1
|
Röscheise J, Klimpel M, Govindarajan P, Otte K, Laux H. Unveiling molecular secrets: Analysis of stable lentiviral packaging cell lines enables identification of novel viral gene functions. Gene Ther 2025:10.1038/s41434-025-00533-w. [PMID: 40234566 DOI: 10.1038/s41434-025-00533-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 03/19/2025] [Accepted: 04/03/2025] [Indexed: 04/17/2025]
Abstract
Lentiviral vectors (LVVs) are widely used in gene therapy due to their ability to infect both dividing and non-dividing cells. For LVV production, the creation of stable packaging cell lines with integrated genes necessary for viral replication offer a more consistent and scalable alternative to transient plasmid transfection approach. Although the development of such stable LVV packaging cell lines has been reported, the molecular changes induced by stable and inducible viral gene expression and the impact of genome integrated viral genes on cellular pathways remain poorly characterized. For better insight, we investigated the molecular characteristics of a stable LVV packaging cell line and its host cell line (HEK293T/17) by comparing differential expressed genes. This pathway analysis revealed significant changes in pathway usage between packaging and host cell lines, influenced by different viral transgenes. Gag-pol expression was found to suppress host translational machinery, while rev and VSV-G expression modulated mitochondrial pathways, including oxidative phosphorylation. HIV-1 tat expression, on the other hand, activated histone-related genes. These regulatory shifts suggest a strategic reprogramming of host cellular states to favor viral replication, curbing protein synthesis and energy production to levels that support viral assembly but impair the host's immune defense and the production of immune-related proteins. Our findings provide a deeper understanding of the molecular changes associated with stable viral gene expression, which can inform the optimization of LVV production in gene therapy applications.
Collapse
Affiliation(s)
- Jona Röscheise
- Institute of Applied Biotechnology, Biberach University of Applied Sciences, Biberach, Germany.
| | - Maximilian Klimpel
- Biopharmaceutical Product Development, CSL Behring Innovation GmbH, Marburg, Germany
| | | | - Kerstin Otte
- Institute of Applied Biotechnology, Biberach University of Applied Sciences, Biberach, Germany
| | - Holger Laux
- Biopharmaceutical Product Development, CSL Behring Innovation GmbH, Marburg, Germany
| |
Collapse
|
2
|
Parman T, Pizzurro DM, Lucas J, Peng Z. Mutagenesis and Carcinogenesis Risk Evaluation for AAV and Lentiviral Gene Therapies. Int J Toxicol 2025:10915818251318248. [PMID: 39935281 DOI: 10.1177/10915818251318248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025]
Abstract
Fueled by the identification and invention of novel gene delivery vectors, gene therapy efforts now hold promise for treating a wide range of diseases and are seen as a crucial part of growth for the biopharmaceutical industry. Currently, recombinant adeno-associated virus vectors (rAAVs) and lentiviral vectors (LVs) are the main vectors used in gene therapies that are approved or tested in human clinical trials. Meanwhile, ongoing research continuously reveals unprecedented knowledge of viral vectors on the host genome, which may subsequently affect the mutagenic and carcinogenic potential of these therapies. This article summarizes the content and addresses the commentary from the scientific symposium entitled "Mutagenesis and Carcinogenesis Risk Evaluation for AAV and Lentiviral Gene Therapies," conducted at the 43rd Annual Meeting of the American College of Toxicology, November 2022 in Denver, CO. The objective is to summarize the current understanding of rAAV and LV related mutagenicity/carcinogenicity risk, describe the methods and interpretation of results to guide risk assessments, as well as the current regulatory landscape on the carcinogenicity and mutagenicity assessment of rAAV and LV gene therapy products.
Collapse
Affiliation(s)
| | | | | | - Zhechu Peng
- Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, USA
| |
Collapse
|
3
|
Khiabani NA, Doustvandi MA, Story D, Nobari SA, Hajizadeh M, Petersen R, Dunbar G, Rossignol J. Glioblastoma therapy: State of the field and future prospects. Life Sci 2024; 359:123227. [PMID: 39537100 DOI: 10.1016/j.lfs.2024.123227] [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: 06/25/2024] [Revised: 09/03/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Glioblastoma (GB) is a cancerous brain tumor that originates from glial cells and leads to thousands of deaths each year and a five-year survival of only 6.8 %. Treatments for GB include surgery, chemotherapy, radiation, and immunotherapy. GB is an incurable fatal disease, necessitating the development of innovative strategies to find a developing effective therapy. Genetic therapies may be crucial in treating GB by identifying the mutations and amplifications of multiple genes, which drive its proliferation and spread. Use of small interfering RNAs (siRNAs) provides a novel technology used to suppress the genes associated with disease, which forms a basis for targeted therapy in GB and its stem cell population, which are recognized for their ability to develop resistance to chemotherapy and tumorigenic capabilities. This review examines the use of siRNAs in GB, emphasizing their effectiveness in suppressing key oncogenes and signaling pathways associated with tumor development, invasion, stemness, and resistance to standard treatments. siRNA-based gene silencing is a promising approach for developing targeted therapeutics against GB and associated stem cell populations, potentially enhancing patient outcomes and survival rates in this devastating disease.
Collapse
Affiliation(s)
- Nadia Allahyarzadeh Khiabani
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, USA; Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, USA; College of Medicine, Central Michigan University, Mount Pleasant, MI, USA
| | | | - Darren Story
- Department of Psychology, Saginaw Valley State University, University Center, MI 48710, USA
| | | | | | - Robert Petersen
- College of Medicine, Central Michigan University, Mount Pleasant, MI, USA
| | - Gary Dunbar
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, USA; Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, USA; Department of Psychology, Central Michigan University, Mount Pleasant, MI, USA
| | - Julien Rossignol
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, USA; Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, USA; College of Medicine, Central Michigan University, Mount Pleasant, MI, USA.
| |
Collapse
|
4
|
de Morais CCPDL, Correia EM, Bonamino MH, de Vasconcelos ZFM. Cell-Penetrating Peptides and CRISPR-Cas9: A Combined Strategy for Human Genetic Disease Therapy. Hum Gene Ther 2024; 35:781-797. [PMID: 39276086 PMCID: PMC11511780 DOI: 10.1089/hum.2024.020] [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: 02/09/2024] [Accepted: 08/29/2024] [Indexed: 09/16/2024] Open
Abstract
The advent of clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated nuclease 9 (Cas9) technology has revolutionized the field of genetic engineering, offering unprecedented potential for the targeted manipulation of DNA sequences. Advances in the mechanism of action of the CRISPR-Cas9 system allowed potential applicability for the treatment of genetic diseases. CRISPR-Cas9's mechanism of action involves the use of an RNA guide molecule to target-specific DNA sequences and the Cas9 enzyme to induce precise DNA cleavage. In the context of the CRISPR-Cas9 system, this review covers nonviral delivery methods for gene editing based on peptide internalization. Here, we describe critical areas of discussion such as immunogenicity, emphasizing the importance of safety, efficiency, and cost-effectiveness, particularly in the context of treating single-mutation genetic diseases using advanced editing techniques genetics as prime editor and base editor. The text discusses the versatility of cell-penetrating peptides (CPPs) in forming complexes for delivering biomolecules, particularly ribonucleoprotein for genome editing with CRISPR-Cas9 in human cells. In addition, it emphasizes the promise of combining CPPs with DNA base editing and prime editing systems. These systems, known for their simplicity and precision, hold great potential for correcting point mutations in human genetic diseases. In summary, the text provides a clear overview of the advantages of using CPPs for genome editing with CRISPR-Cas9, particularly in conjunction with advanced editing systems, highlighting their potential impact on clinical applications in the treatment of single-mutation genetic diseases. [Figure: see text].
Collapse
Affiliation(s)
- Carla Cristina Pedrosa de Lira de Morais
- Cell Processing Center/Umbilical and Placental Cord Blood Bank (CPC/BSCUP), Bone Marrow Transplant Center (CEMO), National Cancer Institute (INCA), Rio de Janeiro, Brazil
- Oswaldo Cruz Foundation (FIOCRUZ), National Institute of Women, Children and Adolescents’ Health Fernandes Figueira (IFF), Rio de Janeiro, Brazil
| | - Eduardo Mannarino Correia
- Cell and Gene Therapy Program, Research Coordination, National Cancer Institute (INCA), Rio de Janeiro, Brazil
| | - Martín Hernán Bonamino
- Cell and Gene Therapy Program, Research Coordination, National Cancer Institute (INCA), Rio de Janeiro, Brazil
| | - Zilton Farias Meira de Vasconcelos
- Oswaldo Cruz Foundation (FIOCRUZ), National Institute of Women, Children and Adolescents’ Health Fernandes Figueira (IFF), Rio de Janeiro, Brazil
| |
Collapse
|
5
|
Mohd Abas MD, Mohd Asri MF, Yusafawi NAS, Rosman NAZ, Baharudin NAZ, Taher M, Susanti D, Khotib J. Advancements of gene therapy in cancer treatment: A comprehensive review. Pathol Res Pract 2024; 261:155509. [PMID: 39121791 DOI: 10.1016/j.prp.2024.155509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 07/23/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024]
Abstract
Cancer is the main contributor for mortality in the world. Conventional therapy that available as the treatment options are chemotherapy, radiotherapy and surgery. However, these treatments are hardly cell-specific most of the time. Nowadays, extensive research and investigations are made to develop cell-specific approaches prior to cancer treatment. Some of them are photodynamic therapy, hyperthermia, immunotherapy, stem cell transplantation and targeted therapy. This review article will be focusing on the development of gene therapy in cancer. The objective of gene therapy is to correct specific mutant genes causing the excessive proliferation of the cell that leads to cancer. There are lots of explorations in the approach to modify the gene. The delivery of this therapy plays a big role in its success. If the inserted gene does not find its way to the target, the therapy is considered a failure. Hence, vectors are needed and the common vectors used are viral, non viral or synthetic, polymer based and lipid based vectors. The advancement of gene therapy in cancer treatment will be focussing on the top three cancer cases in the world which are breast, lung and colon cancer. In breast cancer, the discussed therapy are CRISPR/Cas9, siRNA and gene silencing whereas in colon cancer miRNA and suicide gene therapy and in lung cancer, replacement of tumor suppressor gene, CRISPR/Cas9 and miRNA.
Collapse
Affiliation(s)
- Muhammad Dhiyauddin Mohd Abas
- Department of Pharmaceutical Technology, Faculty of Pharmacy, International Islamic University Malaysia, Jalan Sultan Ahmad Shah, Kuantan, Pahang 25200, Malaysia
| | - Muhammad Fareez Mohd Asri
- Department of Pharmaceutical Technology, Faculty of Pharmacy, International Islamic University Malaysia, Jalan Sultan Ahmad Shah, Kuantan, Pahang 25200, Malaysia
| | - Nur Anis Suffiah Yusafawi
- Department of Pharmaceutical Technology, Faculty of Pharmacy, International Islamic University Malaysia, Jalan Sultan Ahmad Shah, Kuantan, Pahang 25200, Malaysia
| | - Nur Anis Zahra Rosman
- Department of Pharmaceutical Technology, Faculty of Pharmacy, International Islamic University Malaysia, Jalan Sultan Ahmad Shah, Kuantan, Pahang 25200, Malaysia
| | - Nur Arifah Zahidah Baharudin
- Department of Pharmaceutical Technology, Faculty of Pharmacy, International Islamic University Malaysia, Jalan Sultan Ahmad Shah, Kuantan, Pahang 25200, Malaysia
| | - Muhammad Taher
- Department of Pharmaceutical Technology, Faculty of Pharmacy, International Islamic University Malaysia, Jalan Sultan Ahmad Shah, Kuantan, Pahang 25200, Malaysia.
| | - Deny Susanti
- Department of Chemistry, Faculty of Science, International Islamic University Malaysia, Jalan Sultan Ahmad Shah, Kuantan, Pahang 25200, Malaysia.
| | - Junaidi Khotib
- Department of Pharmacy Practice, Faculty of Pharmacy, Airlangga University, Surabaya 60115, Indonesia.
| |
Collapse
|
6
|
Perry C, Mujahid N, Takeuchi Y, Rayat ACME. Insights into product and process related challenges of lentiviral vector bioprocessing. Biotechnol Bioeng 2024; 121:2466-2481. [PMID: 37526313 DOI: 10.1002/bit.28498] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/28/2023] [Accepted: 07/07/2023] [Indexed: 08/02/2023]
Abstract
Lentiviral vectors (LVs) are used in advanced therapies to transduce recipient cells for long term gene expression for therapeutic benefit. The vector is commonly pseudotyped with alternative viral envelope proteins to improve tropism and is selected for enhanced functional titers. However, their impact on manufacturing and the success of individual bioprocessing unit operations is seldom demonstrated. To the best of our knowledge, this is the first study on the processability of different Lentiviral vector pseudotypes. In this work, we compared three envelope proteins commonly pseudotyped with LVs across manufacturing conditions such as temperature and pump flow and across steps common to downstream processing. We have shown impact of filter membrane chemistry on vector recoveries with differing envelopes during clarification and observed complete vector robustness in high shear manufacturing environments using ultra scale-down technologies. The impact of shear during membrane filtration in a tangential flow filtration-mimic showed the benefit of employing higher shear rates, than currently used in LV production, to increase vector recovery. Likewise, optimized anion exchange chromatography purification in monolith format was determined. The results contradict a common perception that lentiviral vectors are susceptible to shear or high salt concentration (up to 1.7 M). This highlights the prospects of improving LV recovery by evaluating manufacturing conditions that contribute to vector losses for specific production systems.
