1
|
Peng J, Zou WW, Wang XL, Zhang ZG, Huo R, Yang L. Viral-mediated gene therapy in pediatric neurological disorders. World J Pediatr 2024; 20:533-555. [PMID: 36607547 DOI: 10.1007/s12519-022-00669-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 11/27/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND Due to the broad application of next-generation sequencing, the molecular diagnosis of genetic disorders in pediatric neurology is no longer an unachievable goal. However, treatments for neurological genetic disorders in children remain primarily symptomatic. On the other hand, with the continuous evolution of therapeutic viral vectors, gene therapy is becoming a clinical reality. From this perspective, we wrote this review to illustrate the current state regarding viral-mediated gene therapy in childhood neurological disorders. DATA SOURCES We searched databases, including PubMed and Google Scholar, using the keywords "adenovirus vector," "lentivirus vector," and "AAV" for gene therapy, and "immunoreaction induced by gene therapy vectors," "administration routes of gene therapy vectors," and "gene therapy" with "NCL," "SMA," "DMD," "congenital myopathy," "MPS" "leukodystrophy," or "pediatric metabolic disorders". We also screened the database of ClinicalTrials.gov using the keywords "gene therapy for children" and then filtered the results with the ones aimed at neurological disorders. The time range of the search procedure was from the inception of the databases to the present. RESULTS We presented the characteristics of commonly used viral vectors for gene therapy for pediatric neurological disorders and summarized their merits and drawbacks, the administration routes of each vector, the research progress, and the clinical application status of viral-mediated gene therapy on pediatric neurological disorders. CONCLUSIONS Viral-mediated gene therapy is on the brink of broad clinical application. Viral-mediated gene therapy will dramatically change the treatment pattern of childhood neurological disorders, and many children with incurable diseases will meet the dawn of a cure. Nevertheless, the vectors must be optimized for better safety and efficacy.
Collapse
Affiliation(s)
- Jing Peng
- Department of Pediatrics, Clinical Research Center for Chidren Neurodevelopmental disablities of Hunan Province, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Wei-Wei Zou
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiao-Lei Wang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhi-Guo Zhang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ran Huo
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Li Yang
- Department of Pediatrics, Clinical Research Center for Chidren Neurodevelopmental disablities of Hunan Province, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
2
|
Kumar SRP, Biswas M, Cao D, Arisa S, Muñoz-Melero M, Lam AK, Piñeros AR, Kapur R, Kaisho T, Kaufman RJ, Xiao W, Shayakhmetov DM, Terhorst C, de Jong YP, Herzog RW. TLR9-independent CD8 + T cell responses in hepatic AAV gene transfer through IL-1R1-MyD88 signaling. Mol Ther 2024; 32:325-339. [PMID: 38053332 PMCID: PMC10861967 DOI: 10.1016/j.ymthe.2023.11.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/14/2023] [Accepted: 11/30/2023] [Indexed: 12/07/2023] Open
Abstract
Upon viral infection of the liver, CD8+ T cell responses may be triggered despite the immune suppressive properties that manifest in this organ. We sought to identify pathways that activate responses to a neoantigen expressed in hepatocytes, using adeno-associated viral (AAV) gene transfer. It was previously established that cooperation between plasmacytoid dendritic cells (pDCs), which sense AAV genomes by Toll-like receptor 9 (TLR9), and conventional DCs promotes cross-priming of capsid-specific CD8+ T cells. Surprisingly, we find local initiation of a CD8+ T cell response against antigen expressed in ∼20% of murine hepatocytes, independent of TLR9 or type I interferons and instead relying on IL-1 receptor 1-MyD88 signaling. Both IL-1α and IL-1β contribute to this response, which can be blunted by IL-1 blockade. Upon AAV administration, IL-1-producing pDCs infiltrate the liver and co-cluster with XCR1+ DCs, CD8+ T cells, and Kupffer cells. Analogous events were observed following coagulation factor VIII gene transfer in hemophilia A mice. Therefore, pDCs have alternative means of promoting anti-viral T cell responses and participate in intrahepatic immune cell networks similar to those that form in lymphoid organs. Combined TLR9 and IL-1 blockade may broadly prevent CD8+ T responses against AAV capsid and transgene product.
Collapse
Affiliation(s)
- Sandeep R P Kumar
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN, USA
| | - Moanaro Biswas
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN, USA
| | - Di Cao
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN, USA
| | - Sreevani Arisa
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN, USA
| | - Maite Muñoz-Melero
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN, USA
| | - Anh K Lam
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN, USA
| | - Annie R Piñeros
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN, USA
| | - Reuben Kapur
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN, USA
| | - Tsuneyasu Kaisho
- Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Randal J Kaufman
- Center for Genetic Disorders and Aging Research, Samford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Weidong Xiao
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN, USA
| | - Dmitry M Shayakhmetov
- Lowance Center for Human Immunology, Emory Vaccine Center, Departments of Pediatrics and Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Cox Terhorst
- Division of Immunology, Beth Israel Deaconess Medical Center (BIDMC), Boston, MA, USA
| | - Ype P de Jong
- Division of Gastroenterology & Hepatology, Weill Cornell Medicine, New York, NY, USA
| | - Roland W Herzog
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN, USA.
| |
Collapse
|
3
|
Sauter MM, Noel H, Brandt CR. The RLR intrinsic antiviral system is expressed in neural retina and restricts lentiviral transduction of human Mueller cells. Exp Eye Res 2023; 236:109647. [PMID: 37689341 PMCID: PMC10834037 DOI: 10.1016/j.exer.2023.109647] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/24/2023] [Accepted: 09/06/2023] [Indexed: 09/11/2023]
Abstract
The retinoic acid-inducible gene I (RIG)-I-like receptor (RLR) family of RNA sensor proteins plays a key role in the innate immune response to viral nucleic acids, including viral gene delivery vectors, but little is known about the expression of RLR proteins in the retina. The purpose of this study was to characterize cell-specific expression patterns of RLR proteins in non-human primate (NHP) neural retina tissue and to examine if RLR pathway signaling restricts viral gene delivery transduction. Since RLR protein signaling converges at the mitochondrial antiviral signaling protein (MAVS), experiments were performed to determine if knockdown of MAVS affected FIVGFP transduction efficiency in the human Mueller cell line MIO-M1. Immunoblotting confirmed expression of RIG-I, melanoma differentiation-associated protein 5 (MDA5), laboratory of genetics and physiology 2 (LGP2), and MAVS proteins in MIO-M1 cells and NHP retina tissue. Double label immunofluorescence (IF) studies revealed RIG-I, LGP2, and MAVS were expressed in Mueller microglial cells in the NHP retina. In addition, LGP2 and MDA5 proteins were detected in cone and retinal ganglion cells (RGC). MDA5 was also present in a subset of calretinin positive amacrine cells, and in nuclei within the inner nuclear layer (INL). Knockdown of MAVS significantly increased the transduction efficiency of the lentiviral vector FIVGFP in MIO-M1 cells, compared to control cells. FIVGFP or AAVGFP challenge did not alter expression of the LGP2, MAVS, MDA5 or RIG-I genes in MIO-M1 cells or NHP retina tissue compared to media treated controls. Our data demonstrate that innate immune response proteins involved in viral RNA sensing, including MDA5, RIG-I, LGP2, and MAVS, are expressed in several cell types within the NHP neural retina. In addition, the MAVS protein restricts non-human lentiviral transduction efficiency in MIO-M1 cells.
