<iframe src="//www.googletagmanager.com/ns.html?id=GTM-5TSRKG" height="0" width="0" style="display: none; visibility: hidden">
Review Article
No access
Published Online: 1 December 2017

Pseudotyped Lentiviral Vectors: One Vector, Many Guises

Publication: Human Gene Therapy Methods
Volume 28, Issue Number 6

Abstract

Viruses have evolved specialized molecular mechanisms to transfer their genome efficiently into host cells. Viruses can be repurposed into viral vectors to achieve controlled gene transfer to desired cells. One of the most popular classes of vectors, lentiviral vectors (LVs), transduce mammalian cells efficiently. LVs are pseudotyped with various heterologous viral envelopes to alter their tropism. While the most common example is the envelope glycoprotein from vesicular stomatitis virus (VSVG), many other viral proteins have also been used. Pseudotyping LVs with a diverse set of naturally occurring or engineered viral envelopes has allowed targeted transduction of specific cell types. Many exciting studies are further uncovering new specificities and shortcomings of pseudotyped LVs. These studies will expand the toolbox to make LVs that cater to the specific requirements of transduction. This review provides a comprehensive overview of various viral envelope pseudotypes used with LVs, their specificities, advantages, and drawbacks.

Get full access to this article

View all available purchase options and get full access to this article.

