1
|
Hannani D, Leplus E, Laulagnier K, Chaperot L, Plumas J. Leveraging a powerful allogeneic dendritic cell line towards neoantigen-based cancer vaccines. Genes Cancer 2023; 14:3-11. [PMID: 36726965 PMCID: PMC9886307 DOI: 10.18632/genesandcancer.229] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 01/20/2023] [Indexed: 01/31/2023] Open
Abstract
In recent years, immunotherapy has finally found its place in the anti-cancer therapeutic arsenal, even becoming standard of care as first line treatment for metastatic forms. The clinical benefit provided by checkpoint blockers such as anti-PD-1/PD-L1 in many cancers revolutionized the field. However, too many patients remain refractory to these treatments due to weak baseline anti-cancer immunity. There is therefore a need to boost the frequency and function of patients' cytotoxic CD8+ cellular effectors by targeting immunogenic and tumor-restricted antigens, such as neoantigens using an efficient vaccination platform. Dendritic cells (DC) are the most powerful immune cell subset for triggering cellular immune response. However, autologous DC-based vaccines display several limitations, such as the lack of reproducibility and the limited number of cells that can be manufactured. Here we discuss the advantages of a new therapeutic vaccine based on an allogeneic Plasmacytoid DC cell line, which is easy to produce and represents a powerful platform for priming and expanding anti-neoantigen cytotoxic CD8+ T-cells.
Collapse
Affiliation(s)
| | | | | | - Laurence Chaperot
- 2R&D Laboratory, Etablissement Français du Sang Auvergne Rhône-Alpes (EFS AURA), Grenoble, France
| | - Joël Plumas
- 1PDC*line Pharma, Grenoble, France,2R&D Laboratory, Etablissement Français du Sang Auvergne Rhône-Alpes (EFS AURA), Grenoble, France,Correspondence to:Joël Plumas, email:
| |
Collapse
|
2
|
Engineering a Human Plasmacytoid Dendritic Cell-Based Vaccine to Prime and Expand Multispecific Viral and Tumor Antigen-Specific T-Cells. Vaccines (Basel) 2021; 9:vaccines9020141. [PMID: 33578850 PMCID: PMC7916617 DOI: 10.3390/vaccines9020141] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/03/2021] [Accepted: 02/08/2021] [Indexed: 11/17/2022] Open
Abstract
Because dendritic cells are crucial to prime and expand antigen-specific CD8+ T-cells, several strategies are designed to use them in therapeutic vaccines against infectious diseases or cancer. In this context, off-the-shelf allogeneic dendritic cell-based platforms are more attractive than individualized autologous vaccines tailored to each patient. In the present study, a unique dendritic cell line (PDC*line) platform of plasmacytoid origin, already used to prime and expand antitumor immunity in melanoma patients, was improved thanks to retroviral engineering. We demonstrated that the clinical-grade PDC*line, transduced with genes encoding viral or tumoral whole proteins, efficiently processed and stably presented the transduced antigens in different human leukocyte antigen (HLA) class I contexts. Moreover, the use of polyepitope constructs allowed the presentation of immunogenic peptides and the expansion of specific cytotoxic effectors. We also demonstrated that the addition of the Lysosome-associated membrane protein-1 (LAMP-1) sequence greatly improved the presentation of some peptides. Lastly, thanks to transduction of new HLA molecules, the PDC platform can benefit many patients through the easy addition of matched HLA-I molecules. The demonstration of the effective retroviral transduction of PDC*line cells strengthens and broadens the scope of the PDC*line platform, which can be used in adoptive or active immunotherapy for the treatment of infectious diseases or cancer.
