Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Short Communication
  • Published:

Development of operational immunologic tolerance with neonatal gene transfer in nonhuman primates: preliminary studies

Abstract

Achieving persistent expression is a prerequisite for effective genetic therapies for inherited disorders. These proof-of-concept studies focused on adeno-associated virus (AAV) administration to newborn monkeys. Serotype rh10 AAV expressing ovalbumin and green fluorescent protein (GFP) was administered intravenously at birth and compared with vehicle controls. At 4 months postnatal age, a second injection was administered intramuscularly, followed by vaccination at 1 year of age with ovalbumin and GFP. Ovalbumin was highest 2 weeks post administration in the treated monkey, which declined but remained detectable thereafter; controls demonstrated no expression. Long-term AAV genome copies were present in myocytes. At 4 weeks, neutralizing antibodies to rh10 were present in the experimental animal only. With AAV9 administration at 4 months, controls showed transient ovalbumin expression that disappeared with the development of strong anti-ovalbumin and anti-GFP antibodies. In contrast, increased and maintained ovalbumin expression was noted in the monkey administered AAV at birth, without antibody development. After vaccination, the experimental monkey maintained levels of ovalbumin without antibodies, whereas controls demonstrated high levels of antibodies. These preliminary studies suggest that newborn AAV administration expressing secreted and intracellular xenogenic proteins may result in persistent expression in muscle, and subsequent vector administration can result in augmented expression without humoral immune responses.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1
Figure 2
Figure 3
Figure 4
Figure 5
Figure 6

Similar content being viewed by others

References

  1. Nathwani AC, Tuddenham EG, Rangarajan S, Rosales C, McIntosh J, Linch DC et al. Adenovirus-associated virus vector-mediated gene transfer in hemophilia B. N Engl J Med 2011; 365: 2357–2365.

    Article  CAS  Google Scholar 

  2. Brantly ML, Chulay JD, Wang L, Mueller C, Humphries M, Spencer LT et al. Sustained transgene expression despite T lymphocyte responses in a clinical trial of rAAV1-AAT gene therapy. Proc Natl Acad Sci USA 2009; 106: 16363–16368.

    Article  CAS  Google Scholar 

  3. Cartier N, Hacein-Bey-Abina S, Bartholomae CC, Veres G, Schmidt M, Kutschera I et al. Hematopoietic stem cell gene therapy with a lentiviral vector in X-linked adrenoleukodystrophy. Science 2009; 326: 818–823.

    Article  CAS  Google Scholar 

  4. Cavazzana-Calvo M, Payen E, Negre O, Wang G, Hehir K, Fusil F et al. Transfusion independence and HMGA2 activation after gene therapy of human beta-thalassaemia. Nature 2010; 467: 318–322.

    Article  CAS  Google Scholar 

  5. Hacein-Bey-Abina S, Hauer J, Lim A, Picard C, Wang GP, Berry CC et al. Efficacy of gene therapy for X-linked severe combined immunodeficiency. N Engl J Med 2010; 363: 355–364.

    Article  CAS  Google Scholar 

  6. Byrne BJ, Falk DJ, Pacak CA, Nayak S, Herzog RW, Elder ME et al. Pompe disease gene therapy. Hum Mol Genet 2011; 20: R61–R68.

    Article  CAS  Google Scholar 

  7. Grez M, Reichenbach J, Schwable J, Seger R, Dinauer MC, Thrasher AJ . Gene therapy of chronic granulomatous disease: the engraftment dilemma. Mol Ther 2011; 19: 28–35.

    Article  CAS  Google Scholar 

  8. Bennett J, Ashtari M, Wellman J, Marshall KA, Cyckowski LL, Chung DC et al. AAV2 gene therapy readministration in three adults with congenital blindness. Sci Transl Med 2012; 4: 120ra15.

    Article  Google Scholar 

  9. Jacobson SG, Cideciyan AV, Ratnakaram R, Heon E, Schwartz SB, Roman AJ et al. Gene therapy for leber congenital amaurosis caused by RPE65 mutations: safety and efficacy in 15 children and adults followed up to 3 years. Arch Ophthalmol 2012; 130: 9–24.

    Article  CAS  Google Scholar 

  10. Scholler J, Brady TL, Binder-Scholl G, Hwang WT, Plesa G, Hege KM et al. Decade-long safety and function of retroviral-modified chimeric antigen receptor T cells. Sci Transl Med 2012; 4: 132ra53.