References

1.
Blomer U, Naldini L, Kafri T, et al. Highly efficient and sustained gene transfer in adult neurons with a lentivirus vector. J Virol 1997;71:6641–6649.
2.
Naldini L, Blomer U, Gallay P, et al. In vivo gene delivery and stable transduction of nondividing cells by a lentiviral vector. Science 1996;272:263–267.
3.
Naldini L, Blomer U, Gage FH, et al. Efficient transfer, integration, and sustained long-term expression of the transgene in adult rat brains injected with a lentiviral vector. Proc Natl Acad Sci U S A 1996;93:11382–11388.
4.
Klatzmann D, Champagne E, Chamaret S, et al. T-lymphocyte T4 molecule behaves as the receptor for human retrovirus LAV. Nature 1984;312:767–768.
5.
Dalgleish AG, Beverley PC, Clapham PR, et al. The CD4 (T4) antigen is an essential component of the receptor for the AIDS retrovirus. Nature 1984;312:763–767.
6.
Moore JP, Trkola A, Dragic T. Co-receptors for HIV-1 entry. Curr Opin Immunol 1997;9:551–562.
7.
Burns JC, Friedmann T, Driever W, et al. Vesicular stomatitis virus G glycoprotein pseudotyped retroviral vectors: concentration to very high titer and efficient gene transfer into mammalian and nonmammalian cells. Proc Natl Acad Sci U S A 1993;90:8033–8037.
8.
Finkelshtein D, Werman A, Novick D, et al. LDL receptor and its family members serve as the cellular receptors for vesicular stomatitis virus. Proc Natl Acad Sci U S A 2013;110:7306–7311.
9.
Hioki H, Kameda H, Nakamura H, et al. Efficient gene transduction of neurons by lentivirus with enhanced neuron-specific promoters. Gene Ther 2007;14:872–882.
10.
Akkina RK, Walton RM, Chen ML, et al. High-efficiency gene transfer into CD34+ cells with a human immunodeficiency virus type 1-based retroviral vector pseudotyped with vesicular stomatitis virus envelope glycoprotein G. J Virol 1996;70:2581–2585.
11.
Amirache F, Levy C, Costa C, et al. 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;123:1422–1424.
12.
Pacchia AL, Adelson ME, Kaul M, et al. An inducible packaging cell system for safe, efficient lentiviral vector production in the absence of HIV-1 accessory proteins. Virology 2001;282:77–86.
13.
Ni Y, Sun S, Oparaocha I, et al. Generation of a packaging cell line for prolonged large-scale production of high-titer HIV-1-based lentiviral vector. J Gene Med 2005;7:818–834.
14.
Farson D, Witt R, McGuinness R, et al. A new-generation stable inducible packaging cell line for lentiviral vectors. Hum Gene Ther 2001;12:981–997.
15.
DePolo NJ, Reed JD, Sheridan PL, et al. VSV-G pseudotyped lentiviral vector particles produced in human cells are inactivated by human serum. Mol Ther 2000;2:218–222.
16.
Sakuma T, Barry MA, Ikeda Y. Lentiviral vectors: basic to translational. Biochem J 2012;443:603–618.
17.
Hu S, Mohan Kumar D, Sax C, et al. Pseudotyping of lentiviral vector with novel vesiculovirus envelope glycoproteins derived from Chandipura and Piry viruses. Virology 2016;488:162–168.
18.
Iyer AV, Pahar B, Boudreaux MJ, et al. Recombinant vesicular stomatitis virus-based West Nile vaccine elicits strong humoral and cellular immune responses and protects mice against lethal challenge with the virulent West Nile virus strain LSU-AR01. Vaccine 2009;27:893–903.
19.
Wong LF, Azzouz M, Walmsley LE, et al. Transduction patterns of pseudotyped lentiviral vectors in the nervous system. Mol Ther 2004;9:101–111.
20.
Humbert O, Gisch DW, Wohlfahrt ME, et al. Development of third-generation cocal envelope producer cell lines for robust lentiviral gene transfer into hematopoietic stem cells and T-cells. Mol Ther 2016;24:1237–1246.
21.
Hwang BY, Schaffer DV. Engineering a serum-resistant and thermostable vesicular stomatitis virus G glycoprotein for pseudotyping retroviral and lentiviral vectors. Gene Ther 2013;20:807–815.
22.
Kafri T, Blomer U, Peterson DA, et al. Sustained expression of genes delivered directly into liver and muscle by lentiviral vectors. Nat Genet 1997;17:314–317.
23.
Peng KW, Pham L, Ye H, et al. Organ distribution of gene expression after intravenous infusion of targeted and untargeted lentiviral vectors. Gene Ther 2001;8:1456–1463.
24.
Sakuma T, De Ravin SS, Tonne JM, et al. Characterization of retroviral and lentiviral vectors pseudotyped with xenotropic murine leukemia virus-related virus envelope glycoprotein. Hum Gene Ther 2010;21:1665–1673.
25.
Diaz RM, Bateman A, Emiliusen L, et al. A lentiviral vector expressing a fusogenic glycoprotein for cancer gene therapy. Gene Ther 2000;7:1656–1663.
26.
