Revisão Acesso aberto Revisado por pares

Surface-Engineered Lentiviral Vectors for Selective Gene Transfer into Subtypes of Lymphocytes

2018; Cell Press; Volume: 12; Linguagem: Inglês

10.1016/j.omtm.2018.10.006

ISSN

2329-0501

Autores

Annika M. Frank, Christian J. Buchholz,

Tópico(s)

Viral Infectious Diseases and Gene Expression in Insects

Resumo

Lymphocytes have always been among the prime targets in gene therapy, even more so since chimeric antigen receptor (CAR) T cells have reached the clinic. However, other gene therapeutic approaches hold great promise as well. The first part of this review provides an overview of current strategies in lymphocyte gene therapy. The second part highlights the importance of precise gene delivery into B and T cells as well as distinct subtypes of lymphocytes. This can be achieved with lentiviral vectors (LVs) pseudotyped with engineered glycoproteins recognizing lymphocyte surface markers as entry receptors. Different strategies for envelope glycoprotein engineering and selection of the targeting ligand are discussed. With a CD8-targeted LV that was recently used to achieve proof of principle for the in vivo reprogramming of CAR T cells, these vectors are becoming a key tool to genetically engineer lymphocytes directly in vivo. Lymphocytes have always been among the prime targets in gene therapy, even more so since chimeric antigen receptor (CAR) T cells have reached the clinic. However, other gene therapeutic approaches hold great promise as well. The first part of this review provides an overview of current strategies in lymphocyte gene therapy. The second part highlights the importance of precise gene delivery into B and T cells as well as distinct subtypes of lymphocytes. This can be achieved with lentiviral vectors (LVs) pseudotyped with engineered glycoproteins recognizing lymphocyte surface markers as entry receptors. Different strategies for envelope glycoprotein engineering and selection of the targeting ligand are discussed. With a CD8-targeted LV that was recently used to achieve proof of principle for the in vivo reprogramming of CAR T cells, these vectors are becoming a key tool to genetically engineer lymphocytes directly in vivo. Gene therapy looks back to a history of around 30 years. Since its early days, cells of the hematopoietic system, including lymphocytes, have been among the prime targets of research and clinical applications. In fact, the first clinical trial was performed in adenosine deaminase (ADA) deficiency-mediated severe combined immunodeficiency (ADA-SCID) patients by transferring an intact ADA gene copy into the patients' T lymphocytes by an ex vivo gene delivery approach using γ-retroviral vectors.1Ferrua F. Aiuti A. Twenty-Five Years of Gene Therapy for ADA-SCID: From Bubble Babies to an Approved Drug.Hum. Gene Ther. 2017; 28: 972-981Crossref PubMed Scopus (0) Google Scholar Although cure of this and other inherited immunodeficiencies in the end turned out to require gene delivery into hematopoietic stem cells (HSCs), B and T lymphocytes have remained in the focus. With two chimeric antigen receptor (CAR) T cell products having achieved marketing authorization, genetically modified T cells are major contributors to the success story of cancer immunotherapy.2June C.H. O'Connor R.S. Kawalekar O.U. Ghassemi S. Milone M.C. CAR T cell immunotherapy for human cancer.Science. 