Collapse
|
3
|
Hacker UT, Bentler M, Kaniowska D, Morgan M, Büning H. Towards Clinical Implementation of Adeno-Associated Virus (AAV) Vectors for Cancer Gene Therapy: Current Status and Future Perspectives. Cancers (Basel) 2020; 12:E1889. [PMID: 32674264 PMCID: PMC7409174 DOI: 10.3390/cancers12071889] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/03/2020] [Accepted: 07/07/2020] [Indexed: 02/06/2023] Open
Abstract
Adeno-associated virus (AAV) vectors have gained tremendous attention as in vivo delivery systems in gene therapy for inherited monogenetic diseases. First market approvals, excellent safety data, availability of large-scale production protocols, and the possibility to tailor the vector towards optimized and cell-type specific gene transfer offers to move from (ultra) rare to common diseases. Cancer, a major health burden for which novel therapeutic options are urgently needed, represents such a target. We here provide an up-to-date overview of the strategies which are currently developed for the use of AAV vectors in cancer gene therapy and discuss the perspectives for the future translation of these pre-clinical approaches into the clinic.
Collapse
Affiliation(s)
- Ulrich T. Hacker
- Department of Oncology, Gastroenterology, Hepatology, Pulmonology, and Infectious Diseases, University Cancer Center Leipzig (UCCL), Leipzig University Medical Center, 04103 Leipzig, Germany;
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; (M.B.); (M.M.)
| | - Martin Bentler
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; (M.B.); (M.M.)
| | - Dorota Kaniowska
- Department of Oncology, Gastroenterology, Hepatology, Pulmonology, and Infectious Diseases, University Cancer Center Leipzig (UCCL), Leipzig University Medical Center, 04103 Leipzig, Germany;
| | - Michael Morgan
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; (M.B.); (M.M.)
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Hildegard Büning
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; (M.B.); (M.M.)
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany
- German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Inhoffenstraße 7, 38124 Braunschweig, Germany
| |
Collapse
|
4
|
Gernoux G, Gruntman AM, Blackwood M, Zieger M, Flotte TR, Mueller C. Muscle-Directed Delivery of an AAV1 Vector Leads to Capsid-Specific T Cell Exhaustion in Nonhuman Primates and Humans. Mol Ther 2020; 28:747-757. [PMID: 31982038 PMCID: PMC7054721 DOI: 10.1016/j.ymthe.2020.01.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 01/08/2020] [Accepted: 01/08/2020] [Indexed: 12/21/2022] Open
Abstract
With the US Food and Drug Administration (FDA) and European Medicines Agency (EMA) approvals for Zolgensma, Luxturna, and Glybera, recombinant adeno-associated viruses (rAAVs) are considered efficient tools for gene transfer. However, studies in animals and humans demonstrate that intramuscular (IM) AAV delivery can trigger immune responses to AAV capsids and/or transgenes. IM delivery of rAAV1 in humans has also been described to induce tolerance to rAAV characterized by the presence of capsid-specific regulatory T cells (Tregs) in periphery. To understand mechanisms responsible for tolerance and parameters involved, we tested 3 muscle-directed administration routes in rhesus monkeys: IM delivery, venous limb perfusion, and the intra-arterial push and dwell method. These 3 methods were well tolerated and led to transgene expression. Interestingly, gene transfer in muscle led to Tregs and exhausted T cell infiltrates in situ at both day 21 and day 60 post-injection. In human samples, an in-depth analysis of the functionality of these cells demonstrates that capsid-specific exhausted T cells are detected after at least 5 years post-vector delivery and that the exhaustion can be reversed by blocking the checkpoint pathway. Overall, our study shows that persisting transgene expression after gene transfer in muscle is mediated by Tregs and exhausted T cells.