Collapse
Affiliation(s)
- Monica M Sauter
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Hongyu Noel
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Curtis R Brandt
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53706, USA; Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, 53706, USA; McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, 53706, USA.
| |
Collapse
|
4
|
Wang Y, Shao W. Innate Immune Response to Viral Vectors in Gene Therapy. Viruses 2023; 15:1801. [PMID: 37766208 PMCID: PMC10536768 DOI: 10.3390/v15091801] [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: 07/12/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023] Open
Abstract
Viral vectors play a pivotal role in the field of gene therapy, with several related drugs having already gained clinical approval from the EMA and FDA. However, numerous viral gene therapy vectors are currently undergoing pre-clinical research or participating in clinical trials. Despite advancements, the innate response remains a significant barrier impeding the clinical development of viral gene therapy. The innate immune response to viral gene therapy vectors and transgenes is still an important reason hindering its clinical development. Extensive studies have demonstrated that different DNA and RNA sensors can detect adenoviruses, adeno-associated viruses, and lentiviruses, thereby activating various innate immune pathways such as Toll-like receptor (TLR), cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING), and retinoic acid-inducible gene I-mitochondrial antiviral signaling protein (RLR-MAVS). This review focuses on elucidating the mechanisms underlying the innate immune response induced by three widely utilized viral vectors: adenovirus, adeno-associated virus, and lentivirus, as well as the strategies employed to circumvent innate immunity.
Collapse
Affiliation(s)
| | - Wenwei Shao
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin 300072, China;
| |
Collapse
|
5
|
Chen SY, Jou IM, Ko PY, Hsu KL, Su WR, Kuo LC, Lee PY, Wu CL, Wu PT. Amelioration of experimental tendinopathy by lentiviral CD44 gene therapy targeting senescence-associated secretory phenotypes. Mol Ther Methods Clin Dev 2022; 26:157-168. [PMID: 35846572 PMCID: PMC9254001 DOI: 10.1016/j.omtm.2022.06.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 06/07/2022] [Indexed: 01/14/2023]
Abstract
CD44 exerts anti-senescence effects in many disease models. We examined senescence in tendinopathy and the effect of CD44 on senescence-associated secretory phenotypes (SASPs). Senescent markers were determined in human tendinopathic long head of bicep (LHB) and normal hamstring tendons. CD44 gene transfer in rat tendinopathic tenocytes stimulated with interleukin (IL)-1β and a rat Achilles tendinopathy model were performed using lentiviral vectors. Expression levels of p53, p21, and p16 and senescence-associated β-galactosidase (SA-β-gal) activity were positively correlated with the severity of human tendinopathy and were higher in rat and human tendinopathic tenocytes than in normal controls. CD44 overexpressed tenocyte transfectants exhibited reduced levels of IL-6, matrix metalloproteinases (MMPs), cyclooxygenase (COX)-2, p53, p21, p16, SA-β-gal, and phospho-nuclear factor (NF)-κB, whereas their collagen type I alpha 1 (COL1A1) and tenomodulin (tnmd) levels were increased when compared with control transfectants under IL-1β-stimulated conditions. In the animal model, CD44 overexpression lowered the ultrasound and histology scores and expression levels of the senescent and SASP markers COX-2 and phospho-NF-κB. Bromodeoxyuridine (BrdU)- and tnmd-positive cell numbers were increased in the LVCD44-transduced tendinopathic tendons. Senescence is positively correlated with tendinopathic severity, and CD44 overexpression may protect the tendinopathic tendons from SASPs via anti-inflammation and maintenance of extracellular matrix homeostasis.
Collapse
Affiliation(s)
- Shih-Yao Chen
- Department of Nursing, College of Nursing, Chung Hwa University of Medical Technology, Tainan 71703, Taiwan
| | - I-Ming Jou
- Department of Orthopaedics, E-Da Hospital, Kaohsiung 82445, Taiwan.,School of Medicine, College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan
| | - Po-Yen Ko
- Department of Orthopaedics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan.,Department of Biomedical Engineering, National Cheng Kung University, Tainan 70101, Taiwan
| | - Kai-Lan Hsu
- Department of Orthopaedics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan.,Department of Biomedical Engineering, National Cheng Kung University, Tainan 70101, Taiwan
| | - Wei-Ren Su
- Department of Orthopaedics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan.,Department of Orthopaedics, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Li-Chieh Kuo
- Department of Occupational Therapy, National Cheng Kung University, Tainan 70101, Taiwan.,Medical Device Innovation Center, National Cheng Kung University, Tainan 70101, Taiwan
| | - Pei-Yuan Lee
- Department of Orthopaedic Surgery, Show Chwan Memorial Hospital, Changhua 50544, Taiwan.,College of Nursing and Health Sciences, Da-Yeh University, Changhua 51500, Taiwan
| | - Chao-Liang Wu
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan.,Department of Medical Research, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 60002, Taiwan
| | - Po-Ting Wu
- Department of Orthopaedics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan.,Department of Biomedical Engineering, National Cheng Kung University, Tainan 70101, Taiwan.,Department of Orthopaedics, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan.,Medical Device Innovation Center, National Cheng Kung University, Tainan 70101, Taiwan.,Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| |
Collapse
|
6
|
Piras F, Kajaste-Rudnitski A. Antiviral immunity and nucleic acid sensing in haematopoietic stem cell gene engineering. Gene Ther 2021; 28:16-28. [PMID: 32661282 PMCID: PMC7357672 DOI: 10.1038/s41434-020-0175-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 07/06/2020] [Indexed: 02/06/2023]
Abstract
The low gene manipulation efficiency of human hematopoietic stem and progenitor cells (HSPC) remains a major hurdle for sustainable and broad clinical application of innovative therapies for a wide range of disorders. Given that all current and emerging gene transfer and editing technologies are bound to expose HSPC to exogenous nucleic acids and most often also to viral vectors, we reason that host antiviral factors and nucleic acid sensors play a pivotal role in the efficacy of HSPC genetic manipulation. Here, we review recent progress in our understanding of vector-host interactions and innate immunity in HSPC upon gene engineering and discuss how dissecting this crosstalk can guide the development of more stealth and efficient gene therapy approaches in the future.
Collapse
Affiliation(s)
- Francesco Piras
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Anna Kajaste-Rudnitski
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
7
|
Wang X, Fu RY, Li C, Chen CY, Firrman J, Konkle BA, Zhang J, Li L, Xiao W, Poncz M, Miao CH. Enhancing therapeutic efficacy of in vivo platelet-targeted gene therapy in hemophilia A mice. Blood Adv 2020; 4:5722-5734. [PMID: 33216891 PMCID: PMC7686911 DOI: 10.1182/bloodadvances.2020002479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 10/15/2020] [Indexed: 11/20/2022] Open
Abstract
Our previous studies demonstrated that intraosseous (IO) infusion of lentiviral vectors (LVs) carrying a modified B domain-deleted factor VIII (FVIII) transgene driven by a megakaryocyte-specific promoter (GP1Bα promoter; G-F8/N6-LV) successfully transduced hematopoietic stem cells (HSCs) to produce FVIII stored in the platelet α-granules. Platelet FVIII corrected the bleeding phenotype with limited efficacy in hemophilia A (HemA) mice with and without preexisting anti-FVIII inhibitors. The present study sought to further enhance the therapeutic efficacy of this treatment protocol by increasing both the efficiency of LV transduction and the functional activity of platelet FVIII. A combined drug regimen of dexamethasone and anti-CD8α monoclonal antibody enhanced the percentage of transduced bone marrow and HSCs over time. In G-F8/N6-LV-treated HemA mice, significant improvement in phenotypic correction was observed on day 84. To improve platelet FVIII functionality, genes encoding FVIII variant F8X10K12 with increased expression or F8N6K12RH with increased functional activity compared with F8/N6 were incorporated into LVs. Treatment with G-F8X10K12-LV in HemA mice produced a higher level of platelet FVIII but induced anti-FVIII inhibitors. After treatment with combined drugs and IO infusion of G-F8/N6K12RH-LV, HemA mice showed significant phenotypic correction without anti-FVIII inhibitor formation. These results indicate that new human FVIII variant F8/N6K12RH combined with immune suppression could significantly enhance the therapeutic efficacy of in vivo platelet-targeted gene therapy for murine HemA via IO delivery. This protocol provides a safe and effective treatment for hemophilia that may be translatable to and particularly beneficial for patients with preexisting inhibitory antibodies to FVIII.