2018; 359: 1361-1365Crossref PubMed Scopus (0) Google Scholar However, many other promising approaches for engineering of both, T and B cells, have been developed to date, which are summarized below as well. Equipping T cells with recombinant receptors recognizing antigens on diseased cells, be it CARs or T cell receptors (TCRs), represents one of the most innovative and successful strategies of T cell engineering for therapeutic purposes to date. Recombinant TCRs have been most extensively studied in the context of cancer, with the first TCR specific for the tumor-associated antigen MART-1 (melanoma-associated antigen recognized by T cells) applied clinically already in 2006.3Morgan R.A. Dudley M.E. Wunderlich J.R. Hughes M.S. Yang J.C. Sherry R.M. Royal R.E. Topalian S.L. Kammula U.S. Restifo N.P. et al.Cancer regression in patients after transfer of genetically engineered lymphocytes.Science. 2006; 314: 126-129Crossref PubMed Scopus (1677) Google Scholar Today, many clinical trials have shown that TCR therapy can be beneficial for patients, with promising results obtained for melanoma, synovial cell sarcoma, and myeloma.4Robbins P.F. Kassim S.H. Tran T.L.N. Crystal J.S. Morgan R.A. Feldman S.A. Yang J.C. Dudley M.E. Wunderlich J.R. Sherry R.M. et al.A pilot trial using lymphocytes genetically engineered with an NY-ESO-1-reactive T-cell receptor: long-term follow-up and correlates with response.Clin. Cancer Res. 2015; 21: 1019-1027Crossref PubMed Scopus (211) Google Scholar, 5Rapoport A.P. Stadtmauer E.A. Binder-Scholl G.K. Goloubeva O. Vogl D.T. Lacey S.F. Badros A.Z. Garfall A. Weiss B. Finklestein J. et al.NY-ESO-1-specific TCR-engineered T cells mediate sustained antigen-specific antitumor effects in myeloma.Nat. Med. 2015; 21: 914-921Crossref PubMed Scopus (250) Google Scholar, 6Morgan R.A. Chinnasamy N. Abate-Daga D. Gros A. Robbins P.F. Zheng Z. Dudley M.E. Feldman S.A. Yang J.C. Sherry R.M. et al.Cancer regression and neurological toxicity following anti-MAGE-A3 TCR gene therapy.J. Immunother. 2013; 36: 133-151Crossref PubMed Scopus (451) Google Scholar Consequently, new studies are on the way involving both well-characterized and new lead TCRs with novel specificities for the treatment of various cancer types.7Karpanen T. Olweus J. T-cell receptor gene therapy--ready to go viral?.Mol. Oncol. 2015; 9: 2019-2042Crossref PubMed Scopus (0) Google Scholar, 8Saudemont A. Jespers L. Clay T. Current Status of Gene Engineering Cell Therapeutics.Front. Immunol. 2018; 9: 153Crossref PubMed Scopus (6) Google Scholar In comparison to response rates achieved with TCR T cell therapy, CAR T cells have been even more successful, as illustrated by the recent approval of the CD19-CAR T cell products Kymriah and Yescarta by the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA).9Maude S.L. Laetsch T.W. Buechner J. Rives S. Boyer M. Bittencourt H. Bader P. Verneris M.R. Stefanski H.E. Myers G.D. et al.Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia.N. Engl. J. Med. 2018; 378: 439-448Crossref PubMed Scopus (248) Google Scholar, 10Neelapu S.S. Locke F.L. Bartlett N.L. Lekakis L.J. Miklos D.B. Jacobson C.A. Braunschweig I. Oluwole O.O. Siddiqi T. Lin Y. et al.Axicabtagene Ciloleucel CAR T-Cell Therapy in Refractory Large B-Cell Lymphoma.N. Engl. J. Med. 2017; 377: 2531-2544Crossref PubMed Scopus (263) Google Scholar Efforts are being put into improving the CAR technology to increase safety and efficacy, reduce production costs, and make it applicable beyond hematological malignancies. Consequently, the number of clinical trials continues to increase exponentially.11Hartmann J. Schüßler-Lenz M. Bondanza A. Buchholz C.J. Clinical development of CAR T cells-challenges and opportunities in translating innovative treatment concepts.EMBO Mol. Med. 2017; 9: 1183-1197Crossref PubMed Scopus (0) Google Scholar In addition, TCR and CAR therapies are now being expanded beyond cancer treatment. Regulatory T cells (Treg cells) representing the immunosuppressive arm of the T cell response have also been modified with CARs. For instance, an approach for the treatment of alloreactivity after organ transplantation used CAR Treg cells recognizing the human leukocyte antigen (HLA) A2.12MacDonald K.G. Hoeppli R.E. Huang Q. Gillies J. Luciani D.S. Orban P.C. Broady R. Levings M.K. Alloantigen-specific regulatory T cells generated with a chimeric antigen receptor.J. Clin. Invest. 