Collapse
Affiliation(s)
- Gwladys Gernoux
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA; Department of Pediatrics, University of Massachusetts Medical School, Worcester, MA, USA
| | - Alisha M Gruntman
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA; Department of Pediatrics, University of Massachusetts Medical School, Worcester, MA, USA; Department of Clinical Sciences, Cummings School of Veterinary Medicine at Tufts University, N. Grafton, MA, USA
| | - Meghan Blackwood
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - Marina Zieger
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - Terence R Flotte
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - Christian Mueller
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA; Department of Pediatrics, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
5
|
Domenger C, Grimm D. Next-generation AAV vectors—do not judge a virus (only) by its cover. Hum Mol Genet 2019; 28:R3-R14. [DOI: 10.1093/hmg/ddz148] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 05/30/2019] [Accepted: 06/17/2019] [Indexed: 12/11/2022] Open
Abstract
AbstractRecombinant adeno-associated viruses (AAV) are under intensive investigation in numerous clinical trials after they have emerged as a highly promising vector for human gene therapy. Best exemplifying their power and potential is the authorization of three gene therapy products based on wild-type AAV serotypes, comprising Glybera (AAV1), Luxturna (AAV2) and, most recently, Zolgensma (AAV9). Nonetheless, it has also become evident that the current AAV vector generation will require improvements in transduction potency, antibody evasion and cell/tissue specificity to allow the use of lower and safer vector doses. To this end, others and we devoted substantial previous research to the implementation and application of key technologies for engineering of next-generation viral capsids in a high-throughput ‘top-down’ or (semi-)rational ‘bottom-up’ approach. Here, we describe a set of recent complementary strategies to enhance features of AAV vectors that act on the level of the recombinant cargo. As examples that illustrate the innovative and synergistic concepts that have been reported lately, we highlight (i) novel synthetic enhancers/promoters that provide an unprecedented degree of AAV tissue specificity, (ii) pioneering genetic circuit designs that harness biological (microRNAs) or physical (light) triggers as regulators of AAV gene expression and (iii) new insights into the role of AAV DNA structures on vector genome stability, integrity and functionality. Combined with ongoing capsid engineering and selection efforts, these and other state-of-the-art innovations and investigations promise to accelerate the arrival of the next generation of AAV vectors and to solidify the unique role of this exciting virus in human gene therapy.
Collapse
Affiliation(s)
- Claire Domenger
- Department of Infectious Diseases/Virology, Heidelberg University Hospital, BioQuant Center, Im Neuenheimer Feld, Heidelberg, Germany
| | - Dirk Grimm
- Department of Infectious Diseases/Virology, Heidelberg University Hospital, BioQuant Center, Im Neuenheimer Feld, Heidelberg, Germany
- German Center for Infection Research (DZIF) and German Center for Cardiovascular Research (DZHK), Heidelberg, Germany
| |
Collapse
|
6
|
Laustsen A, Bak RO, Krapp C, Kjær L, Egedahl JH, Petersen CC, Pillai S, Tang HQ, Uldbjerg N, Porteus M, Roan NR, Nyegaard M, Denton PW, Jakobsen MR. Interferon priming is essential for human CD34+ cell-derived plasmacytoid dendritic cell maturation and function. Nat Commun 2018; 9:3525. [PMID: 30166549 PMCID: PMC6117296 DOI: 10.1038/s41467-018-05816-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 07/25/2018] [Indexed: 12/30/2022] Open
Abstract
Plasmacytoid dendritic cells (pDC) are essential for immune competence. Here we show that pDC precursor differentiated from human CD34+ hematopoietic stem and progenitor cells (HSPC) has low surface expression of pDC markers, and has limited induction of type I interferon (IFN) and IL-6 upon TLR7 and TLR9 agonists treatment; by contrast, cGAS or RIG-I agonists-mediated activation is not altered. Importantly, after priming with type I and II IFN, these precursor pDCs attain a phenotype and functional activity similar to that of peripheral blood-derived pDCs. Data from CRISPR/Cas9-mediated genome editing of HSPCs further show that HSPC-pDCs with genetic modifications can be obtained, and that expression of the IFN-α receptor is essential for the optimal function, but dispensable for the differentiation, of HSPC-pDC percursor. Our results thus demonstrate the biological effects of IFNs for regulating pDC function, and provide the means of generating of gene-modified human pDCs.