Collapse
Affiliation(s)
- Christopher Perry
- Department of Biochemical Engineering, University College London, London, UK
- Division of Infection and Immunology, University College London, London, UK
- Biotherapeutics and Advanced Therapies, Scientific Research and Innovation, Medicines and Healthcare Products Regulatory Agency, South Mimms, Potters Bar, UK
| | - Noor Mujahid
- Department of Biochemical Engineering, University College London, London, UK
| | - Yasu Takeuchi
- Division of Infection and Immunology, University College London, London, UK
- Biotherapeutics and Advanced Therapies, Scientific Research and Innovation, Medicines and Healthcare Products Regulatory Agency, South Mimms, Potters Bar, UK
| | - Andrea C M E Rayat
- Department of Biochemical Engineering, University College London, London, UK
| |
Collapse
|
7
|
Mier NC, Roper DK. Effects of an indole derivative on cell proliferation, transfection, and alternative splicing in production of lentiviral vectors by transient co-transfection. PLoS One 2024; 19:e0297817. [PMID: 38833479 PMCID: PMC11149887 DOI: 10.1371/journal.pone.0297817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 01/12/2024] [Indexed: 06/06/2024] Open
Abstract
Lentiviral vectors derived from human immunodeficiency virus type I are widely used to deliver functional gene copies to mammalian cells for research and gene therapies. Post-transcriptional splicing of lentiviral vector transgene in transduced host and transfected producer cells presents barriers to widespread application of lentiviral vector-based therapies. The present study examined effects of indole derivative compound IDC16 on splicing of lentiviral vector transcripts in producer cells and corresponding yield of infectious lentiviral vectors. Indole IDC16 was shown previously to modify alternative splicing in human immunodeficiency virus type I. Human embryonic kidney 293T cells were transiently transfected by 3rd generation backbone and packaging plasmids using polyethyleneimine. Reverse transcription-quantitative polymerase chain reaction of the fraction of unspliced genomes in human embryonic kidney 293T cells increased up to 31% upon the indole's treatment at 2.5 uM. Corresponding yield of infectious lentiviral vectors decreased up to 4.5-fold in a cell transduction assay. Adjusting timing and duration of IDC16 treatment indicated that the indole's disruption of early stages of transfection and cell cycle had a greater effect on exponential time course of lentiviral vector production than its reduction of post-transcriptional splicing. Decrease in transfected human embryonic kidney 293T proliferation by IDC16 became significant at 10 uM. These findings indicated contributions by early-stage transfection, cell proliferation, and post-transcriptional splicing in transient transfection of human embryonic kidney 293T cells for lentiviral vector production.
Collapse
Affiliation(s)
- Nataly Carolina Mier
- Department of Biological Engineering, Utah State University, Logan, Utah, United States of America
| | - Donald Keith Roper
- Department of Biological Engineering, Utah State University, Logan, Utah, United States of America
| |
Collapse
|
8
|
Prasher P, Sharma M, Agarwal V, Singh SK, Gupta G, Dureja H, Dua K. Cationic cycloamylose based nucleic acid nanocarriers. Chem Biol Interact 2024; 395:111000. [PMID: 38614318 DOI: 10.1016/j.cbi.2024.111000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 04/02/2024] [Accepted: 04/07/2024] [Indexed: 04/15/2024]
Abstract
Nucleic acid delivery by viral and non-viral methods has been a cornerstone for the contemporary gene therapy aimed at correcting the defective genes, replacing of the missing genes, or downregulating the expression of anomalous genes is highly desirable for the management of various diseases. Ostensibly, it becomes paramount for the delivery vectors to intersect the biological barriers for accessing their destined site within the cellular environment. However, the lipophilic nature of biological membranes and their potential to limit the entry of large sized, charged, hydrophilic molecules thus presenting a sizeable challenge for the cellular integration of negatively charged nucleic acids. Furthermore, the susceptibility of nucleic acids towards the degrading enzymes (nucleases) in the lysosomes present in cytoplasm is another matter of concern for their cellular and nuclear delivery. Hence, there is a pressing need for the identification and development of cationic delivery systems which encapsulate the cargo nucleic acids where the charge facilitates their cellular entry by evading the membrane barriers, and the encapsulation shields them from the enzymatic attack in cytoplasm. Cycloamylose bearing a closed loop conformation presents a robust candidature in this regard owing to its remarkable encapsulating tendency towards nucleic acids including siRNA, CpG DNA, and siRNA. The presence of numerous hydroxyl groups on the cycloamylose periphery provides sites for its chemical modification for the introduction of cationic groups, including spermine, (3-Chloro-2 hydroxypropyl) trimethylammonium chloride (Q188), and diethyl aminoethane (DEAE). The resulting cationic cycloamylose possesses a remarkable transfection efficiency and provides stability to cargo oligonucleotides against endonucleases, in addition to modulating the undesirable side effects such as unwanted immune stimulation. Cycloamylose is known to interact with the cell membranes where they release certain membrane components such as phospholipids and cholesterol thereby resulting in membrane destabilization and permeabilization. Furthermore, cycloamylose derivatives also serve as formulation excipients for improving the efficiency of other gene delivery systems. This review delves into the various vector and non-vector-based gene delivery systems, their advantages, and limitations, eventually leading to the identification of cycloamylose as an ideal candidate for nucleic acid delivery. The synthesis of cationic cycloamylose is briefly discussed in each section followed by its application for specific delivery/transfection of a particular nucleic acid.
Collapse
Affiliation(s)
- Parteek Prasher
- Department of Chemistry, University of Petroleum & Energy Studies, Energy Acres, Dehradun, 248007, India.
| | - Mousmee Sharma
- Department of Chemistry, Uttaranchal University, Dehradun, 248007, India
| | - Vipul Agarwal
- Cluster for Advanced Macromolecular Design (CAMD), School of Chemical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India; Faculty of Health, Australian Research Center in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia; School of Medical and Life Sciences, Sunway University, 47500 Sunway City, Malaysia
| | - Gaurav Gupta
- School of Pharmacy, Graphic Era Hill University, Dehradun, 248007, India; Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman 346, United Arab Emirates
| | - Harish Dureja
- Department of Pharmaceutical Sciences, Maharishi Dayanand University, Rohtak, 124001, India
| | - Kamal Dua
- Faculty of Health, Australian Research Center in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
| |
Collapse
|
9
|
Rathod RJ, Sukumaran RK, Kedia N, Kumar J, Nair R, Chandy M, Gandikota L, Radhakrishnan VS. Chimeric Antigen Receptor T-cell based cellular therapies for cancer: An introduction and Indian perspective. Indian J Cancer 2024; 61:204-214. [PMID: 39152647 DOI: 10.4103/ijc.ijc_433_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 11/19/2021] [Indexed: 08/19/2024]
Abstract
Using one's own immune system for curing cancer has been an active field of research in cancer biology and therapeutics. One such opportunity in cellular immunotherapy is adoptive cell transfers. With the recent approval of CAR-T therapy as a cancer treatment, a whole new paradigm of cancer treatment has opened-up, with a ray of hope for relapsed/refractory cancer patients. Despite promising clinical outcomes, the therapy is in its early phase and remains out of reach for most patients due to its high cost and logistic challenges. In India, these therapies are unavailable and further confounded by the economic challenges and a large population. In this review, we discuss various aspects of T-cell immunotherapies with a special focus on CAR-T in the Indian scenario. We touch upon the basic scientific aspects, mechanism of action, manufacturing, clinical aspects and commercial aspects of the CAR-Tcell therapies and its future worldwide and in India.
Collapse
Affiliation(s)
- Reena J Rathod
- Cell and Gene Therapy Division, Intas Pharmaceuticals, Ahmedabad, Gujarat, India
| | - Reghu K Sukumaran
- Hematology Oncology and HCT, Tata Medical Center, Kolkata, West Bengal, India
| | - Neelam Kedia
- Cell and Gene Therapy Division, Intas Pharmaceuticals, Ahmedabad, Gujarat, India
| | - Jeevan Kumar
- Hematology Oncology and HCT, Tata Medical Center, Kolkata, West Bengal, India
| | - Reena Nair
- Hematology Oncology and HCT, Tata Medical Center, Kolkata, West Bengal, India
| | - Mammen Chandy
- Hematology Oncology and HCT, Tata Medical Center, Kolkata, West Bengal, India
| | | | | |
Collapse
|
10
|
Lu B, Lim JM, Yu B, Song S, Neeli P, Sobhani N, K P, Bonam SR, Kurapati R, Zheng J, Chai D. The next-generation DNA vaccine platforms and delivery systems: advances, challenges and prospects. Front Immunol 2024; 15:1332939. [PMID: 38361919 PMCID: PMC10867258 DOI: 10.3389/fimmu.2024.1332939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 01/17/2024] [Indexed: 02/17/2024] Open
Abstract
Vaccines have proven effective in the treatment and prevention of numerous diseases. However, traditional attenuated and inactivated vaccines suffer from certain drawbacks such as complex preparation, limited efficacy, potential risks and others. These limitations restrict their widespread use, especially in the face of an increasingly diverse range of diseases. With the ongoing advancements in genetic engineering vaccines, DNA vaccines have emerged as a highly promising approach in the treatment of both genetic diseases and acquired diseases. While several DNA vaccines have demonstrated substantial success in animal models of diseases, certain challenges need to be addressed before application in human subjects. The primary obstacle lies in the absence of an optimal delivery system, which significantly hampers the immunogenicity of DNA vaccines. We conduct a comprehensive analysis of the current status and limitations of DNA vaccines by focusing on both viral and non-viral DNA delivery systems, as they play crucial roles in the exploration of novel DNA vaccines. We provide an evaluation of their strengths and weaknesses based on our critical assessment. Additionally, the review summarizes the most recent advancements and breakthroughs in pre-clinical and clinical studies, highlighting the need for further clinical trials in this rapidly evolving field.