2016; 126: 1413-1424Crossref PubMed Scopus (78) Google Scholar In autoimmunity, Tregs are equipped with CARs specific for self-antigens,13Skuljec J. Chmielewski M. Happle C. Habener A. Busse M. Abken H. Hansen G. Chimeric Antigen Receptor-Redirected Regulatory T Cells Suppress Experimental Allergic Airway Inflammation, a Model of Asthma.Front. Immunol. 2017; 8: 1125Crossref PubMed Scopus (5) Google Scholar, 14Yoon J. Schmidt A. Zhang A.-H. Königs C. Kim Y.C. Scott D.W. FVIII-specific human chimeric antigen receptor T-regulatory cells suppress T- and B-cell responses to FVIII.Blood. 2017; 129: 238-245Crossref PubMed Google Scholar in some studies in combination with engineered expression of FOXP3.15Fransson M. Piras E. Burman J. Nilsson B. Essand M. Lu B. Harris R.A. Magnusson P.U. Brittebo E. Loskog A.S. CAR/FoxP3-engineered T regulatory cells target the CNS and suppress EAE upon intranasal delivery.J. Neuroinflammation. 2012; 9: 112Crossref PubMed Scopus (72) Google Scholar For treatment of antibody-mediated autoimmune disease, the autoantigen has been presented as an extracellular domain of the chimeric receptor, resulting in T cells redirected to anti-autoantigen B cell receptors that eliminated autoreactive B cells.16Ellebrecht C.T. Bhoj V.G. Nace A. Choi E.J. Mao X. Cho M.J. Di Zenzo G. Lanzavecchia A. Seykora J.T. Cotsarelis G. et al.Reengineering chimeric antigen receptor T cells for targeted therapy of autoimmune disease.Science. 2016; 353: 179-184Crossref PubMed Google Scholar Expanding this approach, it was recently shown that Treg cells expressing such an inverse CAR inhibit autoreactive B cells in a mouse model of hemophilia A.17Zhang A.-H. Yoon J. Kim Y.C. Scott D.W. Targeting Antigen-Specific B Cells Using Antigen-Expressing Transduced Regulatory T Cells.J. Immunol. 2018; 201: 1434-1441Crossref PubMed Scopus (0) Google Scholar To increase T cell responses, cytokine or chemokine receptors as well as costimulatory receptors can be introduced. For instance, expression of CX3CR1 in T cells has improved T cell trafficking in preclinical tumor models.18Siddiqui I. Erreni M. van Brakel M. Debets R. Allavena P. Enhanced recruitment of genetically modified CX3CR1-positive human T cells into Fractalkine/CX3CL1 expressing tumors: importance of the chemokine gradient.J. Immunother. Cancer. 2016; 4: 21Crossref PubMed Scopus (11) Google Scholar Likewise, genetic knockout of inhibitory receptors such as programmed death-1 (PD1) can improve T cell functions.19Zhao Z. Shi L. Zhang W. Han J. Zhang S. Fu Z. Cai J. CRISPR knock out of programmed cell death protein 1 enhances anti-tumor activity of cytotoxic T lymphocytes.Oncotarget. 2017; 9: 5208-5215Crossref PubMed Scopus (14) Google Scholar Moreover, hybrid receptors have been introduced that combine the extracellular domain of an inhibitory receptor with the intracellular part of an activating receptor, thus converting the inhibitory signal into an activating one. Examples include a PD1 extracellular domain fused to a CD28 intracellular domain20Liu X. Ranganathan R. Jiang S. Fang C. Sun J. Kim S. Newick K. Lo A. June C.H. Zhao Y. Moon E.K. A Chimeric Switch-Receptor Targeting PD1 Augments the Efficacy of Second-Generation CAR T Cells in Advanced Solid Tumors.Cancer Res. 2016; 76: 1578-1590Crossref PubMed Scopus (100) Google Scholar and an interleukin (IL)-4 receptor displaying the IL-7 receptor signaling domain.21Leen A.M. Sukumaran S. Watanabe N. Mohammed