Collapse
Affiliation(s)
- A Laustsen
- Department of Biomedicine, Aarhus University, Wilhelm Meyers Alle 4, 8000, Aarhus C, Denmark
| | - R O Bak
- Department of Biomedicine, Aarhus University, Wilhelm Meyers Alle 4, 8000, Aarhus C, Denmark
- Aarhus Institute of Advanced Studies (AIAS), Aarhus University, Høegh-Guldbergs Gade 6B, 8000, Aarhus C, Denmark
- Department of Pediatrics, Stanford University, Stanford, CA, 94305, USA
| | - C Krapp
- Department of Biomedicine, Aarhus University, Wilhelm Meyers Alle 4, 8000, Aarhus C, Denmark
| | - L Kjær
- Department of Biomedicine, Aarhus University, Wilhelm Meyers Alle 4, 8000, Aarhus C, Denmark
| | - J H Egedahl
- Department of Biomedicine, Aarhus University, Wilhelm Meyers Alle 4, 8000, Aarhus C, Denmark
- Department of Urology, University of California, San Francisco, CA, 94158, USA
- The J. David Gladstone Institutes, San Francisco, CA, 94158, USA
| | - C C Petersen
- Department of Biomedicine, Aarhus University, Wilhelm Meyers Alle 4, 8000, Aarhus C, Denmark
| | - S Pillai
- University of California, San Francisco, Blood Systems Research Institute, 270 Masonic Avenue, San Francisco, 94118-4417, CA, USA
| | - H Q Tang
- Department of Obstetrics and Gynaecology, Aarhus University Hospital Skejby, Aarhus, 8200, Denmark
| | - N Uldbjerg
- Department of Obstetrics and Gynaecology, Aarhus University Hospital Skejby, Aarhus, 8200, Denmark
| | - M Porteus
- Department of Pediatrics, Stanford University, Stanford, CA, 94305, USA
| | - N R Roan
- Department of Urology, University of California, San Francisco, CA, 94158, USA
- The J. David Gladstone Institutes, San Francisco, CA, 94158, USA
| | - M Nyegaard
- Department of Biomedicine, Aarhus University, Wilhelm Meyers Alle 4, 8000, Aarhus C, Denmark
| | - P W Denton
- Department of Infectious Diseases, Aarhus University Hospital Skejby, Aarhus, 8200, Denmark
- Department of Clinical Medicine, Aarhus University Hospital Skejby, Aarhus, 8200, Denmark
| | - M R Jakobsen
- Department of Biomedicine, Aarhus University, Wilhelm Meyers Alle 4, 8000, Aarhus C, Denmark.
| |
Collapse
|
7
|
Gernoux G, Wilson JM, Mueller C. Regulatory and Exhausted T Cell Responses to AAV Capsid. Hum Gene Ther 2017; 28:338-349. [PMID: 28323492 PMCID: PMC5399736 DOI: 10.1089/hum.2017.022] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 03/16/2017] [Indexed: 12/31/2022] Open
Abstract
Recombinant adeno-associated viruses (AAVs) are quickly becoming the preferred viral vector for viral gene delivery for the treatment of a wide variety of genetic disorders. However, since their use in a clinical trial targeting hemophilia B patients 10 years ago, immune responses to the AAV capsid appear to have hampered some of the early clinical gene transfer efficacy. Indeed, AAV-based gene transfer has been shown to reactivate capsid-specific memory T cells, which have correlated with a decline in AAV-transduced tissue in some patients. Importantly, clinical trials have also shown that this reactivation can be quelled by administering time-course taper of glucocorticoid steroids before or after dosing. More recently, two clinical studies have shown that AAV gene transfer is not only able to induce a deleterious immune response, but also can result in the initiation of a tolerance to the AAV capsid mediated by regulatory T cells and exhausted T cells. This article reviews clinical trials describing immune responses to AAV, as well as the mechanisms responsible for immune tolerance in chronic infections and how it could apply to AAV-based gene transfer. A better understanding of both cytotoxic and tolerogenic immune responses to recombinant AAV will lead to safer gene transfer protocols in patients.