Collapse
Affiliation(s)
- Xuefeng Wang
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA
| | - Richard Y Fu
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA
| | - Chong Li
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA
| | - Chun-Yu Chen
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA
| | - Jenni Firrman
- Department of Pediatrics, Indiana University, Indianapolis, IN
| | | | - Junping Zhang
- Department of Pediatrics, Indiana University, Indianapolis, IN
| | - Lei Li
- Department of Chemistry, Georgia State University, Atlanta, GA; and
| | - Weidong Xiao
- Department of Pediatrics, Indiana University, Indianapolis, IN
| | - Mortimer Poncz
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Carol H Miao
- Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA
| |
Collapse
|
8
|
Milani M, Annoni A, Moalli F, Liu T, Cesana D, Calabria A, Bartolaccini S, Biffi M, Russo F, Visigalli I, Raimondi A, Patarroyo-White S, Drager D, Cristofori P, Ayuso E, Montini E, Peters R, Iannacone M, Cantore A, Naldini L. Phagocytosis-shielded lentiviral vectors improve liver gene therapy in nonhuman primates. Sci Transl Med 2020; 11:11/493/eaav7325. [PMID: 31118293 DOI: 10.1126/scitranslmed.aav7325] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 01/11/2019] [Accepted: 05/01/2019] [Indexed: 12/13/2022]
Abstract
Liver-directed gene therapy for the coagulation disorder hemophilia showed safe and effective results in clinical trials using adeno-associated viral vectors to replace a functional coagulation factor, although some unmet needs remain. Lentiviral vectors (LVs) may address some of these hurdles because of their potential for stable expression and the low prevalence of preexisting viral immunity in humans. However, systemic LV administration to hemophilic dogs was associated to mild acute toxicity and low efficacy at the administered doses. Here, exploiting intravital microscopy and LV surface engineering, we report a major role of the human phagocytosis inhibitor CD47, incorporated into LV cell membrane, in protecting LVs from uptake by professional phagocytes and innate immune sensing, thus favoring biodistribution to hepatocytes after systemic administration. By enforcing high CD47 surface content, we generated phagocytosis-shielded LVs which, upon intravenous administration to nonhuman primates, showed selective liver and spleen targeting and enhanced hepatocyte gene transfer compared to parental LV, reaching supraphysiological activity of human coagulation factor IX, the protein encoded by the transgene, without signs of toxicity or clonal expansion of transduced cells.
Collapse
Affiliation(s)
- Michela Milani
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy.,Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Andrea Annoni
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | | | | | - Daniela Cesana
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Andrea Calabria
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Sara Bartolaccini
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Mauro Biffi
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Fabio Russo
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Ilaria Visigalli
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | | | | | | | - Patrizia Cristofori
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy.,GlaxoSmithKline R&D UK, Ware SG12 0DP, UK
| | - Eduard Ayuso
- INSERM UMR1089, University of Nantes, CHU de Nantes, 44093 Nantes, France
| | - Eugenio Montini
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | | | | | - Alessio Cantore
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy. .,Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Luigi Naldini
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy. .,Vita-Salute San Raffaele University, 20132 Milan, Italy
| |
Collapse
|
9
|
Cousin C, Oberkampf M, Felix T, Rosenbaum P, Weil R, Fabrega S, Morante V, Negri D, Cara A, Dadaglio G, Leclerc C. Persistence of Integrase-Deficient Lentiviral Vectors Correlates with the Induction of STING-Independent CD8 + T Cell Responses. Cell Rep 2020; 26:1242-1257.e7. [PMID: 30699352 DOI: 10.1016/j.celrep.2019.01.025] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 11/22/2018] [Accepted: 01/07/2019] [Indexed: 12/23/2022] Open
Abstract
Lentiviruses are among the most promising viral vectors for in vivo gene delivery. To overcome the risk of insertional mutagenesis, integrase-deficient lentiviral vectors (IDLVs) have been developed. We show here that strong and persistent specific cytotoxic T cell (CTL) responses are induced by IDLVs, which persist several months after a single injection. These responses were associated with the induction of mild and transient maturation of dendritic cells (DCs) and with the production of low levels of inflammatory cytokines and chemokines. They were independent of the IFN-I, TLR/MyD88, interferon regulatory factor (IRF), retinoic acid induced gene I (RIG-I), and stimulator of interferon genes (STING) pathways but require NF-κB signaling in CD11c+ DCs. Despite the lack of integration of IDLVs, the transgene persists for 3 months in the spleen and liver of IDLV-injected mice. These results demonstrate that the capacity of IDLVs to trigger persistent adaptive responses is mediated by a weak and transient innate response, along with the persistence of the vector in tissues.
Collapse
Affiliation(s)
- Céline Cousin
- Institut Pasteur, Unité de Régulation Immunitaire et Vaccinologie, Equipe Labellisée Ligue Contre le Cancer, 75015 Paris, France; INSERM U1041, 75015 Paris, France
| | - Marine Oberkampf
- Institut Pasteur, Unité de Régulation Immunitaire et Vaccinologie, Equipe Labellisée Ligue Contre le Cancer, 75015 Paris, France; INSERM U1041, 75015 Paris, France
| | - Tristan Felix
- Institut Pasteur, Unité de Régulation Immunitaire et Vaccinologie, Equipe Labellisée Ligue Contre le Cancer, 75015 Paris, France; INSERM U1041, 75015 Paris, France
| | - Pierre Rosenbaum
- Institut Pasteur, Unité de Régulation Immunitaire et Vaccinologie, Equipe Labellisée Ligue Contre le Cancer, 75015 Paris, France; INSERM U1041, 75015 Paris, France
| | - Robert Weil
- Institut Pasteur, Unité Signalisation et Pathogénèse, Département Biologie Cellulaire et Infection, 75015 Paris, France
| | - Sylvie Fabrega
- Plateforme Vecteurs Viraux et Transfert de Gènes, SFR Necker, US 24, UMS 3633, 75014 Paris, France
| | - Valeria Morante
- Department of Infection Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Donatella Negri
- Department of Infection Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Andrea Cara
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | - Gilles Dadaglio
- Institut Pasteur, Unité de Régulation Immunitaire et Vaccinologie, Equipe Labellisée Ligue Contre le Cancer, 75015 Paris, France; INSERM U1041, 75015 Paris, France.
| | - Claude Leclerc
- Institut Pasteur, Unité de Régulation Immunitaire et Vaccinologie, Equipe Labellisée Ligue Contre le Cancer, 75015 Paris, France; INSERM U1041, 75015 Paris, France.
| |
Collapse
|
10
|
Carbonaro-Sarracino DA, Tarantal AF, Lee CCI, Kaufman ML, Wandro S, Jin X, Martinez M, Clark DN, Chun K, Koziol C, Hardee CL, Wang X, Kohn DB. Dosing and Re-Administration of Lentiviral Vector for In Vivo Gene Therapy in Rhesus Monkeys and ADA-Deficient Mice. Mol Ther Methods Clin Dev 2020; 16:78-93. [PMID: 31871959 PMCID: PMC6909201 DOI: 10.1016/j.omtm.2019.11.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 11/04/2019] [Indexed: 12/21/2022]
Abstract
Adenosine deaminase (ADA)-deficient mice and healthy rhesus monkeys were studied to determine the impact of age at treatment, vector dosage, dosing schedule, repeat administration, biodistribution, and immunogenicity after systemic delivery of lentiviral vectors (LVs). In Ada -/- mice, neonatal treatment resulted in broad vector marking across all tissues analyzed, whereas adult treatment resulted in marking restricted to the liver, spleen, and bone marrow. Intravenous administration to infant rhesus monkeys also resulted in dose-dependent marking in the liver, spleen, and bone marrow. Using an ELISA to monitor anti-vector antibody development, Ada -/- neonatal mice did not produce an antibody response, whereas Ada -/- adult mice produced a strong antibody response to vector administration. In mice and monkeys with repeat administration of LV, a strong anti-vector antibody response was shown in response to the second LV administration, which resulted in LV inactivation. Three separate doses administered to immune competent mice resulted in acute toxicity. Pegylation of the vesicular stomatitis virus G protein (VSV-G)-enveloped LVs showed a less robust anti-vector response but did not prevent the inactivation of the second LV administration. These studies identify important factors to consider related to age and timing of administration when implementing systemic delivery of LVs as a potential therapeutic agent.