Collapse
Affiliation(s)
- Bowen Lu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jing Ming Lim
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Boyue Yu
- Department of Environmental Science, Policy, and Management, University of California at Berkeley, Berkeley, CA, United States
| | - Siyuan Song
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Praveen Neeli
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Navid Sobhani
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Pavithra K
- School of Chemistry, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram, India
| | - Srinivasa Reddy Bonam
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
| | - Rajendra Kurapati
- School of Chemistry, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram, India
| | - Junnian Zheng
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dafei Chai
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
11
|
Fischer A, Ehrlich A, Plotkin Y, Ouyang Y, Asulin K, Konstantinos I, Fan C, Nahmias Y, Willner I. Stimuli-Responsive Hydrogel Microcapsules Harnessing the COVID-19 Immune Response for Cancer Therapeutics. Angew Chem Int Ed Engl 2023; 62:e202311590. [PMID: 37675854 DOI: 10.1002/anie.202311590] [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: 08/09/2023] [Revised: 09/04/2023] [Accepted: 09/05/2023] [Indexed: 09/08/2023]
Abstract
The combination of gene therapy and immunotherapy concepts, along recent advances in DNA nanotechnology, have the potential to provide important tools for cancer therapies. We present the development of stimuli-responsive microcapsules, loaded with a viral immunogenetic agent, harnessing the immune response against the Coronavirus Disease 2019, COVID-19, to selectively attack liver cancer cells (hepatoma) or recognize breast cancer or hepatoma, by expression of green fluorescence protein, GFP. The pH-responsive microcapsules, modified with DNA-tetrahedra nanostructures, increased hepatoma permeation by 50 %. Incorporation of a GFP-encoding lentivirus vector inside the tumor-targeting pH-stimulated miRNA-triggered and Alpha-fetoprotein-dictated microcapsules enables the demonstration of neoplasm selectivity, with approximately 5,000-, 8,000- and 50,000-fold more expression in the cancerous cells, respectively. The incorporation of the SARS-CoV-2 spike protein in the gene vector promotes specific recognition of the immune-evading hepatoma by the COVID-19-analogous immune response, which leads to cytotoxic and inflammatory activity, mediated by serum components taken from vaccinated or recovered COVID-19 patients, resulting in effective elimination of the hepatoma (>85 % yield).
Collapse
Affiliation(s)
- Amit Fischer
- Institute of Chemistry, The Minerva Center for Bio-hybrid Complex Systems, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| | - Avner Ehrlich
- Grass Center for Bioengineering, Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| | - Yevgeni Plotkin
- The Department of Anesthesiology, Critical Care and Pain Medicine, Hadassah University Hospital, Jerusalem, 9112001, Israel
- Faculty of Medicine, Hebrew University of Jerusalem Jerusalem, 9112001, (Israel)
| | - Yu Ouyang
- Institute of Chemistry, The Minerva Center for Bio-hybrid Complex Systems, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| | - Klil Asulin
- Grass Center for Bioengineering, Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| | - Ioannidis Konstantinos
- Grass Center for Bioengineering, Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yaakov Nahmias
- Grass Center for Bioengineering, Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| | - Itamar Willner
- Institute of Chemistry, The Minerva Center for Bio-hybrid Complex Systems, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| |
Collapse
|
12
|
He Z, Kwee EJ, Cleveland MH, Cole KD, Lin-Gibson S, He HJ. Quantitation and integrity evaluation of RNA genome in lentiviral vectors by direct reverse transcription-droplet digital PCR (direct RT-ddPCR). Sci Rep 2023; 13:14470. [PMID: 37660227 PMCID: PMC10475045 DOI: 10.1038/s41598-023-41644-x] [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: 06/05/2023] [Accepted: 08/29/2023] [Indexed: 09/04/2023] Open
Abstract
Lentiviral vectors (LV) have proven to be powerful tools for stable gene delivery in both dividing and non-dividing cells. Approval of these LVs for use in clinical applications has been achieved by improvements in LV design. Critically important characteristics concerning quality control are LV titer quantification and the detection of impurities. However, increasing evidence concerning high variability in titration assays indicates poor harmonization of the methods undertaken to date. In this study, we developed a direct reverse transcription droplet digital PCR (Direct RT-ddPCR) approach without RNA extraction and purification for estimation of LV titer and RNA genome integrity. The RNA genome integrity was assessed by RT-ddPCR assays targeted to four distant regions of the LV genome. Results of the analyses showed that direct RT-ddPCR without RNA extraction and purification performs similarly to RT-ddPCR on purified RNA from 3 different LV samples, in terms of robustness and assay variance. Interestingly, these RNA titer results were comparable to physical titers by p24 antigen ELISA (enzyme-linked immunosorbent assay). Moreover, we confirmed the partial degradation or the incomplete RNA genomes in the prepared 3 LV samples. These results may partially explain the discrepancy of the LV particle titers to functional titers. This work not only demonstrates the feasibility of direct RT-ddPCR in determining LV titers, but also provides a method that can be easily adapted for RNA integrity assessment.
Collapse
Affiliation(s)
- Zhiyong He
- Material Measurement Laboratory, National Institute of Standards and Technology, 100 Bureau Drive, MS 8312, Gaithersburg, MD, 20899, USA.
| | - Edward J Kwee
- Material Measurement Laboratory, National Institute of Standards and Technology, 100 Bureau Drive, MS 8312, Gaithersburg, MD, 20899, USA
| | - Megan H Cleveland
- Material Measurement Laboratory, National Institute of Standards and Technology, 100 Bureau Drive, MS 8312, Gaithersburg, MD, 20899, USA
| | - Kenneth D Cole
- Material Measurement Laboratory, National Institute of Standards and Technology, 100 Bureau Drive, MS 8312, Gaithersburg, MD, 20899, USA
| | - Sheng Lin-Gibson
- Material Measurement Laboratory, National Institute of Standards and Technology, 100 Bureau Drive, MS 8312, Gaithersburg, MD, 20899, USA
| | - Hua-Jun He
- Material Measurement Laboratory, National Institute of Standards and Technology, 100 Bureau Drive, MS 8312, Gaithersburg, MD, 20899, USA.
| |
Collapse
|
13
|
Cheng Q, Kang Y, Yao B, Dong J, Zhu Y, He Y, Ji X. Genetically Engineered-Cell-Membrane Nanovesicles for Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302131. [PMID: 37409429 PMCID: PMC10502869 DOI: 10.1002/advs.202302131] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/13/2023] [Indexed: 07/07/2023]
Abstract
The advent of immunotherapy has marked a new era in cancer treatment, offering significant clinical benefits. Cell membrane as drug delivery materials has played a crucial role in enhancing cancer therapy because of their inherent biocompatibility and negligible immunogenicity. Different cell membranes are prepared into cell membrane nanovesicles (CMNs), but CMNs have limitations such as inefficient targeting ability, low efficacy, and unpredictable side effects. Genetic engineering has deepened the critical role of CMNs in cancer immunotherapy, enabling genetically engineered-CMN (GCMN)-based therapeutics. To date, CMNs that are surface modified by various functional proteins have been developed through genetic engineering. Herein, a brief overview of surface engineering strategies for CMNs and the features of various membrane sources is discussed, followed by a description of GCMN preparation methods. The application of GCMNs in cancer immunotherapy directed at different immune targets is addressed as are the challenges and prospects of GCMNs in clinical translation.
Collapse
Affiliation(s)
| | - Yong Kang
- Academy of Medical Engineering and Translational MedicineMedical CollegeTianjin UniversityTianjin300072China
| | - Bin Yao
- Academy of Medical Engineering and Translational MedicineMedical CollegeTianjin UniversityTianjin300072China
| | - Jinrui Dong
- Academy of Medical Engineering and Translational MedicineMedical CollegeTianjin UniversityTianjin300072China
| | - Yalan Zhu
- Jinhua Municipal Central HospitalJinhua321000China
| | - Yiling He
- Jinhua Municipal Central HospitalJinhua321000China
| | - Xiaoyuan Ji
- Academy of Medical Engineering and Translational MedicineMedical CollegeTianjin UniversityTianjin300072China
- Medical CollegeLinyi UniversityLinyi276000China
| |
Collapse
|
14
|
Bomb K, LeValley PJ, Woodward I, Cassel SE, Sutherland BP, Bhattacharjee A, Yun Z, Steen J, Kurdzo E, McCoskey J, Burris D, Levine K, Carbrello C, Lenhoff AM, Fromen CA, Kloxin AM. Cell therapy biomanufacturing: integrating biomaterial and flow-based membrane technologies for production of engineered T-cells. ADVANCED MATERIALS TECHNOLOGIES 2023; 8:2201155. [PMID: 37600966 PMCID: PMC10437131 DOI: 10.1002/admt.202201155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Indexed: 08/22/2023]
Abstract
Adoptive T-cell therapies (ATCTs) are increasingly important for the treatment of cancer, where patient immune cells are engineered to target and eradicate diseased cells. The biomanufacturing of ATCTs involves a series of time-intensive, lab-scale steps, including isolation, activation, genetic modification, and expansion of a patient's T-cells prior to achieving a final product. Innovative modular technologies are needed to produce cell therapies at improved scale and enhanced efficacy. In this work, well-defined, bioinspired soft materials were integrated within flow-based membrane devices for improving the activation and transduction of T cells. Hydrogel coated membranes (HCM) functionalized with cell-activating antibodies were produced as a tunable biomaterial for the activation of primary human T-cells. T-cell activation utilizing HCMs led to highly proliferative T-cells that expressed a memory phenotype. Further, transduction efficiency was improved by several fold over static conditions by using a tangential flow filtration (TFF) flow-cell, commonly used in the production of protein therapeutics, to transduce T-cells under flow. The combination of HCMs and TFF technology led to increased cell activation, proliferation, and transduction compared to current industrial biomanufacturing processes. The combined power of biomaterials with scalable flow-through transduction techniques provides future opportunities for improving the biomanufacturing of ATCTs.
Collapse
Affiliation(s)
- Kartik Bomb
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE
| | - Paige J. LeValley
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE
| | - Ian Woodward
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE
| | - Samantha E. Cassel
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE
| | | | | | - Zaining Yun
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE
| | - Jonathan Steen
- EMD Millipore Corporation, Bedford, MA, an affiliate of Merck, Newark, DE
| | - Emily Kurdzo
- EMD Millipore Corporation, Bedford, MA, an affiliate of Merck, Newark, DE
| | - Jacob McCoskey
- EMD Millipore Corporation, Bedford, MA, an affiliate of Merck, Newark, DE
| | - David Burris
- Mechanical Engineering, University of Delaware, Newark, DE
| | - Kara Levine
- EMD Millipore Corporation, Bedford, MA, an affiliate of Merck, Newark, DE
| | | | - Abraham M. Lenhoff
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE
| | | | - April M. Kloxin
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE
- Material Science and Engineering, University of Delaware, Newark, DE
| |
Collapse
|
15
|
Bell-Hensley A, Zheng H, McAlinden A. Modulation of MicroRNA Expression During In Vitro Chondrogenesis. Methods Mol Biol 2023; 2598:197-215. [PMID: 36355294 PMCID: PMC10069062 DOI: 10.1007/978-1-0716-2839-3_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Since their discovery in 1993, microRNAs (miRNAs) are now recognized as important epigenetic regulators of many mammalian cellular processes including proliferation, apoptosis, metabolism, and differentiation. These small non-coding RNAs function by interacting with specific regions in the 3'-untranslated region of mRNAs, thereby resulting in mRNA degradation or suppression of translation. Since miRNAs have the ability to target many mRNAs within a given cell type, a number of cellular pathways and networks may be regulated as a result. To study the function of miRNAs, a number of methods can be used to modulate their activity in cells such as synthetic mimics or antagomirs for short-term assays or viral-based approaches for longer-term experiments such as cell differentiation assays. In this chapter, we provide our methodology to constitutively overexpress a desired miRNA during in vitro chondrogenesis of human cartilage progenitor cells (CPCs). Specifically, we describe how we obtain CPCs from human articular cartilage specimens, how we generate and titrate lentivirus engineered to overexpress a precursor miRNA, how we transduce CPCs with lentivirus and differentiate them toward the chondrocyte lineage, and how we extract RNA and measure expression levels of the miRNA of interest during in vitro chondrogenesis. We also provide some data from our laboratory demonstrating that we can achieve and maintain miRNA overexpression for up to 14 days in cartilage pellet cultures. We predict that these lentiviral-based approaches will also be useful to study how miRNA modulation of progenitor cells affects cell differentiation and extracellular matrix production within three-dimensional biomaterial scaffolds.