S. Keirnan J. Yanagisawa R. Anurathapan U. Rendon D. Heslop H.E. Rooney C.M. et al.Reversal of tumor immune inhibition using a chimeric cytokine receptor.Mol. Ther. 2014; 22: 1211-1220Abstract Full Text Full Text PDF PubMed Scopus (49) Google Scholar, 22Mohammed S. Sukumaran S. Bajgain P. Watanabe N. Heslop H.E. Rooney C.M. Brenner M.K. Fisher W.E. Leen A.M. Vera J.F. Improving Chimeric Antigen Receptor-Modified T Cell Function by Reversing the Immunosuppressive Tumor Microenvironment of Pancreatic Cancer.Mol. Ther. 2017; 25: 249-258Abstract Full Text Full Text PDF PubMed Scopus (23) Google Scholar Both approaches have resulted in superior antitumoral activities against large solid tumors established in mouse models over conventional CAR T cells used as controls. Besides, cytokine secretion can be engineered into CAR T cells in order to alter the immune environment within the tumor. Inducible release of IL-18 was recently shown to be superior over IL-12 with respect to safety and efficacy in a mouse model of melanoma.23Kunert A. Chmielewski M. Wijers R. Berrevoets C. Abken H. Debets R. Intra-tumoral production of IL18, but not IL12, by TCR-engineered T cells is non-toxic and counteracts immune evasion of solid tumors.OncoImmunology. 2017; 7: e1378842Crossref PubMed Scopus (2) Google Scholar Finally, a lot of effort is currently put into gene therapy concepts targeting HIV-infected cells.24Peterson C.W. Kiem H.-P. Cell and Gene Therapy for HIV Cure.Curr. Top. Microbiol. Immunol. 2018; 417: 211-248PubMed Google Scholar, 25Rogers G.L. Cannon P.M. Gene Therapy Approaches to Human Immunodeficiency Virus and Other Infectious Diseases.Hematol. Oncol. Clin. North Am. 2017; 31: 883-895Abstract Full Text Full Text PDF PubMed Scopus (1) Google Scholar Here, T cells have been modified to express antisense transcripts in an attempt to confer protection from HIV infection.26Levine B.L. Humeau L.M. Boyer J. MacGregor R.-R. Rebello T. Lu X. Binder G.K. Slepushkin V. Lemiale F. Mascola J.R. et al.Gene transfer in humans using a conditionally replicating lentiviral vector.Proc. Natl. Acad. Sci. USA. 2006; 103: 17372-17377Crossref PubMed Scopus (368) Google Scholar Similarly, the modification of T cells with membrane-anchored peptides inhibiting cell-virus fusion as well as TCRs recognizing viral antigens has been explored as a treatment option for HIV patients resistant to antiretroviral therapy.27Egelhofer M. Brandenburg G. Martinius H. Schult-Dietrich P. Melikyan G. Kunert R. Baum C. Choi I. Alexandrov A. von Laer D. Inhibition of human immunodeficiency virus type 1 entry in cells expressing gp41-derived peptides.J. Virol. 2004; 78: 568-575Crossref PubMed Scopus (121) Google Scholar, 28Perica K. Varela J.C. Oelke M. Schneck J. Adoptive T cell immunotherapy for cancer.Rambam Maimonides Med. J. 2015; 6: e0004Crossref PubMed Google Scholar Chronically or even latently infected cells are within special focus of ongoing anti-HIV strategies. Here, genome-modifying enzymes, such as transcription activator-like effector nucleases (TALENs) or CRISPR/Cas that precisely cleave the HIV provirus out of the cellular genome, as well as RNAi attacking viral RNA transcripts have appeared as promising novel tools.29Herrera-Carrillo E. Berkhout B. Attacking HIV-1 RNA versus DNA by sequence-specific approaches: RNAi versus CRISPR-Cas.Biochem. Soc. Trans. 