Collapse
Affiliation(s)
- Denise A. Carbonaro-Sarracino
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Alice F. Tarantal
- Center for Fetal Monkey Gene Transfer for Heart, Lung, and Blood Diseases, University of California, Davis, Davis, CA 95616, USA
- Departments of Pediatrics and Cell Biology and Human Anatomy, School of Medicine, and California National Primate Research Center, University of California, Davis, Davis, CA 95616, USA
| | - C. Chang I. Lee
- Center for Fetal Monkey Gene Transfer for Heart, Lung, and Blood Diseases, University of California, Davis, Davis, CA 95616, USA
- Departments of Pediatrics and Cell Biology and Human Anatomy, School of Medicine, and California National Primate Research Center, University of California, Davis, Davis, CA 95616, USA
| | - Michael L. Kaufman
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Stephen Wandro
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Xiangyang Jin
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Michele Martinez
- Center for Fetal Monkey Gene Transfer for Heart, Lung, and Blood Diseases, University of California, Davis, Davis, CA 95616, USA
- Departments of Pediatrics and Cell Biology and Human Anatomy, School of Medicine, and California National Primate Research Center, University of California, Davis, Davis, CA 95616, USA
| | - Danielle N. Clark
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Krista Chun
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Colin Koziol
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Cinnamon L. Hardee
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Xiaoyan Wang
- Department of General Internal Medicine and Health Services Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Donald B. Kohn
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- The Eli & Edythe Broad Center for Stem Cells and Regenerative Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
11
|
Abstract
Several viral vector-based gene therapy drugs have now received marketing approval. A much larger number of additional viral vectors are in various stages of clinical trials for the treatment of genetic and acquired diseases, with many more in pre-clinical testing. Efficiency of gene transfer and ability to provide long-term therapy make these vector systems very attractive. In fact, viral vector gene therapy has been able to treat or even cure diseases for which there had been no or only suboptimal treatments. However, innate and adaptive immune responses to these vectors and their transgene products constitute substantial hurdles to clinical development and wider use in patients. This review provides an overview of the type of immune responses that have been documented in animal models and in humans who received gene transfer with one of three widely tested vector systems, namely adenoviral, lentiviral, or adeno-associated viral vectors. Particular emphasis is given to mechanisms leading to immune responses, efforts to reduce vector immunogenicity, and potential solutions to the problems. At the same time, we point out gaps in our knowledge that should to be filled and problems that need to be addressed going forward.
Collapse
Affiliation(s)
- Jamie L Shirley
- Gene Therapy Center, University of Massachusetts, Worchester, MA, USA
| | - Ype P de Jong
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, NY, USA
| | - Cox Terhorst
- Division of Immunology, Beth Israel Deaconess Medical Center (BIDMC), Harvard Medical School, Boston, MA, USA
| | - Roland W Herzog
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
12
|
Abstract
Functional studies of specific stem cell populations often require depletion of tissue-specific stem cells in an in vivo model to allow for the interrogation of their contribution to the maintenance and/or regeneration of their home tissue. Depletion methods need an exquisite specificity to uniquely eliminate the target cell type. To achieve such specificity, a commonly used approach has been murine models with expression of the Diphtheria Toxin Receptor (DTR) in the cell of interest. The major caveat of using these DTR-expressing transgenic mice is the need to generate new DTR models for every new cell population of interest. While DTR-expressing models are limited, the number of available GFP-expressing mice is large. To take advantage of this plethora of cell type-specific GFP-reporter mice, we sought to exploit the body's own killer cells as a depletion tool. Thus, we generated a mouse model whose cytotoxic T cells recognize and kill GFP-expressing cells, called the Jedi (Agudo et al., Nat Biotechnol 33:1287-1292, 2015). Jedi T cells now enable the depletion of virtually almost any cell type by using a suitable GFP-expressing transgenic mouse (Agudo et al., Nat Biotechnol 33:1287-1292, 2015; Chen et al., J Clin Invest 128(8):3413-3424, 2018). Here, we explain in detail how to achieve depletion of Lgr5+ stem cells in the intestine with a single injection of Jedi T cells (Agudo et al., Immunity 48:271-285.e5, 2018) with a methodology that can be extrapolated to any other GFP-expressing cell.
Collapse
Affiliation(s)
- Stephen E Sherman
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Judith Agudo
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Immunology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
13
|
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
|
14
|
Piras F, Riba M, Petrillo C, Lazarevic D, Cuccovillo I, Bartolaccini S, Stupka E, Gentner B, Cittaro D, Naldini L, Kajaste-Rudnitski A. Lentiviral vectors escape innate sensing but trigger p53 in human hematopoietic stem and progenitor cells. EMBO Mol Med 2018; 9:1198-1211. [PMID: 28667090 PMCID: PMC5582409 DOI: 10.15252/emmm.201707922] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Clinical application of lentiviral vector (LV)-based hematopoietic stem and progenitor cells (HSPC) gene therapy is rapidly becoming a reality. Nevertheless, LV-mediated signaling and its potential functional consequences on HSPC biology remain poorly understood. We unravel here a remarkably limited impact of LV on the HSPC transcriptional landscape. LV escaped innate immune sensing that instead led to robust IFN responses upon transduction with a gamma-retroviral vector. However, reverse-transcribed LV DNA did trigger p53 signaling, activated also by non-integrating Adeno-associated vector, ultimately leading to lower cell recovery ex vivo and engraftment in vivo These effects were more pronounced in the short-term repopulating cells while long-term HSC frequencies remained unaffected. Blocking LV-induced signaling partially rescued both apoptosis and engraftment, highlighting a novel strategy to further dampen the impact of ex vivo gene transfer on HSPC. Overall, our results shed light on viral vector sensing in HSPC and provide critical insight for the development of more stealth gene therapy strategies.
Collapse
Affiliation(s)
- Francesco Piras
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Michela Riba
- Center for Translational Genomics and Bioinformatics, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Carolina Petrillo
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Dejan Lazarevic
- Center for Translational Genomics and Bioinformatics, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ivan Cuccovillo
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Sara Bartolaccini
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elia Stupka
- Center for Translational Genomics and Bioinformatics, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Bernhard Gentner
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Davide Cittaro
- Center for Translational Genomics and Bioinformatics, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luigi Naldini
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Anna Kajaste-Rudnitski
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
15
|
Modulation of immune responses in lentiviral vector-mediated gene transfer. Cell Immunol 2018; 342:103802. [PMID: 29735164 PMCID: PMC6695505 DOI: 10.1016/j.cellimm.2018.04.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 04/25/2018] [Accepted: 04/26/2018] [Indexed: 02/07/2023]
Abstract
Lentiviral vectors (LV) are widely used vehicles for gene transfer and therapy in pre-clinical animal models and clinical trials with promising safety and efficacy results. However, host immune responses against vector- and/or transgene-derived antigens remain a major obstacle to the success and broad applicability of gene therapy. Here we review the innate and adaptive immunological barriers to successful gene therapy, both in the context of ex vivo and in vivo LV gene therapy, mostly concerning systemic LV delivery and discuss possible means to overcome them, including vector design and production and immune modulatory strategies.