Jang JE, Shaw K, Yu XJ, et al. Specific and stable gene transfer to human embryonic stem cells using pseudotyped lentiviral vectors. Stem Cells Dev 2006;15:109–117.
27.
Bell AJ Jr, Fegen D, Ward M, et al. RD114 envelope proteins provide an effective and versatile approach to pseudotype lentiviral vectors. Exp Biol Med 2010;235:1269–1276.
28.
Di Nunzio F, Piovani B, Cosset FL, et al. Transduction of human hematopoietic stem cells by lentiviral vectors pseudotyped with the RD114-TR chimeric envelope glycoprotein. Hum Gene Ther 2007;18:811–820.
29.
Germain E, Roullin VG, Qiao J, et al. RD114-pseudotyped retroviral vectors kill cancer cells by syncytium formation and enhance the cytotoxic effect of the TK/GCV gene therapy strategy. J Gene Med 2005;7:389–397.
30.
Steffens S, Tebbets J, Kramm CM, Lindemann D, Flake A, Sena-Esteves M. Transduction of human glial and neuronal tumor cells with different lentivirus vector pseudotypes. J Neurooncol 2004;70:281–288.
31.
Hanawa H, Kelly PF, Nathwani AC, et al. Comparison of various envelope proteins for their ability to pseudotype lentiviral vectors and transduce primitive hematopoietic cells from human blood. Mol Ther 2002;5:242–251.
32.
Hanawa H, Hematti P, Keyvanfar K, et al. Efficient gene transfer into rhesus repopulating hematopoietic stem cells using a simian immunodeficiency virus-based lentiviral vector system. Blood 2004;103:4062–4069.
33.
Hale CR, Majumdar S, Elmore J, et al. Essential features and rational design of CRISPR RNAs that function with the Cas RAMP module complex to cleave RNAs. Mol Cell 2012;45:292–302.
34.
Schambach A, Galla M, Modlich U, et al. Lentiviral vectors pseudotyped with murine ecotropic envelope: increased biosafety and convenience in preclinical research. Exp Hematol 2006;34:588–592.
35.
Janssens W, Chuah MK, Naldini L, et al. Efficiency of onco-retroviral and lentiviral gene transfer into primary mouse and human B-lymphocytes is pseudotype dependent. Hum Gene Ther 2003;14:263–276.
36.
Sandrin V, Muriaux D, Darlix JL, et al. Intracellular trafficking of Gag and Env proteins and their interactions modulate pseudotyping of retroviruses. J Virol 2004;78:7153–7164.
37.
Sandrin V, Boson B, Salmon P, et al. Lentiviral vectors pseudotyped with a modified RD114 envelope glycoprotein show increased stability in sera and augmented transduction of primary lymphocytes and CD34+ cells derived from human and nonhuman primates. Blood 2002;100:823–832.
38.
Girard-Gagnepain A, Amirache F, Costa C, et al. Baboon envelope pseudotyped LVs outperform VSV-G-LVs for gene transfer into early-cytokine-stimulated and resting HSCs. Blood 2014;124:1221–1231.
39.
Levy C, Fusil F, Amirache F, et al. Baboon envelope pseudotyped lentiviral vectors efficiently transduce human B cells and allow active factor IX B cell secretion in vivo in NOD/SCIDgammac–/– mice. J Thromb Haemost 2016;14:2478–2492.
40.
Buchholz CJ, Muhlebach MD, Cichutek K. Lentiviral vectors with measles virus glycoproteins—dream team for gene transfer? Trends Biotechnol 2009;27:259–265.
41.
Uhlig KM, Schulke S, Scheuplein VA, et al. Lentiviral protein transfer vectors are an efficient vaccine platform and induce a strong antigen-specific cytotoxic T cell response. J Virol 2015;89:9044–9060.
42.
Frecha C, Costa C, Negre D, et al. Stable transduction of quiescent T cells without induction of cycle progression by a novel lentiviral vector pseudotyped with measles virus glycoproteins. Blood 2008;112:4843–4852.
43.
Zhou Q, Schneider IC, Gallet M, et al. Resting lymphocyte transduction with measles virus glycoprotein pseudotyped lentiviral vectors relies on CD46 and SLAM. Virology 2011;413:149–152.
44.
Frecha C, Levy C, Costa C, et al. Measles virus glycoprotein-pseudotyped lentiviral vector-mediated gene transfer into quiescent lymphocytes requires binding to both SLAM and CD46 entry receptors. J Virol 2011;85:5975–5985.
45.
Funke S, Schneider IC, Glaser S, et al. Pseudotyping lentiviral vectors with the wild-type measles virus glycoproteins improves titer and selectivity. Gene Ther 2009;16:700–705.
46.
Kneissl S, Abel T, Rasbach A, et al. Measles virus glycoprotein-based lentiviral targeting vectors that avoid neutralizing antibodies. PloS One 2012;7:e46667.
47.
Humbert JM, Frecha C, Amirache Bouafia F, et al. Measles virus glycoprotein-pseudotyped lentiviral vectors are highly superior to vesicular stomatitis virus G pseudotypes for genetic modification of monocyte-derived dendritic cells. J Virol 2012;86:5192–5203.
48.
Marino MP, Panigaj M, Ou W, et al. A scalable method to concentrate lentiviral vectors pseudotyped with measles virus glycoproteins. Gene Ther 2015;22:280–285.