2016; 44: 1355-1365Crossref PubMed Scopus (7) Google Scholar However, efficient delivery into resting T lymphocytes in vivo will be crucial for each of these strategies. As main players of the humoral immune response, B lymphocytes have been in the focus of genetic modification strategies as well. Best known for their ability to produce antigen-specific immunoglobulins, they can also act as antigen-presenting cells (APCs) to induce specific immune activation or immune tolerance. Given these essential functions, the genetic modification of B cells is of great interest for both, basic research and therapeutic applications. For instance, B lymphocytes were modified to express costimulatory molecules or pro-inflammatory cytokines for the improvement of their antigen-presenting function. Lee and colleagues30Lee J. Dollins C.M. Boczkowski D. Sullenger B.A. Nair S. Activated B cells modified by electroporation of multiple mRNAs encoding immune stimulatory molecules are comparable to mature dendritic cells in inducing in vitro antigen-specific T-cell responses.Immunology. 2008; 125: 229-240Crossref PubMed Scopus (0) Google Scholar could demonstrate enhanced antigen presentation by B cells that co-expressed the costimulatory ligands OX40L and 4-1BBL and the pro-inflammatory cytokine IL-12p40, which led to the induction of an antigen-specific T cell response in vitro. A similar approach was used to improve immune recognition of B cell malignancies. Introduction of CD80 and granulocyte-macrophage colony-stimulating factor (GM-CSF) into leukemic B cells ex vivo induced an anti-leukemic immune response.31Stripecke R. Cardoso A.A. Pepper K.A. Skelton D.C. Yu X.J. Mascarenhas L. Weinberg K.I. Nadler L.M. Kohn D.B. Lentiviral vectors for efficient delivery of CD80 and granulocyte-macrophage- colony-stimulating factor in human acute lymphoblastic leukemia and acute myeloid leukemia cells to induce antileukemic immune responses.Blood. 2000; 96: 1317-1326Crossref PubMed Google Scholar In autoimmune disease, engineering of antigen presentation by B cells can be used to induce immune tolerance, e.g., by delivering genes encoding antigen-immunoglobulin G (IgG) fusion proteins.32Melo M.E.F. Qian J. El-Amine M. Agarwal R.K. Soukhareva N. Kang Y. Scott D.W. Gene transfer of Ig-fusion proteins into B cells prevents and treats autoimmune diseases.J. Immunol. 2002; 168: 4788-4795Crossref PubMed Google Scholar, 33Wang X. Moghimi B. Zolotukhin I. Morel L.M. Cao O. Herzog R.W. Immune tolerance induction to factor IX through B cell gene transfer: TLR9 signaling delineates between tolerogenic and immunogenic B cells.Mol. Ther. 2014; 22: 1139-1150Abstract Full Text Full Text PDF PubMed Scopus (18) Google Scholar B cells and plasma cells in particular constitute nature's protein-producing factories able to secrete large amounts of antibodies. This unique ability can be hijacked by modifying the cells to secrete a monoclonal antibody or another therapeutic protein of choice. Taking advantage of this strategy, B cells were engineered to secrete neutralizing antibodies or antibody derivatives for the treatment of HIV34Luo X.M. Maarschalk E. O'Connell R.M. Wang P. Yang L. Baltimore D. Engineering human hematopoietic stem/progenitor cells to produce a broadly neutralizing anti-HIV antibody after in vitro maturation to human B lymphocytes.Blood. 2009; 113: 1422-1431Crossref PubMed Scopus (58) Google Scholar, 35Hur E.M. Patel S.N. Shimizu S. Rao D.S. Gnanapragasam P.N.P. An D.S. Yang L. Baltimore D. Inhibitory effect of HIV-specific neutralizing IgA on mucosal transmission of HIV in humanized mice.Blood. 2012; 120: 4571-4582Crossref PubMed Scopus (0) Google Scholar, 36Joseph A. Zheng J.H. Chen K. Dutta M. Chen C. Stiegler G. Kunert R. Follenzi A. Goldstein H. Inhibition of in vivo HIV infection in humanized mice by gene therapy of human hematopoietic stem cells with a lentiviral vector encoding a broadly neutralizing anti-HIV antibody.J. Virol. 