Collapse
|
16
|
Biswas M, Kumar SRP, Terhorst C, Herzog RW. Gene Therapy With Regulatory T Cells: A Beneficial Alliance. Front Immunol 2018; 9:554. [PMID: 29616042 PMCID: PMC5868074 DOI: 10.3389/fimmu.2018.00554] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 03/05/2018] [Indexed: 12/12/2022] Open
Abstract
Gene therapy aims to replace a defective or a deficient protein at therapeutic or curative levels. Improved vector designs have enhanced safety, efficacy, and delivery, with potential for lasting treatment. However, innate and adaptive immune responses to the viral vector and transgene product remain obstacles to the establishment of therapeutic efficacy. It is widely accepted that endogenous regulatory T cells (Tregs) are critical for tolerance induction to the transgene product and in some cases the viral vector. There are two basic strategies to harness the suppressive ability of Tregs: in vivo induction of adaptive Tregs specific to the introduced gene product and concurrent administration of autologous, ex vivo expanded Tregs. The latter may be polyclonal or engineered to direct specificity to the therapeutic antigen. Recent clinical trials have advanced adoptive immunotherapy with Tregs for the treatment of autoimmune disease and in patients receiving cell transplants. Here, we highlight the potential benefit of combining gene therapy with Treg adoptive transfer to achieve a sustained transgene expression. Furthermore, techniques to engineer antigen-specific Treg cell populations, either through reprogramming conventional CD4+ T cells or transferring T cell receptors with known specificity into polyclonal Tregs, are promising in preclinical studies. Thus, based upon these observations and the successful use of chimeric (IgG-based) antigen receptors (CARs) in antigen-specific effector T cells, different types of CAR-Tregs could be added to the repertoire of inhibitory modalities to suppress immune responses to therapeutic cargos of gene therapy vectors. The diverse approaches to harness the ability of Tregs to suppress unwanted immune responses to gene therapy and their perspectives are reviewed in this article.
Collapse
Affiliation(s)
- Moanaro Biswas
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, FL, United States
| | - Sandeep R P Kumar
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, FL, United States
| | - Cox Terhorst
- Division of Immunology, Beth Israel Deaconess Medical Center (BIDMC), Harvard Medical School, Boston, MA, United States
| | - Roland W Herzog
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, FL, United States
| |
Collapse
|
17
|
Sherman A, Biswas M, Herzog RW. Innovative Approaches for Immune Tolerance to Factor VIII in the Treatment of Hemophilia A. Front Immunol 2017; 8:1604. [PMID: 29225598 PMCID: PMC5705551 DOI: 10.3389/fimmu.2017.01604] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 11/07/2017] [Indexed: 01/19/2023] Open
Abstract
Hemophilia A (coagulation factor VIII deficiency) is a debilitating genetic disorder that is primarily treated with intravenous replacement therapy. Despite a variety of factor VIII protein formulations available, the risk of developing anti-dug antibodies (“inhibitors”) remains. Overall, 20–30% of patients with severe disease develop inhibitors. Current clinical immune tolerance induction protocols to eliminate inhibitors are not effective in all patients, and there are no prophylactic protocols to prevent the immune response. New experimental therapies, such as gene and cell therapies, show promising results in pre-clinical studies in animal models of hemophilia. Examples include hepatic gene transfer with viral vectors, genetically engineered regulatory T cells (Treg), in vivo Treg induction using immune modulatory drugs, and maternal antigen transfer. Furthermore, an oral tolerance protocol is being developed based on transgenic lettuce plants, which suppressed inhibitor formation in hemophilic mice and dogs. Hopefully, some of these innovative approaches will reduce the risk of and/or more effectively eliminate inhibitor formation in future treatment of hemophilia A.
Collapse
Affiliation(s)
- Alexandra Sherman
- Department of Pediatrics, University of Florida, Gainesville, FL, United States
| | - Moanaro Biswas
- Department of Pediatrics, University of Florida, Gainesville, FL, United States
| | - Roland W Herzog
- Department of Pediatrics, University of Florida, Gainesville, FL, United States
| |
Collapse
|
18
|
Initial Considerations Before Designing a Promoter Construct. Methods Mol Biol 2017. [PMID: 28801895 DOI: 10.1007/978-1-4939-7223-4_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Before designing a synthetic promoter, it can be helpful to think about its final application. Is the study purely an in vitro exercise in monitoring short-term promoter activity from an episomal vector, or does the promoter eventually need to be permanently active and be integrated into the genome or perhaps even to function in vivo? The final application will have a bearing on promoter design and vector of choice from the start of the study. In this chapter I highlight some of the vector attributes to consider and features that should be thought about.
Collapse
|
19
|
Plasmacytoid and conventional dendritic cells cooperate in crosspriming AAV capsid-specific CD8 + T cells. Blood 2017; 129:3184-3195. [PMID: 28468798 DOI: 10.1182/blood-2016-11-751040] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 05/01/2017] [Indexed: 02/06/2023] Open
Abstract
Adeno-associated virus (AAV) is a replication-deficient parvovirus that is extensively used as a gene therapy vector. CD8+ T-cell responses against the AAV capsid protein can, however, affect therapeutic efficacy. Little is known about the in vivo mechanism that leads to the crosspriming of CD8+ T cells against the input viral capsid antigen. In this study, we report that the Toll-like receptor 9 (TLR9)-MyD88 pattern-recognition receptor pathway is uniquely capable of initiating this response. By contrast, the absence of TLR2, STING, or the addition of TLR4 agonist has no effect. Surprisingly, both conventional dendritic cells (cDCs) and plasmacytoid DCs (pDCs) are required for the crosspriming of capsid-specific CD8+ T cells, whereas other antigen-presenting cells are not involved. TLR9 signaling is specifically essential in pDCs but not in cDCs, indicating that sensing of the viral genome by pDCs activates cDCs in trans to cross-present capsid antigen during CD8+ T-cell activation. Cross-presentation and crosspriming depend not only on TLR9, but also on interferon type I signaling, and both mechanisms can be inhibited by administering specific molecules to prevent induction of capsid-specific CD8+ T cells. Thus, these outcomes directly point to therapeutic interventions and demonstrate that innate immune blockade can eliminate unwanted immune responses in gene therapy.
Collapse
|
20
|
Abstract
Current treatment of hemophilia A (HemA) patients with repeated infusions of factor VIII (FVIII; abbreviated as F8 in constructs) is costly, inconvenient, and incompletely effective. In addition, approximately 25 % of treated patients develop anti-factor VIII immune responses. Gene therapy that can achieve long-term phenotypic correction without the complication of anti-factor VIII antibody formation is highly desired. Lentiviral vector (LV)-mediated gene transfer into hematopoietic stem cells (HSCs) results in stable integration of FVIII gene into the host genome, leading to persistent therapeutic effect. However, ex vivo HSC gene therapy requires pre-conditioning which is highly undesirable for hemophilia patients. The recently developed novel methodology of direct intraosseous (IO) delivery of LVs can efficiently transduce bone marrow cells, generating high levels of transgene expression in HSCs. IO delivery of E-F8-LV utilizing a ubiquitous EF1α promoter generated initially therapeutic levels of FVIII, however, robust anti-FVIII antibody responses ensued neutralized functional FVIII activity in the circulation. In contrast, a single IO delivery of G-FVIII-LV utilizing a megakaryocytic-specific GP1bα promoter achieved platelet-specific FVIII expression, leading to persistent, partial correction of HemA in treated animals. Most interestingly, comparable therapeutic benefit with G-F8-LV was obtained in HemA mice with pre-existing anti-FVIII inhibitors. Platelets is an ideal IO delivery vehicle since FVIII stored in α-granules of platelets is protected from high-titer anti-FVIII antibodies; and that even relatively small numbers of activated platelets that locally excrete FVIII may be sufficient to promote efficient clot formation during bleeding. Additionally, combination of pharmacological agents improved transduction of LVs and persistence of transduced cells and transgene expression. Overall, a single IO infusion of G-F8-LV can generate long-term stable expression of hFVIII in platelets and correct hemophilia phenotype for long term. This approach has high potential to permanently treat FVIII deficiency with and without pre-existing anti-FVIII antibodies.