Collapse
Affiliation(s)
- Austin Bell-Hensley
- Department of Biomedical Engineering, Washington University, St Louis, MO, USA
| | - Hongjun Zheng
- Department of Orthopaedic Surgery, Washington University School of Medicine, St Louis, MO, USA
| | - Audrey McAlinden
- Department of Orthopaedic Surgery, Washington University School of Medicine, St Louis, MO, USA.
- Department of Cell Biology & Physiology, Washington University School of Medicine, St Louis, MO, USA.
- Shriners Hospitals for Children - St Louis, St Louis, MO, USA.
| |
Collapse
|
16
|
Bhuniya A, Pattarayan D, Yang D. Lentiviral vector transduction provides nonspecific immunogenicity for syngeneic tumor models. Mol Carcinog 2022; 61:1073-1081. [PMID: 36161729 DOI: 10.1002/mc.23467] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 07/31/2022] [Accepted: 09/09/2022] [Indexed: 11/07/2022]
Abstract
Lentivirus-based transduction systems are widely used in biological science and cancer biology, including cancer immunotherapy. However, in in vivo transplanted tumor model, the immunogenicity of these transduced cells was not appropriately addressed. Here, we used empty vector-transduced mouse melanoma (B16) and carcinoma (lewis lung carcinoma) cells transplanted tumor model to study the immune response due to the transduction processes. We showed that the overall in vivo tumor growth rate gets reduced in transduced cells only in immune-competent mice but not in nude mice. This data indicate the involvement of the immune system in the in vivo tumor growth restriction in the transduced group. Further studies showed that specific activation of CD8+ T cells might be responsible for restricted tumor growth. Mechanistically, transduced tumor cells show the higher activity of type I interferon, which might play an essential role in this activation. Overall, our data indicate the modulation of the immune system by lentiviral vector transduced tumor cells, which required further studies to explore the mechanisms and better understand the biological significance. Our data also indicate the importance of considering the immunogenicity of transduced cells when analyzing in vivo results, especially in studies related to immunotherapy.
Collapse
Affiliation(s)
- Avishek Bhuniya
- Department of Pharmaceutical Sciences, Center for Pharmacogenetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Dhamotharan Pattarayan
- Department of Pharmaceutical Sciences, Center for Pharmacogenetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Da Yang
- Department of Pharmaceutical Sciences, Center for Pharmacogenetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,UPMC Hillman Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
17
|
Watson-Levings RS, Palmer GD, Levings PP, Dacanay EA, Evans CH, Ghivizzani SC. Gene Therapy in Orthopaedics: Progress and Challenges in Pre-Clinical Development and Translation. Front Bioeng Biotechnol 2022; 10:901317. [PMID: 35837555 PMCID: PMC9274665 DOI: 10.3389/fbioe.2022.901317] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/27/2022] [Indexed: 11/25/2022] Open
Abstract
In orthopaedics, gene-based treatment approaches are being investigated for an array of common -yet medically challenging- pathologic conditions of the skeletal connective tissues and structures (bone, cartilage, ligament, tendon, joints, intervertebral discs etc.). As the skeletal system protects the vital organs and provides weight-bearing structural support, the various tissues are principally composed of dense extracellular matrix (ECM), often with minimal cellularity and vasculature. Due to their functional roles, composition, and distribution throughout the body the skeletal tissues are prone to traumatic injury, and/or structural failure from chronic inflammation and matrix degradation. Due to a mixture of environment and endogenous factors repair processes are often slow and fail to restore the native quality of the ECM and its function. In other cases, large-scale lesions from severe trauma or tumor surgery, exceed the body’s healing and regenerative capacity. Although a wide range of exogenous gene products (proteins and RNAs) have the potential to enhance tissue repair/regeneration and inhibit degenerative disease their clinical use is hindered by the absence of practical methods for safe, effective delivery. Cumulatively, a large body of evidence demonstrates the capacity to transfer coding sequences for biologic agents to cells in the skeletal tissues to achieve prolonged delivery at functional levels to augment local repair or inhibit pathologic processes. With an eye toward clinical translation, we discuss the research progress in the primary injury and disease targets in orthopaedic gene therapy. Technical considerations important to the exploration and pre-clinical development are presented, with an emphasis on vector technologies and delivery strategies whose capacity to generate and sustain functional transgene expression in vivo is well-established.
Collapse
Affiliation(s)
- Rachael S. Watson-Levings
- Department of Orthopaedic Surgery and Sports Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - Glyn D. Palmer
- Department of Orthopaedic Surgery and Sports Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - Padraic P. Levings
- Department of Orthopaedic Surgery and Sports Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - E. Anthony Dacanay
- Department of Orthopaedic Surgery and Sports Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - Christopher H. Evans
- Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, MI, United States
| | - Steven C. Ghivizzani
- Department of Orthopaedic Surgery and Sports Medicine, University of Florida College of Medicine, Gainesville, FL, United States
- *Correspondence: Steven C. Ghivizzani,
| |
Collapse
|
18
|
Avances en terapia génica en humanos: algunos conceptos básicos y un recorrido histórico. REVISTA MÉDICA CLÍNICA LAS CONDES 2022. [DOI: 10.1016/j.rmclc.2022.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
19
|
Abstract
INTRODUCTION Lentiviral vectors have emerged as powerful vectors for vaccination, due to their high efficiency to transduce dendritic cells and to induce long-lasting humoral immunity, CD8+ T cells, and effective protection in numerous preclinical animal models of infection and oncology. AREAS COVERED Here, we reviewed the literature, highlighting the relevance of lentiviral vectors in vaccinology. We recapitulated both their virological and immunological aspects of lentiviral vectors. We compared lentiviral vectors to the gold standard viral vaccine vectors, i.e. adenoviral vectors, and updated the latest results in lentiviral vector-based vaccination in preclinical models. EXPERT OPINION Lentiviral vectors are non-replicative, negligibly inflammatory, and not targets of preexisting immunity in human populations. These are major characteristics to consider in vaccine development. The potential of lentiviral vectors to transduce non-dividing cells, including dendritic cells, is determinant in their strong immunogenicity. Notably, lentiviral vectors can be engineered to target antigen expression to specific host cells. The very weak inflammatory properties of these vectors allow their use in mucosal vaccination, with particular interest in infectious diseases that affect the lungs or brain, including COVID-19. Recent results in various preclinical models have reinforced the interest of these vectors in prophylaxis against infectious diseases and in onco-immunotherapy.
Collapse
Affiliation(s)
- Min-Wen Ku
- Virology Department, Institut Pasteur-TheraVectys Joint Lab, Paris, France
| | - Pierre Charneau
- Virology Department, Institut Pasteur-TheraVectys Joint Lab, Paris, France
| | - Laleh Majlessi
- Virology Department, Institut Pasteur-TheraVectys Joint Lab, Paris, France
| |
Collapse
|
20
|
Poth KM, Texakalidis P, Boulis NM. Chemogenetics: Beyond Lesions and Electrodes. Neurosurgery 2021; 89:185-195. [PMID: 33913505 PMCID: PMC8279839 DOI: 10.1093/neuros/nyab147] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 02/26/2021] [Indexed: 01/14/2023] Open
Abstract
The field of chemogenetics has rapidly expanded over the last decade, and engineered receptors are currently utilized in the lab to better understand molecular interactions in the nervous system. We propose that chemogenetic receptors can be used for far more than investigational purposes. The potential benefit of adding chemogenetic neuromodulation to the current neurosurgical toolkit is substantial. There are several conditions currently treated surgically, electrically, and pharmacologically in clinic, and this review highlights how chemogenetic neuromodulation could improve patient outcomes over current neurosurgical techniques. We aim to emphasize the need to take these techniques from bench to bedside.
Collapse
Affiliation(s)
- Kelly M Poth
- Department of Neurosurgery, Emory University, Atlanta, Georgia, USA
| | | | | |
Collapse
|
21
|
Lyu P, Wang L, Lu B. Virus-Like Particle Mediated CRISPR/Cas9 Delivery for Efficient and Safe Genome Editing. Life (Basel) 2020; 10:366. [PMID: 33371215 PMCID: PMC7766694 DOI: 10.3390/life10120366] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/11/2020] [Accepted: 12/15/2020] [Indexed: 12/26/2022] Open
Abstract
The discovery of designer nucleases has made genome editing much more efficient than before. The designer nucleases have been widely used for mechanistic studies, animal model generation and gene therapy development. However, potential off-targets and host immune responses are issues still need to be addressed for in vivo uses, especially clinical applications. Short term expression of the designer nucleases is necessary to reduce both risks. Currently, various delivery methods are being developed for transient expression of designer nucleases including Zinc Finger Nuclease (ZNF), Transcription Activator-Like Effector Nuclease (TALEN) and Clustered Regularly Interspaced Short Palindromic Repeats/CRISPR-associated (CRISPR/Cas). Recently, virus-like particles are being used for gene editing. In this review, we will talk through commonly used genome editing nucleases, discuss gene editing delivery tools and review the latest literature using virus-like particles to deliver gene editing effectors.
Collapse
Affiliation(s)
- Pin Lyu
- School of Physical Education and Health, Hangzhou Normal University, Hangzhou 311121, China;
| | - Luxi Wang
- Department of Cancer Biology, Comprehensive Cancer Center of Wake Forest Baptist Medical Center, Winston-Salem, NC 27157, USA;
| | - Baisong Lu
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA
| |
Collapse
|
22
|
Page A, Fusil F, Cosset FL. Toward Tightly Tuned Gene Expression Following Lentiviral Vector Transduction. Viruses 2020; 12:v12121427. [PMID: 33322556 PMCID: PMC7764518 DOI: 10.3390/v12121427] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/05/2020] [Accepted: 12/08/2020] [Indexed: 12/12/2022] Open
Abstract
Lentiviral vectors are versatile tools for gene delivery purposes. While in the earlier versions of retroviral vectors, transgene expression was controlled by the long terminal repeats (LTRs), the latter generations of vectors, including those derived from lentiviruses, incorporate internal constitutive or regulated promoters in order to regulate transgene expression. This allows to temporally and/or quantitatively control transgene expression, which is required for many applications such as for clinical applications, when transgene expression is required in specific tissues and at a specific timing. Here we review the main systems that have been developed for transgene regulated expression following lentiviral gene transfer. First, the induction of gene expression can be triggered either by external or by internal cues. Indeed, these regulated vector systems may harbor promoters inducible by exogenous stimuli, such as small molecules (e.g., antibiotics) or temperature variations, offering the possibility to tune rapidly transgene expression in case of adverse events. Second, expression can be indirectly adjusted by playing on inserted sequence copies, for instance by gene excision. Finally, synthetic networks can be developed to sense specific endogenous signals and trigger defined responses after information processing. Regulatable lentiviral vectors (LV)-mediated transgene expression systems have been widely used in basic research to uncover gene functions or to temporally reprogram cells. Clinical applications are also under development to induce therapeutic molecule secretion or to implement safety switches. Such regulatable approaches are currently focusing much attention and will benefit from the development of other technologies in order to launch autonomously controlled systems.
Collapse
|
23
|
Mika T, Ladigan-Badura S, Maghnouj A, Mustafa B, Klein-Scory S, Baraniskin A, Döhring S, Fuchs I, Ehl S, Hahn SA, Schroers R. Altered T-Lymphocyte Biology Following High-Dose Melphalan and Autologous Stem Cell Transplantation With Implications for Adoptive T-Cell Therapy. Front Oncol 2020; 10:568056. [PMID: 33363008 PMCID: PMC7759611 DOI: 10.3389/fonc.2020.568056] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 10/20/2020] [Indexed: 01/18/2023] Open
Abstract
In relapsed and refractory multiple myeloma (MM), adoptive cell therapies (ACT) including CAR-T-cells are under clinical investigation. However, relapse due to T-cell exhaustion or limited persistence is an obstacle. Before ACT are considered in MM, high-dose (HD) melphalan followed by autologous stem-cell transplantation (autoSCT) has been administered in most clinical situations. Yet, the impact of HD chemotherapy on T-cells in MM with respect to ACT is unclear. In this study, T-lymphocytes’ phenotypes, expansion properties, lentiviral transduction efficacy, and gene expression were examined with special respect to patients following HD melphalan. Significant impairment of T-cells’ expansion and transduction rates could be demonstrated. Expansion was diminished due to inherent disadvantages of the predominant T-cell phenotype but restored over time. The quantitative fraction of CD27−/CD28− T-cells before expansion was predictive of T-cell yield. Following autoSCT, the transduction efficacy was reduced by disturbed lentiviral genome integration. Moreover, an unfavorable T-cell phenotype after expansion was demonstrated. In initial analyses of CD107a degranulation impaired T-cell cytotoxicity was detected in one patient following melphalan and autoSCT. The findings of our study have potential implications regarding the time point of leukapheresis for CAR-T-cell manufacturing. Our results point to a preferred interval of more than 3 months until patients should undergo cell separation for CAR-T therapy in the specific situation post-HD melphalan/autoSCT. Monitoring of CD27−/CD28− T-cells, has the potential to influence clinical decision making before apheresis in MM.