2010; 84: 6645-6653Crossref PubMed Scopus (57) Google Scholar and hepatitis C virus (HCV).37Fusil F. Calattini S. Amirache F. Mancip J. Costa C. Robbins J.B. Douam F. Lavillette D. Law M. Defrance T. et al.A Lentiviral Vector Allowing Physiologically Regulated Membrane-anchored and Secreted Antibody Expression Depending on B-cell Maturation Status.Mol. Ther. 2015; 23: 1734-1747Abstract Full Text Full Text PDF PubMed Scopus (0) Google Scholar A very interesting recent study by Hung and colleagues38Hung K.L. Meitlis I. Hale M. Chen C.-Y. Singh S. Jackson S.W. Miao C.H. Khan I.F. Rawlings D.J. James R.G. Engineering Protein-Secreting Plasma Cells by Homology-Directed Repair in Primary Human B Cells.Mol. Ther. 2018; 26: 456-467Abstract Full Text Full Text PDF PubMed Scopus (0) Google Scholar achieved expression of human factor IX (FIX) and B cell-activating factor in human plasma cells by gene editing. Engineered human primary B cells secreting FIX have also been generated by lentiviral transduction.39Levy C. Fusil F. Amirache F. Costa C. Girard-Gagnepain A. Negre D. Bernadin O. Garaulet G. Rodriguez A. Nair N. et al.Baboon envelope pseudotyped lentiviral vectors efficiently transduce human B cells and allow active factor IX B cell secretion in vivo in NOD/SCIDγc-/- mice.J. Thromb. Haemost. 2016; 14: 2478-2492Crossref PubMed Scopus (8) Google Scholar Transplantation of the modified cells into immunodeficient mice resulted in high-level expression of the therapeutic proteins in both studies, thus providing proof of concept for these novel therapeutic options to treat hemophilia. Current approaches for lymphocyte engineering mainly rely on ex vivo gene transfer protocols. Following their isolation from either healthy donors or patients, the cells are activated and subsequently transduced by lentiviral vectors (LVs), the majority of which is pseudotyped with the glycoprotein G of vesicular stomatitis virus (VSV). The modified lymphocytes are then expanded and either used in functional in vitro assays or used for in vivo applications. While ex vivo modification of B lymphocytes is possible and has been successfully applied as outlined above, it is not an easy task. Stable and efficient transduction of B cells by LVs is very difficult to achieve. Quiescent B cells are restrictive to transduction by conventional VSV G-pseudotyped LVs (VSV-LVs) due to a lack of low-density lipoprotein receptor (LDLR) expression,40Amirache F. Lévy C. Costa C. Mangeot P.-E. Torbett B.E. Wang C.X. Nègre D. Cosset F.L. Verhoeyen E. Mystery solved: VSV-G-LVs do not allow efficient gene transfer into unstimulated T cells, B cells, and HSCs because they lack the LDL receptor.Blood. 2014; 123: 1422-1424Crossref PubMed Scopus (47) Google Scholar and, thus, they have to be activated prior to transduction. Efficient activation and culture of primary human B lymphocytes is tedious, as it involves carefully titrated activating stimuli in combination with cytokines followed by co-cultivation with feeder cells.41Frecha C. Lévy C. Cosset F.-L. Verhoeyen E. Advances in the field of lentivector-based transduction of T and B lymphocytes for gene therapy.Mol. Ther. 2010; 18: 1748-1757Abstract Full Text Full Text PDF PubMed Scopus (41) Google Scholar Even under optimal activation and culture conditions, transduction efficiencies with VSV-LVs are notoriously low. The combination of all these difficulties may well serve as an explanation for the much lower number of clinical trials involving engineered B cells as compared to T cells. In comparison, T lymphocyte manipulation by lentiviral transduction is easier to achieve. Still, the cells have to be activated prior to transduction with conventional VSV-LV, because, like B cells, they are otherwise not susceptible for transduction, again due to a lack of LDLR expression.40Amirache F. Lévy C. Costa C. Mangeot P.