jin-Lian
- Department of Gastroenterology, Shanghai Sixth People's Hospital (South), Shanghai Jiaotong University School of Medicine, Shanghai, China Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ji Li-Juan
- Department of Rehabilitation, The Second People's Hospital of Huai'an, Huai'an, China
| |
Collapse
|
37
|
Chen GX, Zhang S, He XH, Liu SY, Ma C, Zou XP. Clinical utility of recombinant adenoviral human p53 gene therapy: current perspectives. Onco Targets Ther 2014; 7:1901-9. [PMID: 25364261 PMCID: PMC4211860 DOI: 10.2147/ott.s50483] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Gene therapy has promised to be a highly effective antitumor treatment by introducing a tumor suppressor gene or the abrogation of an oncogene. Among the potential therapeutic transgenes, the tumor suppressor gene p53 serves as an attractive target. Restoration of wild-type p53 function in tumors can be achieved by introduction of an intact complementary deoxyribonucleic acid copy of the p53 gene using a suitable viral vector, in most cases an adenoviral vector (Adp53). Preclinical in vitro and in vivo studies have shown that Adp53 triggers a dramatic tumor regression response in various cancers. These viruses are engineered to lack certain early proteins and are thus replication defective, including Gendicine, SCH-58500, and Advexin. Several types of tumor-specific p53-expressing conditionally replicating adenovirus vectors (known as replication-competent CRAdp53 vectors) have been developed, such as ONYX 015, AdDelta24-p53, SG600-p53, OBP-702, and H101. Various clinical trials have been conducted to investigate the safety and efficiency of these adenoviral vectors. In this review we will talk about the biological mechanisms, clinical utility, and therapeutic potentials of the replication-deficient Adp53-based and replication-competent CRAdp53-based gene therapy.
Collapse
Affiliation(s)
- Guang-Xia Chen
- Department of Gastroenterology, First People's Hospital of Xuzhou, Xuzhou, Jiangsu Province, People's Republic of China
| | - Shu Zhang
- Department of Gastroenterology, Drum Tower Hospital, Nanjing, People's Republic of China ; Medical School of Nanjing University, Nanjing, People's Republic of China ; Jiangsu Clinical Medical Center of Digestive Disease, Nanjing, People's Republic of China
| | - Xiao-Hua He
- Department of Gastroenterology, First People's Hospital of Xuzhou, Xuzhou, Jiangsu Province, People's Republic of China
| | - Shi-Yu Liu
- Department of Gastroenterology, First People's Hospital of Xuzhou, Xuzhou, Jiangsu Province, People's Republic of China
| | - Chao Ma
- Department of Gastroenterology, Drum Tower Hospital, Nanjing, People's Republic of China ; Medical School of Nanjing University, Nanjing, People's Republic of China ; Jiangsu Clinical Medical Center of Digestive Disease, Nanjing, People's Republic of China
| | - Xiao-Ping Zou
- Department of Gastroenterology, Drum Tower Hospital, Nanjing, People's Republic of China ; Medical School of Nanjing University, Nanjing, People's Republic of China ; Jiangsu Clinical Medical Center of Digestive Disease, Nanjing, People's Republic of China
| |
Collapse
|
38
|
Wang D, Gao G. State-of-the-art human gene therapy: part II. Gene therapy strategies and clinical applications. DISCOVERY MEDICINE 2014; 18:151-161. [PMID: 25227756 PMCID: PMC4440458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
In Part I of this Review (Wang and Gao, 2014), we introduced recent advances in gene delivery technologies and explained how they have powered some of the current human gene therapy applications. In Part II, we expand the discussion on gene therapy applications, focusing on some of the most exciting clinical uses. To help readers to grasp the essence and to better organize the diverse applications, we categorize them under four gene therapy strategies: (1) gene replacement therapy for monogenic diseases, (2) gene addition for complex disorders and infectious diseases, (3) gene expression alteration targeting RNA, and (4) gene editing to introduce targeted changes in host genome. Human gene therapy started with the simple idea that replacing a faulty gene with a functional copy can cure a disease. It has been a long and bumpy road to finally translate this seemingly straightforward concept into reality. As many disease mechanisms unraveled, gene therapists have employed a gene addition strategy backed by a deep knowledge of what goes wrong in diseases and how to harness host cellular machinery to battle against diseases. Breakthroughs in other biotechnologies, such as RNA interference and genome editing by chimeric nucleases, have the potential to be integrated into gene therapy. Although clinical trials utilizing these new technologies are currently sparse, these innovations are expected to greatly broaden the scope of gene therapy in the near future.
Collapse
Affiliation(s)
- Dan Wang
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Guangping Gao
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA
- Department of Microbiology and Physiology Systems, University of Massachusetts Medical School, Worcester, MA 01605, USA
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| |
Collapse
|
39
|
Affiliation(s)
- Helen E Heslop
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, and Houston Methodist Hospital, Houston, Texas, USA
| |
Collapse
|
40
|
|