Collapse
Affiliation(s)
- Gwladys Gernoux
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts
| | - James M. Wilson
- Gene Therapy Program, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Christian Mueller
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts
| |
Collapse
|
8
|
Achard C, Guillerme JB, Bruni D, Boisgerault N, Combredet C, Tangy F, Jouvenet N, Grégoire M, Fonteneau JF. Oncolytic measles virus induces tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-mediated cytotoxicity by human myeloid and plasmacytoid dendritic cells. Oncoimmunology 2016; 6:e1261240. [PMID: 28197384 PMCID: PMC5283625 DOI: 10.1080/2162402x.2016.1261240] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 11/11/2016] [Accepted: 11/11/2016] [Indexed: 12/18/2022] Open
Abstract
Attenuated measles virus (MV) is currently being evaluated in clinical trials as an oncolytic therapeutic agent. Originally used for its lytic activity against tumor cells, it is now admitted that the effectiveness of MV also lies in its ability to initiate antitumor immune responses through the activation of dendritic cells (DCs). In this study, we investigated the capacity of oncolytic MV to convert human blood myeloid CD1c+ DCs and plasmacytoid DCs (pDCs) into cytotoxic effectors. We found that MV induces the expression of the cytotoxic protein TNF-related apoptosis-inducing ligand (TRAIL) on the surface of DCs. We demonstrate that the secretion of interferon-α (IFN-α) by DCs in response to MV is responsible for this TRAIL expression. Several types of PRRs (pattern recognition receptors) have been implicated in MV genome recognition, including RLRs (RIG-I-like receptors) and TLRs (Toll-like receptors). We showed that CD1c+ DCs secrete modest amounts of IFN-α and express TRAIL in an RLR-dependent manner upon exposure to MV. In pDCs, MV is recognized by RLRs and also by TLR7, leading to the secretion of high amounts of IFN-α and TRAIL expression. Finally, we showed that MV-stimulated DCs induce TRAIL-mediated cell death of Jurkat cells, confirming their acquisition of cytotoxic functions. Our results demonstrate that MV can activate cytotoxic myeloid CD1c+ DCs and pDCs, which may participate to the antitumor immune response.
Collapse
Affiliation(s)
- Carole Achard
- INSERM, UMR892, Institut de Recherche en Santé de l'Université de Nantes, Nantes, France; CNRS, UMR6299, Institut de Recherche en Santé de l'Université de Nantes, Nantes, France; Université de Nantes, Nantes, France
| | - Jean-Baptiste Guillerme
- INSERM, UMR892, Institut de Recherche en Santé de l'Université de Nantes, Nantes, France; CNRS, UMR6299, Institut de Recherche en Santé de l'Université de Nantes, Nantes, France; Université de Nantes, Nantes, France
| | - Daniela Bruni
- CNRS-UMR3569, Unité de Génomique Virale et Vaccination, Institut Pasteur , Paris, France
| | - Nicolas Boisgerault
- INSERM, UMR892, Institut de Recherche en Santé de l'Université de Nantes, Nantes, France; CNRS, UMR6299, Institut de Recherche en Santé de l'Université de Nantes, Nantes, France; Université de Nantes, Nantes, France
| | - Chantal Combredet
- CNRS-UMR3569, Unité de Génomique Virale et Vaccination, Institut Pasteur , Paris, France
| | - Frédéric Tangy
- CNRS-UMR3569, Unité de Génomique Virale et Vaccination, Institut Pasteur , Paris, France
| | - Nolwenn Jouvenet
- CNRS-UMR3569, Unité de Génomique Virale et Vaccination, Institut Pasteur , Paris, France
| | - Marc Grégoire
- INSERM, UMR892, Institut de Recherche en Santé de l'Université de Nantes, Nantes, France; CNRS, UMR6299, Institut de Recherche en Santé de l'Université de Nantes, Nantes, France; Université de Nantes, Nantes, France
| | - Jean-François Fonteneau
- INSERM, UMR892, Institut de Recherche en Santé de l'Université de Nantes, Nantes, France; CNRS, UMR6299, Institut de Recherche en Santé de l'Université de Nantes, Nantes, France; Université de Nantes, Nantes, France
| |
Collapse
|
9
|
Cheng M, Zhang X, Yu H, Du P, Plumas J, Chaperot L, Su L, Zhang L. Characterization of species-specific genes regulated by E2-2 in human plasmacytoid dendritic cells. Sci Rep 2015; 5:10752. [PMID: 26182859 PMCID: PMC4505321 DOI: 10.1038/srep10752] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 04/27/2015] [Indexed: 01/05/2023] Open