Collapse
Affiliation(s)
- Thomas Mika
- Department of Medicine, Hematology and Oncology, Ruhr-University Bochum, Bochum, Germany.,Department of Molecular Gastrointestinal Oncology, Ruhr-University Bochum, Bochum, Germany
| | - Swetlana Ladigan-Badura
- Department of Medicine, Hematology and Oncology, Ruhr-University Bochum, Bochum, Germany.,Department of Molecular Gastrointestinal Oncology, Ruhr-University Bochum, Bochum, Germany
| | - Abdelouahid Maghnouj
- Department of Molecular Gastrointestinal Oncology, Ruhr-University Bochum, Bochum, Germany
| | - Bakr Mustafa
- Department of Molecular Gastrointestinal Oncology, Ruhr-University Bochum, Bochum, Germany
| | - Susanne Klein-Scory
- Department of Medicine, Hematology and Oncology, Ruhr-University Bochum, Bochum, Germany
| | - Alexander Baraniskin
- Department of Medicine, Hematology and Oncology, Ruhr-University Bochum, Bochum, Germany
| | - Sascha Döhring
- Department of Molecular Gastrointestinal Oncology, Ruhr-University Bochum, Bochum, Germany
| | - Ilka Fuchs
- Center for Chronic Immunodeficiency, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Stephan Ehl
- Center for Chronic Immunodeficiency, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Stephan A Hahn
- Department of Molecular Gastrointestinal Oncology, Ruhr-University Bochum, Bochum, Germany
| | - Roland Schroers
- Department of Medicine, Hematology and Oncology, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
24
|
Yang Y, Lu R, Gao F, Zhang J, Liu F. Berberine induces lipolysis in porcine adipocytes by activating the AMP‑activated protein kinase pathway. Mol Med Rep 2020; 21:2603-2614. [DOI: 10.3892/mmr.2020.11070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 03/17/2020] [Indexed: 11/05/2022] Open
Affiliation(s)
- Yongqing Yang
- Department of Biological Science, College of Life Science, Shanxi Normal University, Linfen, Shanxi 041000, P.R. China
| | - Rongsheng Lu
- Department of Biological Science, College of Life Science, Shanxi Normal University, Linfen, Shanxi 041000, P.R. China
| | - Fangfang Gao
- Department of Biological Science, College of Life Science, Shanxi Normal University, Linfen, Shanxi 041000, P.R. China
| | - Jie Zhang
- Department of Biological Science, College of Life Science, Shanxi Normal University, Linfen, Shanxi 041000, P.R. China
| | - Fenglan Liu
- Department of Biological Science, College of Life Science, Shanxi Normal University, Linfen, Shanxi 041000, P.R. China
| |
Collapse
|
25
|
Peng Y, Yang T, Tang X, Chen F, Wang S. Construction of an Inducible CRISPR/Cas9 System for CXCR4 Gene and Demonstration of its Effects on MKN-45 Cells. Cell Biochem Biophys 2020; 78:23-30. [PMID: 31875277 DOI: 10.1007/s12013-019-00898-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 12/14/2019] [Indexed: 12/13/2022]
Abstract
The CRISPR/Cas9 system is an effective tool for gene editing. However, this conventional expression system cannot control the timing of gene editing and does not utilize resistance screening markers. Therefore, carrying out CRISPR/Cas9 experiments is extremely inconvenient. Our aim is to develop an inducible lentiviral vector-based gene-editing system for C-X-C chemokine receptor 4 (CXCR4) by CRISPR/Cas9, and to demonstrate its function in MKN-45 cell. The DNA fragments of Blasticidin and T2A-GFP were produced using the lenti-Cas9-BLAST and PX458 plasmids as templates. The PCR products were harvested and cloned into the lenti-guide-puro plasmid to yield the lenti-guide-BLAST-GFP plasmid. Three double-stranded guide RNA (gRNA) sequences targeting the exon 2 of CXCR4 gene were designed online (http://crispr.mit.edu), synthesized, and recombined into the lenti-guide-BLAST-GFP plasmid, to yield the lenti-guide-BLAST-GFP-gRNA plasmid. The pCW-Cas9 and lenti-guide-BLAST-GFP-gRNA plasmids were packaged with lentiviral vectors, which were then transfected into MKN-45 cells, to identify the CXCR4 gene-editing effects using the T7 endonuclease 1 (T7E1) and Western blot assays. The lenti-guide-BLAST-GFP and lenti-guide-BLAST-GFP-gRNA plasmids were successfully constructed and packaged, to yield lentiviral particles. Transfection of the pCW-Cas9 and lenti-guide-BLAST-GFP-gRNA viral vectors could decrease the expression of CXCR4 protein, and lead to gene editing in MKN-45 cells. The efficiencies of gRNA-1, gRNA-2, and gRNA-3 were 45.6%, 53.6%, and 56.7%, respectively. Furthermore, the chemotactic efficiency of the dual viral vector-infected MKN-45 cells was significantly decreased in response to SDF-1. The numbers of migratory cells in the lower chamber of the transwell system were 30.0 ± 0.23, 29.7 ± 1.55, 28.2 ± 1.11 and 36.1 ± 2.00 cells per field (400×) for gRNA-1, gRNA-2, gRNA-3 and the control, respectively (P < 0.05). We constructed an inducible CXCR4 gene-editing, dual-vector CRISPR/Cas9 system, which could induce CXCR4 gene editing in MKN-45 cells in a doxycycline-dependent manner and thus reduce the migration of MKN-45 cells.
Collapse
Affiliation(s)
- Yanhua Peng
- Department of Anesthesiology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, No. 136, 2nd Zhongshan Rd, Yuzhong District, Chongqing, China
- Department of Anesthesiology, People's Hospital of Deyang City, Taishan North Road 173, Deyang, 618000, China
| | - Taobo Yang
- Department of Anesthesiology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, No. 136, 2nd Zhongshan Rd, Yuzhong District, Chongqing, China
| | - Xixi Tang
- Department of Anesthesiology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, No. 136, 2nd Zhongshan Rd, Yuzhong District, Chongqing, China
| | - Fei Chen
- Department of Anesthesiology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, No. 136, 2nd Zhongshan Rd, Yuzhong District, Chongqing, China
| | - Shouyong Wang
- Department of Anesthesiology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, No. 136, 2nd Zhongshan Rd, Yuzhong District, Chongqing, China.
| |
Collapse
|
26
|
Joshi J, Rubart M, Zhu W. Optogenetics: Background, Methodological Advances and Potential Applications for Cardiovascular Research and Medicine. Front Bioeng Biotechnol 2020; 7:466. [PMID: 32064254 PMCID: PMC7000355 DOI: 10.3389/fbioe.2019.00466] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 12/19/2019] [Indexed: 12/27/2022] Open
Abstract
Optogenetics is an elegant approach of precisely controlling and monitoring the biological functions of a cell, group of cells, tissues, or organs with high temporal and spatial resolution by using optical system and genetic engineering technologies. The field evolved with the need to precisely control neurons and decipher neural circuity and has made great accomplishments in neuroscience. It also evolved in cardiovascular research almost a decade ago and has made considerable progress in both in vitro and in vivo animal studies. Thus, this review is written with an objective to provide information on the evolution, background, methodical advances, and potential scope of the field for cardiovascular research and medicine. We begin with a review of literatures on optogenetic proteins related to their origin, structure, types, mechanism of action, methods to improve their performance, and the delivery vehicles and methods to express such proteins on target cells and tissues for cardiovascular research. Next, we reviewed historical and recent literatures to demonstrate the scope of optogenetics for cardiovascular research and regenerative medicine and examined that cardiac optogenetics is vital in mimicking heart diseases, understanding the mechanisms of disease progression and also in introducing novel therapies to treat cardiac abnormalities, such as arrhythmias. We also reviewed optogenetics as promising tools in providing high-throughput data for cardiotoxicity screening in drug development and also in deciphering dynamic roles of signaling moieties in cell signaling. Finally, we put forth considerations on the need of scaling up of the optogenetic system, clinically relevant in vivo and in silico models, light attenuation issues, and concerns over the level, immune reactions, toxicity, and ectopic expression with opsin expression. Detailed investigations on such considerations would accelerate the translation of cardiac optogenetics from present in vitro and in vivo animal studies to clinical therapies.
Collapse
Affiliation(s)
- Jyotsna Joshi
- Department of Cardiovascular Medicine, Physiology and Biomedical Engineering, Mayo Clinic, Phoenix, AZ, United States
| | - Michael Rubart
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Wuqiang Zhu
- Department of Cardiovascular Medicine, Physiology and Biomedical Engineering, Mayo Clinic, Phoenix, AZ, United States
| |
Collapse
|
27
|
Yang Y, Liu F, Lu R, Jia J. Berberine Inhibits Adipogenesis in Porcine Adipocytes
via
AMP‐Activated Protein Kinase‐Dependent and ‐Independent Mechanisms. Lipids 2019; 54:667-678. [DOI: 10.1002/lipd.12200] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 09/20/2019] [Accepted: 10/08/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Yongqing Yang
- College of Life ScienceShanxi Normal University The First Gongyuan Road, Linfen Shanxi Province 041000 People's Republic of China
| | - Fenglan Liu
- College of Life ScienceShanxi Normal University The First Gongyuan Road, Linfen Shanxi Province 041000 People's Republic of China
| | - Rongsheng Lu
- College of Life ScienceShanxi Normal University The First Gongyuan Road, Linfen Shanxi Province 041000 People's Republic of China
| | - Junli Jia
- College of Life ScienceShanxi Normal University The First Gongyuan Road, Linfen Shanxi Province 041000 People's Republic of China
| |
Collapse
|
28
|
Moore N, Chevillet JR, Healey LJ, McBrine C, Doty D, Santos J, Teece B, Truslow J, Mott V, Hsi P, Tandon V, Borenstein JT, Balestrini J, Kotz K. A Microfluidic Device to Enhance Viral Transduction Efficiency During Manufacture of Engineered Cellular Therapies. Sci Rep 2019; 9:15101. [PMID: 31641163 PMCID: PMC6806008 DOI: 10.1038/s41598-019-50981-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 09/23/2019] [Indexed: 02/08/2023] Open
Abstract
The development and approval of engineered cellular therapies are revolutionizing approaches to treatment of diseases. However, these life-saving therapies require extensive use of inefficient bioprocessing equipment and specialized reagents that can drive up the price of treatment. Integration of new genetic material into the target cells, such as viral transduction, is one of the most costly and labor-intensive steps in the production of cellular therapies. Approaches to reducing the costs associated with gene delivery have been developed using microfluidic devices to increase overall efficiency. However, these microfluidic approaches either require large quantities of virus or pre-concentration of cells with high-titer viral particles. Here, we describe the development of a microfluidic transduction device (MTD) that combines microfluidic spatial confinement with advective flow through a membrane to efficiently colocalize target cells and virus particles. We demonstrate that the MTD can improve the efficiency of lentiviral transduction for both T-cell and hematopoietic stem-cell (HSC) targets by greater than two fold relative to static controls. Furthermore, transduction saturation in the MTD is reached with only half the virus required to reach saturation under static conditions. Moreover, we show that MTD transduction does not adversely affect cell viability or expansion potential.