Collapse
Affiliation(s)
- Carol H Miao
- Seattle Children's Research Institute, Seattle, WA USA ; Department of Pediatrics, University of Washington, Seattle, WA USA
| |
Collapse
|
21
|
Borsotti C, Borroni E, Follenzi A. Lentiviral vector interactions with the host cell. Curr Opin Virol 2016; 21:102-108. [PMID: 27637073 DOI: 10.1016/j.coviro.2016.08.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 08/29/2016] [Accepted: 08/31/2016] [Indexed: 10/21/2022]
Abstract
Lentiviral vectors (LVs)-mediated gene transfer is an efficient method for ex vivo and in vivo gene therapy. Actually, LVs have been used in several clinical trials and therapeutic correction was reached in affected patients. However, in order to be effective gene therapy needs to be efficient without detrimental effects for target cells. Successful cell transduction by LVs can be hampered by several factors such as the activation of innate immune sensors during cell transduction and different restriction factors (RFs) inhibiting viral replication inside the cells. Therefore, a better knowledge of host-vector interactions is important for the development of more efficient gene therapy strategies improving the LVs platform by limiting harmful responses.
Collapse
Affiliation(s)
- Chiara Borsotti
- Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara 28100, Italy
| | - Ester Borroni
- Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara 28100, Italy
| | - Antonia Follenzi
- Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara 28100, Italy.
| |
Collapse
|
22
|
Aranyi T, Stockholm D, Yao R, Poinsignon C, Wiart T, Corre G, Touleimat N, Tost J, Galy A, Paldi A. Systemic epigenetic response to recombinant lentiviral vectors independent of proviral integration. Epigenetics Chromatin 2016; 9:29. [PMID: 27408621 PMCID: PMC4940770 DOI: 10.1186/s13072-016-0077-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 06/28/2016] [Indexed: 01/03/2023] Open
Abstract
Background Lentiviral vectors (LV) are widely used for various gene transfer or gene therapy applications. The effects of LV on target cells are expected to be limited to gene delivery. Yet, human hematopoietic CD34+ cells respond to functional LVs as well as several types of non-integrating LVs by genome-wide DNA methylation changes. Results A new algorithm for the analysis of 450K Illumina data showed that these changes were marked by de novo methylation. The same 4126 cytosines located in islands corresponding to 1059 genes were systematically methylated. This effect required cellular entry of the viral particle in the cells but not the genomic integration of the vector cassette. Some LV preparations induced only mild sporadic changes while others had strong effects suggesting that LV batch heterogeneity may be related to the extent of the epigenetic response. Conclusion These findings identify a previously uncharacterized but consistent cellular response to viral components and provide a novel example of environmentally modified epigenome. Electronic supplementary material The online version of this article (doi:10.1186/s13072-016-0077-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tamas Aranyi
- Université Evry Val d'Essonne, UMRS_951, Genethon, 91002 Evry, France
| | - Daniel Stockholm
- Ecole Pratique des Hautes Etudes, PSL Research University, UMRS_951, Genethon, 1 bis rue de l'Internationale, 91002 Evry, France
| | | | | | | | | | - Nizar Touleimat
- Centre National de Génotypage, CEA-Institut de Génomique, 2, rue Gaston Crémieux, 91000 Evry, France
| | - Jörg Tost
- Centre National de Génotypage, CEA-Institut de Génomique, 2, rue Gaston Crémieux, 91000 Evry, France
| | - Anne Galy
- Inserm, U951, Genethon, 1 bis rue de l'Internationale, 91002 Evry, France.,Genethon, 91002 Evry, France
| | - Andràs Paldi
- Ecole Pratique des Hautes Etudes, PSL Research University, UMRS_951, Genethon, 1 bis rue de l'Internationale, 91002 Evry, France
| |
Collapse
|
23
|
|
24
|
Griffin DO, Goff SP. Restriction of HIV-1-based lentiviral vectors in adult primary marrow-derived and peripheral mobilized human CD34+ hematopoietic stem and progenitor cells occurs prior to viral DNA integration. Retrovirology 2016; 13:14. [PMID: 26945863 PMCID: PMC4779582 DOI: 10.1186/s12977-016-0246-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 02/18/2016] [Indexed: 01/10/2023] Open
Abstract
Background Gene therapy is currently being attempted using a number of delivery vehicles including lentiviral-based vectors. The delivery and insertion of a gene using lentiviral-based vectors involves multiple discrete steps, including reverse transcription of viral RNA into DNA, nuclear entry, integration of viral DNA into the host genome and expression of integrated genes. Transduction of murine stem cells by the murine leukemia viruses is inefficient because the expression of the integrated DNA is profoundly blocked. Transduction of human stem cells by lentivirus vectors is also inefficient, but the cause and specific part of the retroviral lifecycle where this block occurs is unknown. Results Here we demonstrate that the dominant point of restriction of an HIV-1-based lentiviral vector in adult human hematopoietic stem and progenitor cells (HSPCs) from bone marrow and also those obtained following peripheral mobilization is prior to viral DNA integration. We specifically show that restriction of HSPCs to an HIV-1-based lentiviral vector is prior to formation of nuclear DNA forms. Conclusions Murine restriction of MLV and human cellular restriction of HIV-1 are fundamentally different. While murine restriction of MLV occurs post integration, human restriction of HIV-1 occurs before integration.
Collapse
Affiliation(s)
- Daniel O Griffin
- Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, HHSC 1310c, 701 West 168th Street, New York, NY, 10032, USA. .,Division of Infectious Diseases, Department of Medicine, Columbia University Medical Center, New York, NY, 10032, USA.
| | - Stephen P Goff
- Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, HHSC 1310c, 701 West 168th Street, New York, NY, 10032, USA. .,Howard Hughes Medical Institute, Columbia University Medical Center, New York, NY, 10032, USA. .,Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
25
|
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
|
26
|
Abstract
Hemophilia is an X-linked inherited bleeding disorder consisting of two classifications, hemophilia A and hemophilia B, depending on the underlying mutation. Although the disease is currently treatable with intravenous delivery of replacement recombinant clotting factor, this approach represents a significant cost both monetarily and in terms of quality of life. Gene therapy is an attractive alternative approach to the treatment of hemophilia that would ideally provide life-long correction of clotting activity with a single injection. In this review, we will discuss the multitude of approaches that have been explored for the treatment of both hemophilia A and B, including both in vivo and ex vivo approaches with viral and nonviral delivery vectors.