Abstract
Dendritic cells (DCs) are sentinels of the immune system and comprise two distinct subsets: conventional DCs (cDCs) and plasmacytoid DCs (pDCs). Human pDCs are distinguished from mouse pDCs phenotypically and functionally. Basic helix-loop-helix protein E2-2 is defined as an essential transcription factor for mouse pDC development, cell fate maintenance and gene programe. It is unknown whether E2-2 regulation contributes to this species-specific difference. Here we investigated the function of E2-2 in human pDCs and screened human-specific genes regulated by E2-2. Reduced E2-2 expression in human pDC cell line GEN2.2 resulted in diminished IFN-α production in response to CpG but elevated antigen presentation capacity. Gene expression profiling showed that E2-2 silence down-regulated pDC signature genes but up-regulated cDC signature genes. Thirty human-specific genes regulated by E2-2 knockdown were identified. Among these genes, we confirmed that expression of Siglec-6 was inhibited by E2-2. Further more, Siglec-6 was expressed at a higher level on a human pDC subset with drastically lower expression of E2-2. Collectively, these results highlight that E2-2 modulates pDC function in a species-specific manner, which may provide insights for pDC development and functions.
Collapse
Affiliation(s)
- Menglan Cheng
- Key Laboratory of Immunity and Infection, Institute of Biophysics, University of Chinese Academy of Sciences, Beijing, China
| | - Xuyuan Zhang
- Key Laboratory of Immunity and Infection, Institute of Biophysics, University of Chinese Academy of Sciences, Beijing, China
| | - Haisheng Yu
- Key Laboratory of Immunity and Infection, Institute of Biophysics, University of Chinese Academy of Sciences, Beijing, China
| | - Peishuang Du
- Key Laboratory of Immunity and Infection, Institute of Biophysics, University of Chinese Academy of Sciences, Beijing, China
| | - Joël Plumas
- Department of Research and Development, EFS Rh ône-Alpes Grenoble, La Tronche, France
| | - Laurance Chaperot
- Department of Research and Development, EFS Rh ône-Alpes Grenoble, La Tronche, France
| | - Lishan Su
- 1] Key Laboratory of Immunity and Infection, Institute of Biophysics, University of Chinese Academy of Sciences, Beijing, China [2] Lineberger Comprehensive Cancer Center, Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Liguo Zhang
- Key Laboratory of Immunity and Infection, Institute of Biophysics, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
10
|
Intrinsic transgene immunogenicity gears CD8(+) T-cell priming after rAAV-mediated muscle gene transfer. Mol Ther 2014; 23:697-706. [PMID: 25492560 DOI: 10.1038/mt.2014.235] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 12/02/2014] [Indexed: 01/18/2023] Open
Abstract
Antitransgene CD8(+) T-cell responses are an important hurdle after recombinant adeno-associated virus (rAAV) vector-mediated gene transfer. Indeed, depending on the mutational genotype of the host, transgene amino-acid sequences of foreign origin can elicit deleterious cellular and humoral responses. We compared here two different major histocompatibility complex (MHC) class I epitopes of an engineered ovalbumin transgene delivered in muscle tissue by rAAV1 vector and found very different strength of CD8 responses, muscle destruction being correlated with the course of the immunodominant response. We further demonstrate that robust CD8(+) T-cell priming can occur through the cross-presentation pathway but requires the presence of either a strong MHC class II epitope or antibodies to the transgene product. Finally, manipulating transgene subcellular localization, we found that provided we avoid transgene expression in antigen presenting cells, the poorly accessible cytosolic form of ovalbumin transgene lacking strong MHC II epitope, evades CD8(+) T-cell priming and remains permanently expressed in muscle with no immune cell infiltration. Our results demonstrate that the intrinsic immunogenicity of transgenes delivered with rAAV vector in muscle can be manipulated in a rational manner to avoid adverse immune responses.