Collapse
Affiliation(s)
- Nathan Moore
- Cell and Tissue Engineering, 555 Technology Square, Draper, Cambridge, MA, 02139, USA.
| | - John R Chevillet
- Cell and Tissue Engineering, 555 Technology Square, Draper, Cambridge, MA, 02139, USA
| | - Laura J Healey
- Cell and Tissue Engineering, 555 Technology Square, Draper, Cambridge, MA, 02139, USA
| | - Connor McBrine
- Synthetic Biology, 555 Technology Square, Draper, Cambridge, MA, 02139, USA
| | - Daniel Doty
- Cell and Tissue Engineering, 555 Technology Square, Draper, Cambridge, MA, 02139, USA
| | - Jose Santos
- Biological Microsystems, 555 Technology Square, Draper, Cambridge, MA, 02139, USA
| | - Bryan Teece
- Biological Microsystems, 555 Technology Square, Draper, Cambridge, MA, 02139, USA
| | - James Truslow
- Biological Microsystems, 555 Technology Square, Draper, Cambridge, MA, 02139, USA
| | - Vienna Mott
- Biological Microsystems, 555 Technology Square, Draper, Cambridge, MA, 02139, USA
| | - Peter Hsi
- Cell and Tissue Engineering, 555 Technology Square, Draper, Cambridge, MA, 02139, USA
| | - Vishal Tandon
- Biological Microsystems, 555 Technology Square, Draper, Cambridge, MA, 02139, USA
| | | | - Jenna Balestrini
- Biological Microsystems, 555 Technology Square, Draper, Cambridge, MA, 02139, USA
| | - Kenneth Kotz
- Biological Microsystems, 555 Technology Square, Draper, Cambridge, MA, 02139, USA
| |
Collapse
|
29
|
Ipseiz N, Czubala MA, Bart VMT, Davies LC, Jenkins RH, Brennan P, Taylor PR. Effective In Vivo Gene Modification in Mouse Tissue-Resident Peritoneal Macrophages by Intraperitoneal Delivery of Lentiviral Vectors. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 16:21-31. [PMID: 31720306 PMCID: PMC6838965 DOI: 10.1016/j.omtm.2019.10.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 10/14/2019] [Indexed: 12/22/2022]
Abstract
Tissue-resident macrophages exhibit specialized phenotypes dependent on their in vivo physiological niche. Investigation of their function often relies upon complex whole mouse transgenic studies. While some appropriate lineage-associated promoters exist, there are no options for tissue-specific targeting of macrophages. We have developed full protocols for in vivo productive infection (defined by stable transgene expression) of tissue-resident macrophages with lentiviral vectors, enabling RNA and protein overexpression, including expression of small RNA species such as shRNA, to knock down and modulate gene expression. These approaches allow robust infection of peritoneal tissue-resident macrophages without significant infection of other cell populations. They permit rapid functional study of macrophages in homeostatic and inflammatory settings, such as thioglycolate-induced peritonitis, while maintaining the cells in their physiological context. Here we provide detailed protocols for the whole workflow: viral production, purification, and quality control; safety considerations for administration of the virus to mice; and assessment of in vivo transduction efficiency and the low background levels of inflammation induced by the virus. In summary, we present a quick and accessible protocol for the rapid assessment of gene function in peritoneal tissue-resident macrophages in vivo.
Collapse
Affiliation(s)
- Natacha Ipseiz
- Systems Immunity Research Institute, Cardiff University School of Medicine, Tenovus Building, Heath Park, Cardiff CF14 4XN, UK
| | - Magdalena A Czubala
- Systems Immunity Research Institute, Cardiff University School of Medicine, Tenovus Building, Heath Park, Cardiff CF14 4XN, UK
| | - Valentina M T Bart
- Systems Immunity Research Institute, Cardiff University School of Medicine, Tenovus Building, Heath Park, Cardiff CF14 4XN, UK
| | - Luke C Davies
- Systems Immunity Research Institute, Cardiff University School of Medicine, Tenovus Building, Heath Park, Cardiff CF14 4XN, UK
| | - Robert H Jenkins
- Systems Immunity Research Institute, Cardiff University School of Medicine, Tenovus Building, Heath Park, Cardiff CF14 4XN, UK
| | - Paul Brennan
- Division of Cancer and Genetics, Cardiff University, School of Medicine, Cancer and Genetics Building, Heath Park, Cardiff CF14 4XN, UK
| | - Philip R Taylor
- Systems Immunity Research Institute, Cardiff University School of Medicine, Tenovus Building, Heath Park, Cardiff CF14 4XN, UK.,UK Dementia Research Institute at Cardiff, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK
| |
Collapse
|
30
|
Timmins LM, Patel RS, Teryek MS, Parekkadan B. Real-time transfer of lentiviral particles by producer cells using an engineered coculture system. Cytotechnology 2019; 71:1019-1031. [PMID: 31515650 PMCID: PMC6787137 DOI: 10.1007/s10616-019-00343-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 09/06/2019] [Indexed: 11/25/2022] Open
Abstract
Lentiviruses are quite effective gene delivery systems for stable production of genetically engineered human cells. However, prior to using lentivirus to deliver genetic materials to cells of interest, the normal course of production of these lentiviruses involves a lengthy collection, purification, preservation, and quantification process. In this report, we demonstrate the ability for producer HEK293T cells to simultaneously produce lentiviral particles and transduce (i.e., infect) target cells through a membrane-based coculture system in a continuous, real-time mode which negates the need for a separate viral collection and quantification process. The coculture system was evaluated for major design features such as variations in HEK293T seeding density, target cell type densities, as well as membrane porosities to identify key relationships between lentiviral particle production rate and infection kinetics for adherent and suspension cell types. As a proof-of-concept for the creation of an engineered cell immunotherapy, we describe the ability to engineer human T cells isolated from PBMCs under the control of this coculture system in under 6 days with a GFP construct. These studies suggest the capability to combine and more closely automate the transfection/transduction process in order to facilitate well-timed and cost-effective transduction of target cell types. These experiments provide novel insight into the forthcoming transition into improved manufacturing systems for viral production and subsequent cell engineering.
Collapse
Affiliation(s)
- Lauren M Timmins
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08902, USA
| | - Riya S Patel
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08902, USA
| | - Matthew S Teryek
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08902, USA
| | - Biju Parekkadan
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08902, USA.
| |
Collapse
|
31
|
Abstract
Viral vectors are a promising tool for effective delivery of genetic material into cells. They take advantage of the natural ability of a virus to deliver a genetic payload into cells while being genetically modified such that their ability to replicate is crippled or removed. Here, an updated overview of routinely used viral vectors, including adeno-associated viruses (AAV), retroviruses/lentiviruses, and adenoviruses (Ads), is provided, as well as perspectives on their advantages and disadvantages in research and gene therapy. © 2018 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Yong Hong Chen
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Megan S Keiser
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Beverly L Davidson
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania.,University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
32
|
Ye L, Lou Y, Lu L, Fan X. Mesothelin-targeted second generation CAR-T cells inhibit growth of mesothelin-expressing tumors in vivo. Exp Ther Med 2018; 17:739-747. [PMID: 30651858 PMCID: PMC6307389 DOI: 10.3892/etm.2018.7015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 08/31/2018] [Indexed: 12/22/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) and mesothelioma are renowned for being diagnosed at a late stage and poor prognosis. Although surgery, chemotherapy, and radiotherapy have yielded successful outcomes, the improvement on the survival rate of NSCLC and mesothelioma have been less marked. Recently, adoptive immunotherapy, particularly chimeric antigen receptor T (CAR-T) cell therapy demonstrated promise for improving the survival of acute lymphoblastic leukemia with minimum toxicity. However, its application in solid tumors still warrants in-depth investigations and multiple consistent trial results, particularly in eliminating 'off-tumor' toxicity. To explore CAR-T therapy in NSCLC and mesothelioma, second-generation CAR-T cells were constructed targeting mesothelin (MSLN), which is abundant in NSCLC and mesothelioma but is under expressed in normal tissues. The second-generation design incorporated co-stimulatory CD28 and 4-1BB signaling domains to enhance the proliferation. Following the successful analysis of CAR-T cells by flow cytometry, cytotoxicity experiments were performed using the LDH kit to verify the killing effect of CAR-T cells on target cells. Otherwise, the in vivo killing tumor activity of MSLN CAR-T cells was verified by constructing a mouse model using tumor-derived cells from patients to inoculate the mice. When the effector-to-target ratio is >0.5:1, CAR-T MSLN cells exhibited significantly higher ability to kill tumor cells than T cells. In in vivo experiments, mice whose tail vein was injected with CAR-T MSLN cells demonstrated significantly slower tumor growth. Without continuous administration, both groups became gradually synchronized in growth of tumor size, which suggests that the persistence of CAR-T cells is an important issue in preclinical studies.
Collapse
Affiliation(s)
- Lin Ye
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Yuqing Lou
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Liming Lu
- Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Xiaohong Fan
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| |
Collapse
|
33
|
Vio V, Riveros AL, Tapia-Bustos A, Lespay-Rebolledo C, Perez-Lobos R, Muñoz L, Pismante P, Morales P, Araya E, Hassan N, Herrera-Marschitz M, Kogan MJ. Gold nanorods/siRNA complex administration for knockdown of PARP-1: a potential treatment for perinatal asphyxia. Int J Nanomedicine 2018; 13:6839-6854. [PMID: 30498346 PMCID: PMC6207385 DOI: 10.2147/ijn.s175076] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background Perinatal asphyxia interferes with neonatal development, resulting in long-term deficits associated with systemic and neurological diseases. Despite the important role of poly (ADP-ribose) polymerase 1 (PARP-1) in the regulation of gene expression and DNA repair, overactivation of PARP-1 in asphyxia-exposed animals worsens the ATP-dependent energetic crisis. Inhibition of PARP-1 offers a therapeutic strategy for diminishing the effects of perinatal asphyxia. Methods We designed a nanosystem that incorporates a specific siRNA for PARP-1 knockdown. The siRNA was complexed with gold nanorods (AuNR) conjugated to the peptide CLPFFD for brain targeting. Results The siRNA was efficiently delivered into PC12 cells, resulting in gene silencing. The complex was administered intraperitoneally in vivo to asphyxia-exposed rat pups, and the ability of the AuNR-CLPFFD/siRNA complex to reach the brain was demonstrated. Conclusion The combination of a nanosystem for delivery and a specific siRNA for gene silencing resulted in effective inhibition of PARP-1 in vivo.
Collapse
Affiliation(s)
- Valentina Vio
- Department of Pharmacological and Toxicology Chemistry, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile, .,Program of Molecular and Clinical Pharmacology, Medical Faculty, Universidad de Chile, Santiago, Chile,
| | - Ana L Riveros
- Department of Pharmacological and Toxicology Chemistry, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile,
| | - Andrea Tapia-Bustos
- Program of Molecular and Clinical Pharmacology, Medical Faculty, Universidad de Chile, Santiago, Chile,
| | - Carolyne Lespay-Rebolledo
- Program of Molecular and Clinical Pharmacology, Medical Faculty, Universidad de Chile, Santiago, Chile,
| | - Ronald Perez-Lobos
- Program of Molecular and Clinical Pharmacology, Medical Faculty, Universidad de Chile, Santiago, Chile,
| | - Luis Muñoz
- Chemical Meteorology Section, Comisión Chilena de Energía Nuclear, Santiago, Chile
| | - Paola Pismante
- Chemical Meteorology Section, Comisión Chilena de Energía Nuclear, Santiago, Chile
| | - Paola Morales
- Program of Molecular and Clinical Pharmacology, Medical Faculty, Universidad de Chile, Santiago, Chile, .,Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Eyleen Araya
- Departamento de Ciencias Quimicas, Facultad de Ciencias Exactas, Universidad Andres Bello, Santiago, Chile
| | - Natalia Hassan
- Department of Pharmacological and Toxicology Chemistry, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile, .,Programa Institucional de Fomento a la Investigación, Desarrollo e Innovación, Universidad Tecnológica Metropolitana, Santiago, Chile
| | - Mario Herrera-Marschitz
- Program of Molecular and Clinical Pharmacology, Medical Faculty, Universidad de Chile, Santiago, Chile,
| | - Marcelo J Kogan
- Department of Pharmacological and Toxicology Chemistry, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile, .,Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile,
| |
Collapse
|
34
|
Alfranca A, Campanero MR, Redondo JM. New Methods for Disease Modeling Using Lentiviral Vectors. Trends Mol Med 2018; 24:825-837. [PMID: 30213701 DOI: 10.1016/j.molmed.2018.08.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 08/02/2018] [Accepted: 08/03/2018] [Indexed: 12/11/2022]
Abstract
Lentiviral vectors (LVs) transduce quiescent cells and provide stable integration to maintain transgene expression. Several approaches have been adopted to optimize LV safety profiles. Similarly, LV targeting has been tailored through strategies including the modification of envelope components, the use of specific regulatory elements, and the selection of appropriate administration routes. Models of aortic disease based on a single injection of pleiotropic LVs have been developed that efficiently transduce the three aorta layers in wild type mice. This approach allows the dissection of pathways involved in aortic aneurysm formation and the identification of targets for gene therapy in aortic diseases. LVs provide a fast, efficient, and affordable alternative to genetically modified mice to study disease mechanisms and develop therapeutic tools.