49.
Witting SR, Vallanda P, Gamble AL. Characterization of a third generation lentiviral vector pseudotyped with Nipah virus envelope proteins for endothelial cell transduction. Gene Ther 2013;20:997–1005.
50.
Palomares K, Vigant F, Van Handel B, et al. Nipah virus envelope-pseudotyped lentiviruses efficiently target ephrinB2-positive stem cell populations in vitro and bypass the liver sink when administered in vivo. J Virol 2013;87:2094–2108.
51.
Kobayashi K, Kato S, Inoue K, et al. Altering entry site preference of lentiviral vectors into neuronal cells by pseudotyping with envelope glycoproteins. Methods Mol Biol 2016;1382:175–186.
52.
Mazarakis ND, Azzouz M, Rohll JB, et al. Rabies virus glycoprotein pseudotyping of lentiviral vectors enables retrograde axonal transport and access to the nervous system after peripheral delivery. Hum Mol Genet 2001;10:2109–2121.
53.
Hirano M, Kato S, Kobayashi K, et al. Highly efficient retrograde gene transfer into motor neurons by a lentiviral vector pseudotyped with fusion glycoprotein. PloS One 2013;8:e75896.
54.
Hislop JN, Islam TA, Eleftheriadou I, et al. Rabies virus envelope glycoprotein targets lentiviral vectors to the axonal retrograde pathway in motor neurons. J Biol Chem 2014;289:16148–16163.
55.
Kato S, Inoue K, Kobayashi K, et al. Efficient gene transfer via retrograde transport in rodent and primate brains using a human immunodeficiency virus type 1-based vector pseudotyped with rabies virus glycoprotein. Hum Gene Ther 2007;18:1141–1151.
56.
Schoderboeck L, Riad S, Bokor AM, et al. Chimeric rabies SADB19-VSVg-pseudotyped lentiviral vectors mediate long-range retrograde transduction from the mouse spinal cord. Gene Ther 2015;22:357–364.
57.
Carpentier DC, Vevis K, Trabalza A, et al. Enhanced pseudotyping efficiency of HIV-1 lentiviral vectors by a rabies/vesicular stomatitis virus chimeric envelope glycoprotein. Gene Ther 2012;19:761–774.
58.
Cronin J, Zhang XY, Reiser J. Altering the tropism of lentiviral vectors through pseudotyping. Curr Gene Ther 2005;5:387–398.
59.
Kato S, Kobayashi K, Kobayashi K. Improved transduction efficiency of a lentiviral vector for neuron-specific retrograde gene transfer by optimizing the junction of fusion envelope glycoprotein. J Neurosci Methods 2014;227:151–158.
60.
Trabalza A, Eleftheriadou I, Sgourou A, et al. Enhanced central nervous system transduction with lentiviral vectors pseudotyped with RVG/HIV-1gp41 chimeric envelope glycoproteins. J Virol 2014;88:2877–2890.
61.
Colin A, Faideau M, Dufour N, et al. Engineered lentiviral vector targeting astrocytes in vivo. Glia 2009;57:667–679.
62.
Hachiya A, Sriwiriyanont P, Patel A, et al. Gene transfer in human skin with different pseudotyped HIV-based vectors. Gene Ther 2007;14:648–656.
63.
Medina MF, Kobinger GP, Rux J, et al. Lentiviral vectors pseudotyped with minimal filovirus envelopes increased gene transfer in murine lung. Mol Ther 2003;8:777–789.
64.
Dylla DE, Xie L, Michele DE, et al. Altering alpha-dystroglycan receptor affinity of LCMV pseudotyped lentivirus yields unique cell and tissue tropism. Genet Vaccines Ther 2011;9:8.
65.
Zhang C, Hu B, Xiao L, et al. Pseudotyping lentiviral vectors with lymphocytic choriomeningitis virus glycoproteins for transduction of dendritic cells and in vivo immunization. Hum Gene Ther Methods 2014;25:328–338.
66.
Limberis MP, Bell CL, Heath J, et al. Activation of transgene-specific T cells following lentivirus-mediated gene delivery to mouse lung. Mol Ther 2010;18:143–150.
67.
Watson DJ, Kobinger GP, Passini MA, et al. Targeted transduction patterns in the mouse brain by lentivirus vectors pseudotyped with VSV, Ebola, Mokola, LCMV, or MuLV envelope proteins. Mol Ther 2002;5:528–537.
68.
Kumar M, Bradow BP, Zimmerberg J. Large-scale production of pseudotyped lentiviral vectors using baculovirus GP64. Hum Gene Ther 2003;14:67–77.
69.
Schauber CA, Tuerk MJ, Pacheco CD, et al. Lentiviral vectors pseudotyped with baculovirus gp64 efficiently transduce mouse cells in vivo and show tropism restriction against hematopoietic cell types in vitro. Gene Ther 2004;11:266–275.
70.
Sinn PL, Burnight ER, Hickey MA, et al. Persistent gene expression in mouse nasal epithelia following feline immunodeficiency virus-based vector gene transfer. J Virol 2005;79:12818–12827.
71.
Buckley SM, Howe SJ, Sheard V, et al. Lentiviral transduction of the murine lung provides efficient pseudotype and developmental stage-dependent cell-specific transgene expression. Gene Ther 2008;15:1167–1175.