Collapse
|
11
|
Boisgerault F, Gross DA, Ferrand M, Poupiot J, Darocha S, Richard I, Galy A. Prolonged gene expression in muscle is achieved without active immune tolerance using microrRNA 142.3p-regulated rAAV gene transfer. Hum Gene Ther 2014; 24:393-405. [PMID: 23427817 DOI: 10.1089/hum.2012.208] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Gene transfer efficacy is limited by unwanted immunization against transgene products. In some models, immunization may be avoided by regulating transgene expression with mir142.3p target sequences. Yet, it is unclear if such a strategy controls T-cell responses following recombinant adeno-associated viral vector (rAAV)-mediated gene transfer, particularly in muscle. In mice, intramuscular rAAV1 gene delivery of a tagged human sarcoglycan muscle protein is robustly immunogenic and leads to muscle destruction. In this model, the simple insertion of mir142.3p-target sequences in the transgene expression cassette modifies the outcome of gene transfer, providing high and persistent levels of muscle transduction in C57Bl/6 mice. Such regulated vector fails to prime specific CD4 and CD8 T cells; although, transgene tolerance seems to result from ignorance and could be broken by a robust antigenic challenge. While effective in normal mice, the mir142.3p-regulated transgene remains immunogenic in sarcoglycan-deficient dystrophic mice. In these mice, transgene expression is only prolonged but does not persist as effector CD4 and CD8 T-cell responses develop. Thus, using a mir142.3p-regulated transgene can improve rAAV muscle gene transfer results, but the level of efficacy depends on the context of application. In normal muscle, this strategy is sufficient to prevent immunization and functions even more effectively than tissue-specific promoters. In dystrophic models, additional strategies are required to fully control T-cell responses.
Collapse
Affiliation(s)
- Florence Boisgerault
- Genethon, Molecular Immunology and Innovative Biotherapies Group, Evry F91002 France
| | | | | | | | | | | | | |
Collapse
|
12
|
Chen JH, Yu YS, Chen XH, Liu HH, Zang GQ, Tang ZH. Enhancement of CTLs induced by DCs loaded with ubiquitinated hepatitis B virus core antigen. World J Gastroenterol 2012; 18:1319-27. [PMID: 22493545 PMCID: PMC3319958 DOI: 10.3748/wjg.v18.i12.1319] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Revised: 02/01/2012] [Accepted: 02/16/2012] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate whether hepatitis B virus (HBV) could induce a hepatitis B virus core antigen (HBcAg)-specific cytotoxic T lymphocyte (CTL) response in vitro by dendritic cells (DCs) transduced with lentiviral vector-encoding ubiquitinated hepatitis B virus core antigen (LV-Ub-HBcAg).