Collapse
Affiliation(s)
- Arantzazu Alfranca
- Department of Immunology, Hospital Universitario de La Princesa, Madrid, Spain; CIBERCV, Madrid, Spain.
| | - Miguel R Campanero
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM, Madrid, Spain; CIBERCV, Madrid, Spain
| | - Juan Miguel Redondo
- Gene Regulation in Cardiovascular Remodeling and Inflammation Group, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain; CIBERCV, Madrid, Spain.
| |
Collapse
|
35
|
Tasyurek HM, Altunbas HA, Balci MK, Griffith TS, Sanlioglu S. Therapeutic Potential of Lentivirus-Mediated Glucagon-Like Peptide-1 Gene Therapy for Diabetes. Hum Gene Ther 2018; 29:802-815. [PMID: 29409356 DOI: 10.1089/hum.2017.180] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Postprandial glucose-induced insulin secretion from the islets of Langerhans is facilitated by glucagon-like peptide-1 (GLP-1)-a metabolic hormone with insulinotropic properties. Among the variety of effects it mediates, GLP-1 induces delta cell secretion of somatostatin, inhibits alpha cell release of glucagon, reduces gastric emptying, and slows food intake. These events collectively contribute to weight loss over time. During type 2 diabetes (T2DM), however, the incretin response to glucose is reduced and accompanied by a moderate reduction in GLP-1 secretion. To compensate for the reduced incretin effect, a human immunodeficiency virus-based lentiviral vector was generated to deliver DNA encoding human GLP-1 (LentiGLP-1), and the anti-diabetic efficacy of LentiGLP-1 was tested in a high-fat diet/streptozotocin-induced model of T2DM. Therapeutic administration of LentiGLP-1 reduced blood glucose levels in obese diabetic Sprague Dawley rats, along with improving insulin sensitivity and glucose tolerance. Normoglycemia was correlated with increased blood GLP-1 and pancreatic beta cell regeneration in LentiGLP-1-treated rats. Plasma triglyceride levels were also normalized after LentiGLP-1 injection. Collectively, these data suggest the clinical potential of GLP-1 gene transfer therapy for the treatment of T2DM.
Collapse
Affiliation(s)
- Hale M Tasyurek
- 1 Human Gene and Cell Therapy Center of Akdeniz University Hospitals , Antalya, Turkey
| | - Hasan Ali Altunbas
- 2 Department of Internal Medicine, Division of Endocrinology and Metabolism, Akdeniz University Faculty of Medicine, Antalya, Turkey
| | - Mustafa Kemal Balci
- 2 Department of Internal Medicine, Division of Endocrinology and Metabolism, Akdeniz University Faculty of Medicine, Antalya, Turkey
| | - Thomas S Griffith
- 3 Department of Urology, University of Minnesota , School of Medicine, Minneapolis, Minnesota
| | - Salih Sanlioglu
- 1 Human Gene and Cell Therapy Center of Akdeniz University Hospitals , Antalya, Turkey
| |
Collapse
|
36
|
Angelina C, Tan ISY, Choo Z, Lee OZJ, Pervaiz S, Chen ZX. KIF1Bβ increases ROS to mediate apoptosis and reinforces its protein expression through O 2- in a positive feedback mechanism in neuroblastoma. Sci Rep 2017; 7:16867. [PMID: 29203804 PMCID: PMC5715000 DOI: 10.1038/s41598-017-17192-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 11/22/2017] [Indexed: 01/15/2023] Open
Abstract
Relapse-prone, poor prognosis neuroblastoma is frequently characterized by deletion of chr1p36 where tumor suppressor gene KIF1Bβ resides. Interestingly, many 1p36-positive patients failed to express KIF1Bβ protein. Since altered cellular redox status has been reported to be involved in cell death and protein modification, we investigated the relationship between reactive oxygen species (ROS) and KIF1Bβ. Here, we showed that wild-type KIF1Bβ protein expression positively correlates with superoxide (O2-) and total ROS levels in neuroblastoma cells, unlike apoptotic loss-of-function KIF1Bβ mutants. Overexpression of KIF1Bβ apoptotic domain variants increases total ROS and, specifically O2-, whereas knockdown of endogenous KIF1Bβ decreases ROS and O2-. Interestingly, O2- increases KIF1Bβ protein expression, independent of the proteasomal degradation pathway. Scavenging O2- or ROS decreases KIF1Bβ protein expression and subsequent apoptosis. Moreover, treatment with investigational redox compound Gliotoxin increases O2-, KIF1Bβ protein expression, apoptosis and colony formation inhibition. Overall, our findings suggest that ROS and O2- may be important downstream effectors of KIF1Bβ-mediated apoptosis. Subsequently, O2- produced may increase KIF1Bβ protein expression in a positive feedback mechanism. Therefore, ROS and, specifically O2-, may be critical regulators of KIF1Bβ-mediated apoptosis and its protein expression in neuroblastoma.
Collapse
Affiliation(s)
- Clara Angelina
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Irene Sze Ying Tan
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Zhang'e Choo
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Oswald Zhao Jian Lee
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Shazib Pervaiz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, Singapore.,Singapore-MIT Alliance, Singapore, Singapore
| | - Zhi Xiong Chen
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore. .,KK Women's and Children's Hospital, Singapore, Singapore.
| |
Collapse
|
37
|
Tokgun O, Fiorentino FP, Tokgun PE, Yokota J, Akca H. Design of a Lentiviral Vector for the Inducible Expression of MYC: A New Strategy for Construction Approach. Mol Biotechnol 2017; 59:200-206. [DOI: 10.1007/s12033-017-0006-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
38
|
Taheri-Kafrani A, Shirzadfar H, Tavassoli-Kafrani E. Dendrimers and Dendrimers-Grafted Superparamagnetic Iron Oxide Nanoparticles: Synthesis, Characterization, Functionalization, and Biological Applications in Drug Delivery Systems. NANO- AND MICROSCALE DRUG DELIVERY SYSTEMS 2017:75-94. [DOI: 10.1016/b978-0-323-52727-9.00005-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
39
|
Genetic Manipulation with Viral Vectors to Assess Metabolism and Adipose Tissue Function. Methods Mol Biol 2017; 1566:109-124. [PMID: 28244045 DOI: 10.1007/978-1-4939-6820-6_11] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Viral vectors have become widely used tools for genetic manipulation of adipose tissues to understand the biology and function of adipocytes in metabolism. There are a number of different viral vectors commonly used: retrovirus, lentivirus, adenovirus, and adeno-associated virus (AAV). Here, we review examples from the literature and describe methods to transduce adipocytes and adipose tissues using retrovirus, lentivirus, adenovirus, and AAV to ascertain gene function in adipose biology.
Collapse
|
40
|
Abstract
The immune system evolved to distinguish non-self from self to protect the organism. As cancer is derived from our own cells, immune responses to dysregulated cell growth present a unique challenge. This is compounded by mechanisms of immune evasion and immunosuppression that develop in the tumour microenvironment. The modern genetic toolbox enables the adoptive transfer of engineered T cells to create enhanced anticancer immune functions where natural cancer-specific immune responses have failed. Genetically engineered T cells, so-called 'living drugs', represent a new paradigm in anticancer therapy. Recent clinical trials using T cells engineered to express chimeric antigen receptors (CARs) or engineered T cell receptors (TCRs) have produced stunning results in patients with relapsed or refractory haematological malignancies. In this Review we describe some of the most recent and promising advances in engineered T cell therapy with a particular emphasis on what the next generation of T cell therapy is likely to entail.
Collapse
MESH Headings
- Antigen Presentation
- Antigens, CD19/immunology
- Antigens, Neoplasm/immunology
- Clinical Trials as Topic
- Costimulatory and Inhibitory T-Cell Receptors/genetics
- Costimulatory and Inhibitory T-Cell Receptors/immunology
- Cytokines/metabolism
- Forecasting
- Gene Editing
- Gene Transfer Techniques
- Genetic Engineering
- HLA Antigens/immunology
- Hematologic Neoplasms/immunology
- Hematologic Neoplasms/therapy
- Humans
- Immunotherapy, Adoptive/adverse effects
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/trends
- Models, Immunological
- Neoplasms/immunology
- Neoplasms/therapy
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Syndrome
- T-Cell Antigen Receptor Specificity
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/transplantation
- Tumor Escape
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Andrew D Fesnak
- Department of Pathology and Laboratory Medicine and Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104-5156, USA
| | - Carl H June
- Department of Pathology and Laboratory Medicine and Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104-5156, USA
| | - Bruce L Levine
- Department of Pathology and Laboratory Medicine and Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104-5156, USA
| |
Collapse
|
41
|
Chen F, Fan C, Gu X, Zhang H, Liu Q, Gao X, Lu J, He B, Lai X. Construction of Anti-CD20 Single-Chain Antibody-CD28-CD137-TCRζ Recombinant Genetic Modified T Cells and its Treatment Effect on B Cell Lymphoma. Med Sci Monit 2015. [PMID: 26195067 PMCID: PMC4537073 DOI: 10.12659/msm.893791] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Background Immunotherapy has been explored as a new therapy for B cell lymphoma, which is a non-Hodgkin’s lymphoma. Because CD20 is a B lymphocyte-specific marker, anti-CD20 single chain-tagged T lymphocytes have already begun to be experimentally used in B cell lymphoma treatment, but its use is still limited because of its unspecific targeting. T cells transfected with CD28 and CD137 can significantly improve the ability of cytokines secretion and anti-tumor effect, as well as extending T cell survival time and improving their proliferation ability. Material/Methods Genes containing anti-CD20-CD28-CD137-TCRζ were constructed. After cloning and sequencing, the plasmid was constructed and packaged by lentivirus. It was transfected to the peripheral blood T lymphocyte after identification transfection to induce the fusion protein expression. The cells were incubated with Raji cells and the LDH test was performed to detect the cytotoxic effect of CAR-T cells; the tumor volume and survival rate were measured to observe its inhibitory effect on B cell lymphoma in nude mice. Results Gene with anti-CD20-CD28-CD137-TCRζ was successfully constructed and transfected to the T cell surface. LDH assay revealed that CAR-T cells can kill the Raji cells with a killing rate of 32.89±6.26%. It can significantly inhibit B cell lymphoma growth in nude mice. Conclusions T lymphocytes transfected with anti-CD20-CD28-CD137-TCRζ fusion gene can kill B cell lymphoma, which could provide a new strategy for tumor treatment.