72.
Kang Y, Stein CS, Heth JA, et al. In vivo gene transfer using a nonprimate lentiviral vector pseudotyped with Ross River virus glycoproteins. J Virol 2002;76:9378–9388.
73.
Kahl CA, Marsh J, Fyffe J, et al. Human immunodeficiency virus type 1-derived lentivirus vectors pseudotyped with envelope glycoproteins derived from Ross River virus and Semliki Forest virus. J Virol 2004;78:1421–1430.
74.
Jakobsson J, Nielsen TT, Staflin K, et al. Efficient transduction of neurons using Ross River glycoprotein-pseudotyped lentiviral vectors. Gene Ther 2006;13:966–973.
75.
Kahl CA, Pollok K, Haneline LS, et al. Lentiviral vectors pseudotyped with glycoproteins from Ross River and vesicular stomatitis viruses: variable transduction related to cell type and culture conditions. Mol Ther 2005;11:470–482.
76.
Kolokoltsov AA, Weaver SC, Davey RA. Efficient functional pseudotyping of oncoretroviral and lentiviral vectors by Venezuelan equine encephalitis virus envelope proteins. J Virol 2005;79:756–763.
77.
Poluri A, Ainsworth R, Weaver SC, et al. Functional pseudotyping of human immunodeficiency virus type 1 vectors by Western equine encephalitis virus envelope glycoprotein. J Virol 2008;82:12580–12584.
78.
Salvador B, Zhou Y, Michault A, et al. Characterization of Chikungunya pseudotyped viruses: Identification of refractory cell lines and demonstration of cellular tropism differences mediated by mutations in E1 glycoprotein. Virology2009;393:33–41.
79.
Eleftheriadou I, Dieringer M, Poh XY, et al. Selective transduction of astrocytic and neuronal CNS subpopulations by lentiviral vectors pseudotyped with Chikungunya virus envelope. Biomaterials 2017;123:1–14.
80.
McKay T, Patel M, Pickles RJ, et al. Influenza M2 envelope protein augments avian influenza hemagglutinin pseudotyping of lentiviral vectors. Gene Ther 2006;13:715–724.
81.
Patel M, Giddings AM, Sechelski J, et al. High efficiency gene transfer to airways of mice using influenza hemagglutinin pseudotyped lentiviral vectors. J Gene Med 2013;15:51–62.
82.
Schlecht-Louf G, Mangeney M, El-Garch H, et al. A targeted mutation within the feline leukemia virus (FeLV) envelope protein immunosuppressive domain to improve a canarypox virus-vectored FeLV vaccine. J Virol 2014;88:992–1001.
83.
Bupp K, Roth MJ. Targeting a retroviral vector in the absence of a known cell-targeting ligand. Hum Gene Ther 2003;14:1557–1564.
84.
Sarangi A, Bupp K, Roth MJ. Identification of a retroviral receptor used by an envelope protein derived by peptide library screening. Proc Natl Acad Sci U S A 2007;104:11032–11037.
85.
Snitkovsky S, Young JA. Targeting retroviral vector infection to cells that express heregulin receptors using a TVA-heregulin bridge protein. Virology 2002;292:150–155.
86.
Zhou Q, Uhlig KM, Muth A, et al. Exclusive transduction of human CD4+ T cells upon systemic delivery of CD4-targeted lentiviral vectors. J Immunol 2015;195:2493–2501.
87.
Yang L, Bailey L, Baltimore D, et al. Targeting lentiviral vectors to specific cell types in vivo. Proc Natl Acad Sci U S A 2006;103:11479–11484.
88.
Zhang X, Roth MJ. Antibody-directed lentiviral gene transduction in early immature hematopoietic progenitor cells. J Gene Med 2010;12:945–955.
89.
Maurice M, Verhoeyen E, Salmon P, et al. Efficient gene transfer into human primary blood lymphocytes by surface-engineered lentiviral vectors that display a T cell-activating polypeptide. Blood 2002;99:2342–2350.
90.
Zhou Q, Schneider IC, Edes I, et al. T-cell receptor gene transfer exclusively to human CD8(+) cells enhances tumor cell killing. Blood 2012;120:4334–4342.
91.
Kneissl S, Zhou Q, Schwenkert M, et al. CD19 and CD20 targeted vectors induce minimal activation of resting B lymphocytes. PloS One 2013;8:e79047.
92.
Anliker B, Abel T, Kneissl S, et al. Specific gene transfer to neurons, endothelial cells and hematopoietic progenitors with lentiviral vectors. Nat Methods 2010;7:929–935.
93.
Bender RR, Muth A, Schneider IC, et al. Receptor-targeted Nipah virus glycoproteins improve cell-type selective gene delivery and reveal a preference for membrane-proximal cell attachment. PLoS Pathog 2016;12:e1005641.
94.
Kutner RH, Zhang XY, Reiser J. Production, concentration and titration of pseudotyped HIV-1-based lentiviral vectors. Nat Protoc 2009;4:495–505.
95.
Merten OW, Hebben M, Bovolenta C. Production of lentiviral vectors. Mol Ther Methods Clin Dev 2016;3:16017.
96.
Ausubel LJ, Hall C, Sharma A, et al. Production of CGMP-grade lentiviral vectors. Bioprocess Int 2012;10:32–43.