-E. Torbett B.E. Wang C.X. Nègre D. Cosset F.L. Verhoeyen E. Mystery solved: VSV-G-LVs do not allow efficient gene transfer into unstimulated T cells, B cells, and HSCs because they lack the LDL receptor.Blood. 2014; 123: 1422-1424Crossref PubMed Scopus (47) Google Scholar Mainly due to the paramount success of CAR T cell therapy, the protocols for T cell isolation, activation, lentiviral transduction, and expansion have been extensively improved in recent years. Current state-of-the-art T cell activation relies on stimulation of the TCR activation pathway via CD3- and CD28-specific antibodies in combination with cytokines such as IL-7 and IL-15.42Xu Y. Zhang M. Ramos C.A. Durett A. Liu E. Dakhova O. Liu H. Creighton C.J. Gee A.P. Heslop H.E. et al.Closely related T-memory stem cells correlate with in vivo expansion of CAR.CD19-T cells and are preserved by IL-7 and IL-15.Blood. 2014; 123: 3750-3759Crossref PubMed Scopus (139) Google Scholar, 43Casucci M. Falcone L. Camisa B. Norelli M. Porcellini S. Stornaiuolo A. Ciceri F. Traversari C. Bordignon C. Bonini C. Bondanza A. Extracellular NGFR Spacers Allow Efficient Tracking and Enrichment of Fully Functional CAR-T Cells Co-Expressing a Suicide Gene.Front. Immunol. 2018; 9: 507Crossref PubMed Scopus (1) Google Scholar The need for activation of lymphocytes prior to transduction with conventional LVs harbors some disadvantages. First, it adds to the complexity of the overall procedure, involving additional steps before transduction of the cells can be carried out. This increases duration and costs of the manufacturing process. Second, the stimuli applied for activation in combination with the prolonged ex vivo culture likely change the cells, which can negatively impact on the quality of the final product. As a result, naive cells could differentiate into less preferential phenotypes that exhibit a higher degree of exhaustion, lower proliferative capacity, shorter in vivo persistence, and less functionality. This can have very important implications for therapeutic success. For instance, it has been shown that a central memory (CD45RO+/CD45RA+/CD62L+) or stem cell memory (CD45RO+/CD45RA−/CD62L+)44Golubovskaya V. Wu L. Different Subsets of T Cells, Memory, Effector Functions, and CAR-T Immunotherapy.Cancers (Basel). 2016; 8: 36Crossref PubMed Scopus (0) Google Scholar phenotype is beneficial for T cell persistence and function in vivo.45Berger C. Jensen M.C. Lansdorp P.M. Gough M. Elliott C. Riddell S.R. Adoptive transfer of effector CD8+ T cells derived from central memory cells establishes persistent T cell memory in primates.J. Clin. Invest. 2008; 118: 294-305Crossref PubMed Scopus (503) Google Scholar In this regard, a positive correlation of a CAR T cell central memory phenotype and a positive clinical response has been observed in several clinical studies,42Xu Y. Zhang M. Ramos C.A. Durett A. Liu E. Dakhova O. Liu H. Creighton C.J. Gee A.P. Heslop H.E. et al.Closely related T-memory stem cells correlate with in vivo expansion of CAR.CD19-T cells and are preserved by IL-7 and IL-15.Blood. 2014; 123: 3750-3759Crossref PubMed Scopus (139) Google Scholar, 46Louis C.U. Savoldo B. Dotti G. Pule M. Yvon E. Myers G.D. Rossig C. Russell H.V. Diouf O. Liu E. et al.Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma.Blood. 2011; 118: 6050-6056Crossref PubMed Scopus (450) Google Scholar, 47Fraietta J.A. Lacey S.F. Orlando E.J. Pruteanu-Malinici I. Gohil M. Lundh S. Boesteanu A.C. Wang Y. O'Connor R.S. Hwang W.T. et al.Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia.Nat. Med. 2018; 24: 563-571Crossref PubMed Scopus (0) Google Scholar and, consequently, the infusion of purified central memory CAR T cells is now being investigated as well.48Wang X. Popplewell L.L. Wagner J.R. Naranjo A. Blanchard M.S. Mott M.R. Norris A.P. Wong C.W. Urak R.Z. Chang W.C. et al.Phase 1 studies of central memory-derived CD19 CAR T-cell therapy following autologous HSCT in patients with B-cell NHL.Blood. 