Collapse
Affiliation(s)
- Fei Chen
- Department of Nephrology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China (mainland)
| | - Chuming Fan
- Intensive Care Unit, Hospital of Yunnan Province, Kunming, Yunnan, China (mainland)
| | - Xuezhong Gu
- Department of Hematology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China (mainland)
| | - Haixi Zhang
- Department of Hematology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China (mainland)
| | - Qian Liu
- Department of Hematology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China (mainland)
| | - Xiaoli Gao
- Department of Hematology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China (mainland)
| | - Jie Lu
- Department of Hematology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China (mainland)
| | - Baoli He
- Animal Laboratory, Kunming Medical University, Kunming, Yunnan, China (mainland)
| | - Xun Lai
- Department of Hematology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China (mainland)
| |
Collapse
|
42
|
Kajaste-Rudnitski A, Naldini L. Cellular innate immunity and restriction of viral infection: implications for lentiviral gene therapy in human hematopoietic cells. Hum Gene Ther 2015; 26:201-9. [PMID: 25808164 DOI: 10.1089/hum.2015.036] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Hematopoietic gene therapy has tremendous potential to treat human disease. Nevertheless, for gene therapy to be efficacious, effective gene transfer into target cells must be reached without inducing detrimental effects on their biological properties. This remains a great challenge for the field as high vector doses and prolonged ex vivo culture conditions are still required to reach significant transduction levels of clinically relevant human hematopoietic stem and progenitor cells (HSPCs), while other potential target cells such as primary macrophages can hardly be transduced. The reasons behind poor permissiveness of primary human hematopoietic cells to gene transfer partly reside in the retroviral origin of lentiviral vectors (LVs). In particular, host antiviral factors referred to as restriction factors targeting the retroviral life cycle can hamper LV transduction efficiency. Furthermore, LVs may activate innate immune sensors not only in differentiated hematopoietic cells but also in HSPCs, with potential consequences on transduction efficiency as well as their biological properties. Therefore, better understanding of the vector-host interactions in the context of hematopoietic gene transfer is important for the development of safer and more efficient gene therapy strategies. In this review, we briefly summarize the current knowledge regarding innate immune recognition of lentiviruses in primary human hematopoietic cells as well as discuss its relevance for LV-based ex vivo gene therapy approaches.
Collapse
Affiliation(s)
- Anna Kajaste-Rudnitski
- 1 Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute , Milan 20132, Italy
| | | |
Collapse
|
43
|
Levine BL. Performance-enhancing drugs: design and production of redirected chimeric antigen receptor (CAR) T cells. Cancer Gene Ther 2015; 22:79-84. [DOI: 10.1038/cgt.2015.5] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 12/21/2014] [Accepted: 12/23/2014] [Indexed: 02/02/2023]
|
44
|
Vaginal challenge with an SIV-based dual reporter system reveals that infection can occur throughout the upper and lower female reproductive tract. PLoS Pathog 2014; 10:e1004440. [PMID: 25299616 PMCID: PMC4192600 DOI: 10.1371/journal.ppat.1004440] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 08/29/2014] [Indexed: 12/21/2022] Open
Abstract
The majority of new HIV infections occur in women as a result of heterosexual intercourse, overcoming multiple innate barriers to infection within the mucosa. However, the avenues through which infection is established, and the nature of bottlenecks to transmission, have been the source of considerable investigation and contention. Using a high dose of a single round non-replicating SIV-based vector containing a novel dual reporter system, we determined the sites of infection by the inoculum using the rhesus macaque vaginal transmission model. Here we show that the entire female reproductive tract (FRT), including the vagina, ecto- and endocervix, along with ovaries and local draining lymph nodes can contain transduced cells only 48 hours after inoculation. The distribution of infection shows that virions quickly disseminate after exposure and can access target cells throughout the FRT, with an apparent preference for infection in squamous vaginal and ectocervical mucosa. JRFL enveloped virions infect diverse CD4 expressing cell types, with T cells resident throughout the FRT representing the primary target. These findings establish a new perspective that the entire FRT is susceptible and virus can reach as far as the ovary and local draining lymph nodes. Based on these findings, it is essential that protective mechanisms for prevention of HIV acquisition must be present at protective levels throughout the entire FRT to provide complete protection. There is currently a great effort world-wide to develop interventions such as vaccines and microbicides to decrease, or hopefully block, HIV transmission. To model the infection of women, the field utilizes the rhesus macaque vaginal transmission model. Understanding the initial events leading to infection after viral challenge of the female reproductive tract (FRT) is crucial for the development of functional prevention strategies. To this end, we developed a novel method for detecting infection in the rhesus macaque FRT after vaginal inoculation. This method utilizes single round replication defective vector that expresses dual reporter proteins, Luciferase and mCherry. Monitoring Luciferase expression allows us to identify the sites of infection within the intact FRT, while fluorescent protein mCherry allows us to visualize the single infected cells. Our studies revealed that virus can access the entire upper and lower reproductive tract. Infection occurs primarily in vaginal and ectocervical tissue, but can spread as far as the ovary and local draining lymph nodes. All classically defined susceptible cell types can be infected with the broadly tropic HIV envelope utilized in this study. Prevention strategies aimed at protecting from HIV infection should consider the entire FRT architecture as potentially susceptible and design interventions accordingly.
Collapse
|
45
|
Petersen GF, Hilbert B, Trope G, Kalle W, Strappe P. Efficient transduction of equine adipose-derived mesenchymal stem cells by VSV-G pseudotyped lentiviral vectors. Res Vet Sci 2014; 97:616-22. [PMID: 25443656 DOI: 10.1016/j.rvsc.2014.09.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 09/03/2014] [Accepted: 09/08/2014] [Indexed: 11/30/2022]
Abstract
Equine adipose-derived mesenchymal stem cells (EADMSC) provide a unique cell-based approach for treatment of a variety of equine musculoskeletal injuries, via regeneration of diseased or damaged tissue, or the secretion of immunomodulatory molecules. These capabilities can be further enhanced by genetic modification using lentiviral vectors, which provide a safe and efficient method of gene delivery. We investigated the suitability of lentiviral vector technology for gene delivery into EADMSC, using GFP expressing lentiviral vectors pseudotyped with the G glycoprotein from the vesicular stomatitis virus (V-GFP) or, for the first time, the baculovirus gp64 envelope protein (G-GFP). In this study, we produced similarly high titre V-GFP and G-GFP lentiviral vectors. Flow cytometric analysis showed efficient transduction using V-GFP; however G-GFP exhibited a poor ability to transduce EADMSC. Transduction resulted in sustained GFP expression over four passages, with minimal effects on cell viability and doubling time, and an unaltered chondrogenic differentiation potential.
Collapse
Affiliation(s)
- Gayle F Petersen
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, NSW, Australia
| | - Bryan Hilbert
- School of Animal and Veterinary Sciences, Charles Sturt University, Wagga Wagga, NSW, Australia
| | - Gareth Trope
- School of Animal and Veterinary Sciences, Charles Sturt University, Wagga Wagga, NSW, Australia
| | - Wouter Kalle
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, NSW, Australia
| | - Padraig Strappe
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, NSW, Australia.
| |
Collapse
|
46
|
Abstract
Glucagon-like peptide (GLP)-1 is an incretin hormone with several antidiabetic functions including stimulation of glucose-dependent insulin secretion, increase in insulin gene expression and beta-cell survival. Despite the initial technical difficulties and profound inefficiency of direct gene transfer into the pancreas that seriously restricted in vivo gene transfer experiments with GLP-1, recent exploitation of various routes of gene delivery and alternative means of gene transfer has permitted the detailed assessment of the therapeutic efficacy of GLP-1 in animal models of type 2 diabetes (T2DM). As a result, many clinical benefits of GLP-1 peptide/analogues observed in clinical trials involving induction of glucose tolerance, reduction of hyperglycaemia, suppression of appetite and food intake linked to weight loss have been replicated in animal models using gene therapy. Furthermore, GLP-1-centered gene therapy not only improved insulin sensitivity, but also reduced abdominal and/or hepatic fat associated with obesity-induced T2DM with drastic alterations in adipokine profiles in treated subjects. Thus, a comprehensive assessment of recent GLP-1-mediated gene therapy approaches with detailed analysis of current hurdles and resolutions, is discussed.
Collapse
|
47
|
Abstract
Skeletal muscle satellite cells (myoblasts) are the primary stem cells of skeletal muscle which contribute to growth, maintenance, and repair of the muscles. Satellite cells are the first stem cells used for cellular cardiomyoplasty more than 20 years ago. The isolation, culture, labeling, and identification of satellite cells are described in detail here. The implantation and outcomes of cellular cardiomyoplasty using satellite cells have been summarized in the previous chapter (Chapter 1).
Collapse
|
48
|
Wu J, Wang D, Ruan D, He Q, Zhang Y, Wang C, Xin H, Xu C, Liu Y. Prolonged expansion of human nucleus pulposus cells expressing human telomerase reverse transcriptase mediated by lentiviral vector. J Orthop Res 2014; 32:159-66. [PMID: 23983186 DOI: 10.1002/jor.22474] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2013] [Accepted: 07/31/2013] [Indexed: 02/04/2023]
Abstract
Human degenerative disc disease (DDD) is characterized by progressive loss of human nucleus pulposus (HNP) cells and extracellular matrix, in which the massive deposition are secreted by HNP cells. Cell therapy to supplement HNP cells to degenerated discs has been thought to be a promising strategy to treat DDD. However, obtaining a large quality of fully functional HNP cells has been severely hampered by limited proliferation capacity of HNP cells in vitro. Previous studies have used lipofectamine or recombinant adeno-associated viral (rAAV) vectors to deliver human telomerase reverse transcriptase (hTERT) into ovine or HNP cells to prolong the activity of nucleus pulposus cells with limited success. Here we developed a lentiviral vector bearing both hTERT and a gene encoding green fluorescence protein (L-hTERT/EGFP). This vector efficiently mediated both hTERT and EGFP into freshly isolated HNP cells. The expressions of both transgenes in L-hTERT/EGFP transduced HNP cells were detected up to day 210 post viral infection, which was twice as long as rAAV vector did. Furthermore, we observed restored telomerase activity, maintained telomere length, delayed cell senescence, and increased cell proliferation rate in those L-hTERT/EGFP transduced HNP cells. Our study suggests that lentiviral vector might be a useful gene delivery vehicle for HNP cell therapy to treat DDD.
Collapse
Affiliation(s)
- Jianhong Wu
- Department of Orthopaedic Surgery, Navy General Hospital, No.6 Fu-cheng Road, Beijing, 100048, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Conroy R, Seto B. Multifunctional Nanoscale Delivery Systems for Nucleic Acids. ENGINEERING IN TRANSLATIONAL MEDICINE 2014:475-512. [DOI: 10.1007/978-1-4471-4372-7_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
50
|
Sicard F, Gayral M, Lulka H, Buscail L, Cordelier P. Targeting miR-21 for the therapy of pancreatic cancer. Mol Ther 2013; 21:986-94. [PMID: 23481326 DOI: 10.1038/mt.2013.35] [Citation(s) in RCA: 177] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Despite tremendous efforts worldwide from clinicians and cancer scientists, pancreatic ductal adenocarcinoma (PDA) remains a deadly disease for which no cure is available. Recently, microRNAs (miRNAs) have emerged as key actors in carcinogenesis and we demonstrated that microRNA-21 (miR-21), oncomiR is expressed early during PDA. In the present study, we asked whether targeting miR-21 in human PDA-derived cell lines using lentiviral vectors (LVs) may impede tumor growth. We demonstrated that LVs-transduced human PDA efficiently downregulated miR-21 expression, both in vitro and in vivo. Consequently, cell proliferation was strongly inhibited and PDA-derived cell lines died by apoptosis through the mitochondrial pathway. In vivo, miR-21 depletion stopped the progression of a very aggressive model of PDA, to induce cell death by apoptosis; furthermore, combining miR-21 targeting and chemotherapeutic treatment provoked tumor regression. We demonstrate herein for the first time that targeting oncogenic miRNA strongly inhibit pancreatic cancer tumor growth both in vitro and in vivo. Because miR-21 is overexpressed in most human tumors; therapeutic delivery of miR-21 antagonists may still be beneficial for a large number of cancers for which no cure is available.
Collapse
Affiliation(s)
- Flavie Sicard
- INSERM U1037, Cancer Research Center of Toulouse, Toulouse, France
| | | | | | | | | |
Collapse
|