Collapse
Affiliation(s)
- Geoffrey L Rogers
- University of Florida, Department of Pediatrics, Division of Cellular and Molecular Therapy, Gainesville, FL 32610
| | - Roland W Herzog
- University of Florida, Department of Pediatrics, Division of Cellular and Molecular Therapy, Gainesville, FL 32610
| |
Collapse
|
27
|
Rogers GL, Herzog RW. One microRNA controls both angiogenesis and TLR-mediated innate immunity to nucleic acids. Mol Ther 2014; 22:249-250. [PMID: 24487566 DOI: 10.1038/mt.2013.299] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Geoffrey L Rogers
- Department of Pediatrics, University of Florida, Gainesville, Florida, USA
| | - Roland W Herzog
- Department of Pediatrics, University of Florida, Gainesville, Florida, USA.
| |
Collapse
|
28
|
Cyclosporin a and rapamycin relieve distinct lentiviral restriction blocks in hematopoietic stem and progenitor cells. Mol Ther 2014; 23:352-62. [PMID: 25270076 DOI: 10.1038/mt.2014.193] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 09/22/2014] [Indexed: 02/06/2023] Open
Abstract
Improving hematopoietic stem and progenitor cell (HSPC) permissiveness to HIV-derived lentiviral vectors (LVs) remains a challenge for the field of gene therapy as high vector doses and prolonged ex vivo culture are still required to achieve clinically relevant transduction levels. We report here that Cyclosporin A (CsA) and Rapamycin (Rapa) significantly improve LV gene transfer in human and murine HSPC. Both compounds increased LV but not gammaretroviral transduction and acted independently of calcineurin and autophagy. Improved gene transfer was achieved across all CD34(+) subpopulations, including in long-term SCID repopulating cells. Effects of CsA were specific of HSPC and opposite to its known impact on HIV replication. Mutating the Cyclophilin A binding pocket of the viral capsid (CA) further improved transduction in combination with CsA. Tracking of the LV genome fate revealed that CsA relieves a CA-dependent early block and increases integration, while Rapa acts early in LV infection independently of the viral CA. In agreement, only Rapa was able to improve transduction by an integrase-defective LV harboring wild-type CA. Overall, our findings pave the way for more efficient and sustainable LV gene therapy in human HSPCs and shed light on the multiple innate barriers specifically hampering LV transduction in these cells.
Collapse
|
29
|
Blockade of type I interferon (IFN) production by retroviral replicating vectors and reduced tumor cell responses to IFN likely contribute to tumor selectivity. J Virol 2014; 88:10066-77. [PMID: 24965455 DOI: 10.1128/jvi.02300-13] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
UNLABELLED We developed a Moloney mouse leukemia virus (MLV)-based retroviral replicating vector (RRV), Toca 511, which has displayed tumor specificity in resected brain tumor material and blood in clinical trials. Here, we investigated the interaction between Toca 511 and human host cells, and we show that RRVs do not induce type I interferon (IFN) responses in cultured human tumor cells or cultured human primary cells. However, exogenous type I IFN inhibited RRV replication in tumor cells and induced IFN-regulated genes, albeit at a lower level than in primary cells. Unexpectedly, RRVs did not induce IFN-α production upon incubation in vitro with human plasmacytoid dendritic cells (pDCs), whereas lentivirus vector and heat-treated RRVs did. Coincubation of RRVs with heat-treated RRVs or with lentivirus vector suppressed IFN-α production in pDCs, suggesting that native RRV has a dominant inhibitory effect on type I IFN induction. This effect is sensitive to trypsin treatment. In addition, heat treatment inactivated that activity but exposed an immune-stimulatory activity. The immune-stimulating component is sensitive to deglycosidases, trypsin, and phospholipase C treatment. Experiments with retroviral nonreplicating vectors and virus-like particles demonstrated that the immunosuppressive activity is not associated with the amphotropic envelope or the glyco-Gag protein. In summary, our data provide evidence that RRVs do not directly trigger type I IFN responses in IFN-responsive tumor cells. Moreover, RRVs appear to carry a heat-labile component that actively suppresses activation of cellular innate immune responses in pDCs. Inhibition of IFN induction by RRVs and the reduced response to IFN should facilitate tumor-specific infection in vivo. IMPORTANCE RRVs have a convincing preference for replicating in tumor cells in animal models, and we observed similar preferences in the initial treatment of human glioblastoma patients. This study investigates the basis for the interaction between RRV and human host cells (tumor versus nontumor) in vitro. We found that RRVs do not trigger an IFN-α/β response in tumor cells, but the cells are capable of responding to type I IFNs and of producing them when stimulated with known agonists. Surprisingly, the data show that RRVs can actively inhibit induction of cellular innate immunity and that this inhibitory activity is heat labile and trypsin sensitive and not attributable to the envelope protein. These data partially explain the observed in vivo tumor specificity.
Collapse
|
30
|
Carbonaro Sarracino D, Tarantal AF, Lee CCI, Martinez M, Jin X, Wang X, Hardee CL, Geiger S, Kahl CA, Kohn DB. Effects of vector backbone and pseudotype on lentiviral vector-mediated gene transfer: studies in infant ADA-deficient mice and rhesus monkeys. Mol Ther 2014; 22:1803-16. [PMID: 24925206 DOI: 10.1038/mt.2014.88] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Accepted: 05/11/2014] [Indexed: 01/05/2023] Open
Abstract
Systemic delivery of a lentiviral vector carrying a therapeutic gene represents a new treatment for monogenic disease. Previously, we have shown that transfer of the adenosine deaminase (ADA) cDNA in vivo rescues the lethal phenotype and reconstitutes immune function in ADA-deficient mice. In order to translate this approach to ADA-deficient severe combined immune deficiency patients, neonatal ADA-deficient mice and newborn rhesus monkeys were treated with species-matched and mismatched vectors and pseudotypes. We compared gene delivery by the HIV-1-based vector to murine γ-retroviral vectors pseudotyped with vesicular stomatitis virus-glycoprotein or murine retroviral envelopes in ADA-deficient mice. The vesicular stomatitis virus-glycoprotein pseudotyped lentiviral vectors had the highest titer and resulted in the highest vector copy number in multiple tissues, particularly liver and lung. In monkeys, HIV-1 or simian immunodeficiency virus vectors resulted in similar biodistribution in most tissues including bone marrow, spleen, liver, and lung. Simian immunodeficiency virus pseudotyped with the gibbon ape leukemia virus envelope produced 10- to 30-fold lower titers than the vesicular stomatitis virus-glycoprotein pseudotype, but had a similar tissue biodistribution and similar copy number in blood cells. The relative copy numbers achieved in mice and monkeys were similar when adjusted to the administered dose per kg. These results suggest that this approach can be scaled-up to clinical levels for treatment of ADA-deficient severe combined immune deficiency subjects with suboptimal hematopoietic stem cell transplantation options.
Collapse
Affiliation(s)
- Denise Carbonaro Sarracino
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, California, USA
| | - Alice F Tarantal
- 1] Center for Fetal Monkey Gene Transfer for Heart, Lung, and Blood Diseases, University of California, Davis, California USA [2] Departments of Pediatrics and Cell Biology and Human Anatomy, University of California, Davis, CA, USA
| | - C Chang I Lee
- Center for Fetal Monkey Gene Transfer for Heart, Lung, and Blood Diseases, University of California, Davis, California USA
| | - Michele Martinez
- Center for Fetal Monkey Gene Transfer for Heart, Lung, and Blood Diseases, University of California, Davis, California USA
| | - Xiangyang Jin
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, California, USA
| | - Xiaoyan Wang
- Department of General Internal Medicine and Health Services Research, University of California, Los Angeles California, USA
| | - Cinnamon L Hardee
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, California, USA
| | - Sabine Geiger
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, California, USA
| | - Christoph A Kahl
- 1] Division of Research Immunology/BMT, Children's Hospital Los Angeles, Los Angeles, California, USA [2] Current address: Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Donald B Kohn
- 1] Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, California, USA [2] Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| |
Collapse
|
31
|
The miR-126-VEGFR2 axis controls the innate response to pathogen-associated nucleic acids. Nat Immunol 2013; 15:54-62. [PMID: 24270517 PMCID: PMC3896265 DOI: 10.1038/ni.2767] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 10/03/2013] [Indexed: 12/19/2022]
Abstract
MicroRNA-126 (miR-126) is a microRNA predominately expressed by endothelial cells and controls angiogenesis. We found miR-126 was required for the innate response to pathogen-associated nucleic acids, and that miR-126-deficient mice had increased susceptibility to pseudotyped-HIV infection. miRNA profiling and deep-sequencing indicated that miR-126 was highly and specifically expressed by plasmacytoid dendritic cells (pDCs). miR-126 controlled the survival and function of pDCs, and regulated expression ofinnate response genes, including Tlr7, Tlr9 and Nfkb1, as well as Kdr, which encodes VEGF-receptor 2 (VEGFR2). Deletion of Kdr in DCs resulted in reduced type I interferon production, supporting a role for VEGFR2 in miR-126 regulation of pDCs. These studies identify the miR-126–VEGFR2 axis as an important regulator of the innate response that operates through multiscale control of pDCs.