2016; 127: 2980-2990Crossref PubMed Scopus (69) Google Scholar Likewise, a central memory phenotype leads to functionally superior TCR-modified T cells.49Wu F. Zhang W. Shao H. Bo H. Shen H. Li J. Liu Y. Wang T. Ma W. Huang S. Human effector T cells derived from central memory cells rather than CD8(+)T cells modified by tumor-specific TCR gene transfer possess superior traits for adoptive immunotherapy.Cancer Lett. 2013; 339: 195-207Abstract Full Text Full Text PDF PubMed Scopus (0) Google Scholar Minimal manipulation of lymphocytes during genetic modification is thus of tremendous clinical relevance. Substantial progress toward transducing resting lymphocytes has been achieved by replacing the VSV G by envelopes of viruses that use a receptor expressed on quiescent cells for cell entry. Examples include the glycoproteins of measles virus (MV)50Frecha C. Costa C. Nègre D. Gauthier E. Russell S.J. Cosset F.-L. Verhoeyen E. Stable transduction of quiescent T cells without induction of cycle progression by a novel lentiviral vector pseudotyped with measles virus glycoproteins.Blood. 2008; 112: 4843-4852Crossref PubMed Scopus (0) Google Scholar, 51Frecha C. Lévy C. Costa C. Nègre D. Amirache F. Buckland R. Russell S.J. Cosset F.L. Verhoeyen E. Measles virus glycoprotein-pseudotyped lentiviral vector-mediated gene transfer into quiescent lymphocytes requires binding to both SLAM and CD46 entry receptors.J. Virol. 2011; 85: 5975-5985Crossref PubMed Scopus (45) Google Scholar, 52Zhou Q. Schneider I.C. Gallet M. Kneissl S. Buchholz C.J. Resting lymphocyte transduction with measles virus glycoprotein pseudotyped lentiviral vectors relies on CD46 and SLAM.Virology. 2011; 413: 149-152Crossref PubMed Scopus (13) Google Scholar or the envelope protein of Baboon endogenous retrovirus (BaEV),39Levy C. Fusil F. Amirache F. Costa C. Girard-Gagnepain A. Negre D. Bernadin O. Garaulet G. Rodriguez A. Nair N. et al.Baboon envelope pseudotyped lentiviral vectors efficiently transduce human B cells and allow active factor IX B cell secretion in vivo in NOD/SCIDγc-/- mice.J. Thromb. Haemost. 2016; 14: 2478-2492Crossref PubMed Scopus (8) Google Scholar, 53Girard-Gagnepain A. Amirache F. Costa C. Lévy C. Frecha C. Fusil F. Nègre D. Lavillette D. Cosset F.L. Verhoeyen E. Baboon envelope pseudotyped LVs outperform VSV-G-LVs for gene transfer into early-cytokine-stimulated and resting HSCs.Blood. 2014; 124: 1221-1231Crossref PubMed Scopus (31) Google Scholar which have been successfully used for the ex vivo modification of fully resting or minimally stimulated lymphocytes. The BaEV-pseudotyped LV was initially found to transduce mildly stimulated human HSCs at very high efficiency, thereby clearly outperforming VSV-LV.53Girard-Gagnepain A. Amirache F. Costa C. Lévy C. Frecha C. Fusil F. Nègre D. Lavillette D. Cosset F.L. Verhoeyen E. Baboon envelope pseudotyped LVs outperform VSV-G-LVs for gene transfer into early-cytokine-stimulated and resting HSCs.Blood. 2014; 124: 1221-1231Crossref PubMed Scopus (31) Google Scholar Subsequently, its clinical relevance was demonstrated by a study using the vector to deliver FIX into in vivo-differentiated human plasma B cells.39Levy C. Fusil F. Amirache F. Costa C. Girard-Gagnepain A. Negre D. Bernadin O. Garaulet G. Rodriguez A. Nair N. et al.Baboon envelope pseudotyped lentiviral vectors efficiently transduce human B cells and allow active factor IX B cell secretion in vivo in NOD/SCIDγc-/- mice.J. Thromb. Haemost. 2016; 14: 2478-2492Crossref PubMed Scopus (8) Google Scholar Following transduction by BaEV-LVFIX in vitro, f

Referência(s)