Information & Authors

Information

Published In

cover image Human Gene Therapy Methods
Human Gene Therapy Methods
Volume 28Issue Number 6December 2017
Pages: 291 - 301
PubMed: 28870117

History

Published in print: December 2017
Published online: 1 December 2017
Published ahead of print: 4 September 2017
Published ahead of production: 4 September 2017
Accepted: 27 August 2017
Received: 16 May 2017

Permissions

Request permissions for this article.

Topics

    Authors

    Affiliations

    Alok V. Joglekar
    Department of Biology and Biological Engineering, California Institute of Technology, Pasadena, California.
    Salemiz Sandoval
    Department of Biology and Biological Engineering, California Institute of Technology, Pasadena, California.

    Notes

    These authors contributed equally to this work.
    *
    Correspondence: Dr. Alok Vishnu Joglekar, Department of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125. E-mail: [email protected]

    Author Disclosure

    S.S. is a full time employee of PACT Pharma, Inc. A.V.J. has no financial disclosures.

    Metrics & Citations

    Metrics

    Citations

    Export citation

    Select the format you want to export the citations of this publication.

    View Options

    Get Access

    Access content

    To read the fulltext, please use one of the options below to sign in or purchase access.

    Society Access

    If you are a member of a society that has access to this content please log in via your society website and then return to this publication.

    Restore your content access

    Enter your email address to restore your content access:

    Note: This functionality works only for purchases done as a guest. If you already have an account, log in to access the content to which you are entitled.

    View options

    PDF/EPUB

    View PDF/ePub

    Full Text

    View Full Text

    Media

    Figures

    Other

    Tables

    Share

    Share

    Copy the content Link

    Share on social media

    Back to Top