Collapse
|
32
|
Deng S, Zhou Y, Ouyang D, Xiong J, Zhang L, Tu C, Zhang K, Song Z, Zhang F. The effect of dexamethasone on lentiviral vector infection is associated with importin α. Biomed Rep 2013; 2:137-141. [PMID: 24649085 DOI: 10.3892/br.2013.194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 10/30/2013] [Indexed: 02/02/2023] Open
Abstract
Importin α (Imα) plays an important role during the shuttling of the HIV-1 preintegration complex (PIC) from the cytoplasm to the nucleus. Imα may bind to the glucocorticoid receptor (GR), which is localized to nucleus following hormone binding. However, it remains unclear whether the binding of dexamethasone (Dex) to GR affects the Imα redistribution and, thus, alters PIC import. In our study, 293T cells were transfected with the lentiviral vector (LV) carrying the luciferase (Luci) gene following Dex or RU486 pretreatment. The Luci activity (LucA) in the Dex or RU486 group was significantly higher compared to that in the control group (P≤0.01). The effects of Dex and RU486 were inhibited by the Imα inhibitor Bimax1 (P≤0.01), although the inhibitory effect of Bimax1 was alleviated by increasing the Dex dose. Furthermore, it was observed that the LucA in the 30-min Dex treatment group was lower compared to that in the 30-min Dex pretreatment group (P≤0.01). These results suggested that Dex may improve PIC import via increasing the cytoplasmic Imα levels. Kunming mice were transfected in vivo with the LV, either 30 min or 15 h following an intraperitoneal injection of Dex. The LucA in the liver of the 30-min group mice was significantly lower compared to that of the 15-h group mice (P≤0.01), suggesting that the effect of Dex on LV infection depends mainly on the suppression of immune and inflammatory responses in vivo. Taken together, our data indicated that the effect of Dex on LV infection may be associated with Imα, constituting a novel signaling pathway mediating the effects of Dex on HIV-1 infection.
Collapse
Affiliation(s)
- Shengchang Deng
- School of Medicine, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Ying Zhou
- School of Medicine, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Dong Ouyang
- Jiangxi Police College, Nanchang, Jiangxi 330103, P.R. China
| | - Junping Xiong
- School of Medicine, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Lei Zhang
- School of Medicine, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Changchun Tu
- School of Medicine, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Keping Zhang
- School of Medicine, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zengliang Song
- Department of Neurosurgery, The Third People's Hospital of Nanchang, Nanchang, Jiangxi 330009, P.R. China
| | - Fanglin Zhang
- School of Medicine, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
33
|
Schambach A, Zychlinski D, Ehrnstroem B, Baum C. Biosafety features of lentiviral vectors. Hum Gene Ther 2013; 24:132-42. [PMID: 23311447 DOI: 10.1089/hum.2012.229] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Over the past decades, lentiviral vectors have evolved as a benchmark tool for stable gene transfer into cells with a high replicative potential. Their relatively flexible genome and ability to transduce many forms of nondividing cells, combined with the potential for cell-specific pseudotyping, provides a rich resource for numerous applications in experimental platforms and therapeutic settings. Here, we give an overview of important biosafety features of lentiviral vectors, with detailed discussion of (i) the principles of the lentiviral split-genome design used for the construction of packaging cells; (ii) the relevance of modifications introduced into the lentiviral long terminal repeat (deletion of enhancer/promoter sequences and introduction of insulators); (iii) the basic features of mRNA processing, including the Rev/Rev-responsive element (RRE) interaction and the modifications of the 3' untranslated region of lentiviral vectors with various post-transcriptional regulatory elements affecting transcriptional termination, polyadenylation, and differentiation-specific degradation of mRNA; and (iv) the characteristic integration pattern with the associated risk of transcriptional interference with cellular genes. We conclude with considerations regarding the importance of cell targeting via envelope modifications. Along this course, we address canonical biosafety issues encountered with any type of viral vector: the risks of shedding, mobilization, germline transmission, immunogenicity, and insertional mutagenesis.
Collapse
Affiliation(s)
- Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, D-30625 Hannover, Germany
| | | | | | | |
Collapse
|
34
|
Shi Y, Falahati R, Zhang J, Flebbe-Rehwaldt L, Gaensler KML. Role of antigen-specific regulatory CD4+CD25+ T cells in tolerance induction after neonatal IP administration of AAV-hF.IX. Gene Ther 2013; 20:987-96. [PMID: 23759700 PMCID: PMC3795474 DOI: 10.1038/gt.2013.22] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Revised: 01/07/2013] [Accepted: 02/08/2013] [Indexed: 01/10/2023]
Abstract
Neonatal AAV8-mediated Factor IX (F.IX) gene delivery was applied as a model for exploring mechanisms of tolerance induction during immune ontogeny. Intraperitoneal delivery of AAV8/ Factor IX (hF.IX) during weeks 1–4 of life, over a 20-fold dose range, directed stable hF.IX expression, correction of coagulopathy in F.IX-null hemophilia B mice, and induction of tolerance to hF.IX; however, only primary injection at 1–2 days of life enabled increasing AAV8-mediated hF.IX expression after re-administration, due to the absence of anti-viral capsid antibodies. Adoptive splenocyte transfer from tolerized mice demonstrated induction of CD4+CD25+ T regulatory (Treg) populations that specifically suppressed anti-hF.IX antibody responses, but not responses to third party antigen. Induction of hF.IX antibodies was only observed in tolerized mice after in vivo CD4+CD25+ cell depletion and hF.IX challenge. Thus, primary injection of AAV during a critical period in the first week of life does not elicit antiviral responses, enabling re-administration of AAV and augmentation of hF.IX levels. Expansion of hF.IX-specific CD4+CD25+ Tregs has a major role in tolerance induction early in immune ontogeny. Neonatal gene transfer provides a useful approach for defining the ontogeny of immune responses and may suggest approaches for inducing tolerance in the context of genetic therapies.
Collapse
Affiliation(s)
- Y Shi
- Department of Medicine, University of California, San Francisco, CA, USA
| | | | | | | | | |
Collapse
|
35
|
Annoni A, Goudy K, Akbarpour M, Naldini L, Roncarolo MG. Immune responses in liver-directed lentiviral gene therapy. Transl Res 2013; 161:230-40. [PMID: 23360745 DOI: 10.1016/j.trsl.2012.12.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 12/13/2012] [Accepted: 12/17/2012] [Indexed: 01/13/2023]
Abstract
The use of lentiviral vectors (LV)s for in vivo gene therapy is an ideal platform for treating many types of disease. Since LVs can transduce a wide array of cells, support long-term gene expression, and be modified to enhance cell targeting, LVs are a powerful modality to deliver life-long therapeutic proteins. A major limitation facing the use of LVs for in vivo gene therapy is the induction of immune responses, which can reduce the transduction efficiency of LV, eliminate the transduced cells, and inhibit the effect of the therapeutic protein. LV strategies designed to restrict transgene expression to the liver to exploit its naturally tolerogenic properties have proven to significantly reduce the induction of pathogenic immune responses and increase therapeutic efficacy. In this review, we outline the immunological hurdles facing in vivo LV gene therapy and highlight the advantages and limitations of using liver-directed LV gene therapy.
Collapse
Affiliation(s)
- Andrea Annoni
- San Raffaele Telethon Institute for Gene Therapy (HSR-TIGET), Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
| | | | | | | | | |
Collapse
|