METHODS: Recombinant LV-Ub-HBcAg were transfected into highly susceptible 293 T cells to obtain high virus titres. Bone marrow-derived DCs isolated from BALB/c mice were cultured with recombinant granulocyte-macrophage colony-stimulating factor and recombinant interleukin (IL)-4. LV-Ub-HBcAg, lentiviral vector-encoding hepatitis B virus core antigen (LV-HBcAg), lentiviral vector (LV) or lipopolysaccharide were added to induce DC maturation, and the DC phenotypes were analyzed by flow cytometry. The level of IL-12 in the supernatant was detected by enzyme-linked immunosorbent assay (ELISA). T lymphocytes were proliferated using Cell Counting Kit-8. DCs were cultured and induced to mature using different LVs, and co-cultured with allogeneic T cells to detect the secretion levels of IL-2, IL-4, IL-10 and interferon-γ in the supernatants of T cells by ELISA. Intracellular cytokines of proliferative T cells were analyzed by flow cytometry, and specific CTL activity was measured by a lactate dehydrogenase release assay.
RESULTS: LV-Ub-HBcAg-induced DCs secreted more IL-12 and upregulated the expression of CD80, CD86 and major histocompatibility class II. DCs sensitised by different LVs effectively promoted cytokine secretion; the levels of IL-2 and interferon-γ induced by LV-Ub-HBcAg were higher than those induced by LV-HBcAg. Compared with LV-HBcAg-transduced DCs, LV-Ub-HBcAg-transduced DCs more efficiently stimulated the proliferation of T lymphocytes and generated HBcAg-specific cytotoxic T lymphocytes.
CONCLUSION: LV-Ub-HBcAg effectively induced DC maturation. The mature DCs efficiently induced T cell polarisation to Th1 and generated HBcAg-specific CTLs.
Collapse
|
13
|
Chung J, Rossi JJ, Jung U. Current progress and challenges in HIV gene therapy. Future Virol 2011; 6:1319-1328. [PMID: 22754586 DOI: 10.2217/fvl.11.113] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
HIV-1 causes AIDS, a syndrome that affects millions of people globally. Existing HAART is efficient in slowing down disease progression but cannot eradicate the virus. Furthermore the severity of the side effects and the emergence of drug-resistant mutants call for better therapy. Gene therapy serves as an attractive alternative as it reconstitutes the immune system with HIV-resistant cells and could thereby provide a potential cure. The feasibility of this approach was first demonstrated with the 'Berlin patient', who was functionally cured from HIV/AIDS with undetectable HIV-1 viral load after transplantation of bone marrow harboring a naturally occurring CCR5 mutation that blocks viral entry. Here, we give an overview of the current status of HIV gene therapy and remaining challenges and obstacles.
Collapse
Affiliation(s)
- Janet Chung
- Division of Molecular & Cell Biology, Beckman Research Institute of the City of Hope, 1500 East Duarte Road, CA 91010, USA
| | | | | |
Collapse
|
14
|
Wang H, Zhang L, Kung SKP. Emerging applications of lentiviral vectors in dendritic cell-based immunotherapy. Immunotherapy 2010; 2:685-95. [DOI: 10.2217/imt.10.44] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Dendritic cells are professional antigen-presenting cells that initiate, regulate and shape the induction of specific immune responses. The ability to use dendritic cells in the induction of antigen-specific tolerance, antigen-specific immunity or specific differentiation of T-helper subsets holds great promise in dendritic cell-based immunotherapy of various diseases such as cancer, viral infections, allergy, as well as autoimmunity. Replication-incompetent HIV-1-based lentiviral vector is now emerging as a promising delivery system to genetically modify dendritic cells through antigen recognition, costimulatory molecules and/or polarization signals for the manipulation of antigen-specific immunity in vivo. This article discusses some of the recent advances in the uses of lentiviral vectors in dendritic cell-based immunotherapy.
Collapse
Affiliation(s)
- Huiming Wang
- University of Manitoba, Department of Immunology, Room 417 Apotex Center, 750 McDermot Avenue, Winnipeg, Manitoba, R3E 0T5, Canada
| | - Liang Zhang
- University of Manitoba, Department of Immunology, Room 417 Apotex Center, 750 McDermot Avenue, Winnipeg, Manitoba, R3E 0T5, Canada
| | | |
Collapse
|