    Article  Google Scholar 

  11. Fields PA, Arruda VR, Armstrong E, Chu K, Mingozzi F, Hagstrom JN et al. Risk and prevention of anti-factor IX formation in AAV-mediated gene transfer in the context of a large deletion of F9. Mol Ther 2001; 4: 201–210.

    Article  CAS  Google Scholar 

  12. Herzog RW, Mount JD, Arruda VR, High KA, Lothrop CD Jr. . Muscle-directed gene transfer and transient immune suppression result in sustained partial correction of canine hemophilia B caused by a null mutation. Mol Ther 2001; 4: 192–200.

    Article  CAS  Google Scholar 

  13. Saint-Remy JM . Immunology of factor VIII inhibitors. Semin Thromb Hemost 2002; 28: 265–268.

    Article  CAS  Google Scholar 

  14. Kishnani PS, Goldenberg PC, DeArmey SL, Heller J, Benjamin D, Young S et al. Cross-reactive immunologic material status affects treatment outcomes in Pompe disease infants. Mol Genet Metab 2010; 99: 26–33.

    Article  CAS  Google Scholar 

  15. Hu C, Busuttil RW, Lipshutz GS . RH10 provides superior transgene expression in mice when compared with natural AAV serotypes for neonatal gene therapy. J Gene Med 2010; 12: 766–778.

    Article  CAS  Google Scholar 

  16. Hu C, Lipshutz GS . AAV-based neonatal gene therapy for hemophilia A: long-term correction and avoidance of immune responses in mice. Gene Ther 2012; 19: 1166–1176.

    Article  CAS  Google Scholar 

  17. Gau CL, Rosenblatt RA, Cerullo V, Lay FD, Dow AC, Livesay J et al. Short-term correction of arginase deficiency in a neonatal murine model with a helper-dependent adenoviral vector. Mol Ther 2009; 17: 1155–1163.

    Article  CAS  Google Scholar 

  18. Hu C, Cela RG, Suzuki M, Lee B, Lipshutz GS . Neonatal helper-dependent adenoviral vector gene therapy mediates correction of hemophilia A and tolerance to human factor VIII. Proc Natl Acad Sci USA 2011; 108: 2082–2087.

    Article  CAS  Google Scholar 

  19. Binny C, McIntosh J, Della Peruta M, Kymalainen H, Tuddenham EG, Buckley SM et al. AAV-mediated gene transfer in the perinatal period results in expression of FVII at levels that protect against fatal spontaneous hemorrhage. Blood 2012; 119: 957–966.

    Article  CAS  Google Scholar 

  20. Ridge JP, Fuchs EJ, Matzinger P . Neonatal tolerance revisited: turning on newborn T cells with dendritic cells. Science 1996; 271: 1723–1726.

    Article  CAS  Google Scholar 

  21. Tarantal AF, Skarlatos SI . Center for Fetal Monkey Gene Transfer for Heart, Lung, and Blood diseases: an NHLBI resource for the gene therapy community. Hum Gene Ther 2012; 23: 1130–1135.

    Article  CAS  Google Scholar 

  22. Lipshutz GS, Flebbe-Rehwaldt L, Gaensler KM . Reexpression following readministration of an adenoviral vector in adult mice after initial in utero adenoviral administration. Mol Ther 2000; 2: 374–380.

    Article  CAS  Google Scholar 

  23. Bogue M, Candeias S, Benoist C, Mathis D . A special repertoire of alpha:beta T cells in neonatal mice. EMBO J 1991; 10: 3647–3654.

    Article  CAS  Google Scholar 

  24. Chen N, Field EH . Enhanced type 2 and diminished type 1 cytokines in neonatal tolerance. Transplantation 1995; 59: 933–941.

    Article  CAS  Google Scholar 

  25. Darrasse-Jeze G, Marodon G, Salomon BL, Catala M, Klatzmann D . Ontogeny of CD4+CD25+ regulatory/suppressor T cells in human fetuses. Blood 2005; 105: 4715–4721.

    Article  CAS  Google Scholar 

  26. Mestas J, Hughes CC . Of mice and not men: differences between mouse and human immunology. J Immunol 2004; 172: 2731–2738.

    Article  CAS  Google Scholar 

  27. Bontrop RE . Non-human primates: essential partners in biomedical research. Immunol Rev 2001; 183: 5–9.

    Article  CAS  Google Scholar 

  28. Horn PA, Thomasson BM, Wood BL, Andrews RG, Morris JC, Kiem HP . Distinct hematopoietic stem/progenitor cell populations are responsible for repopulating NOD/SCID mice compared with nonhuman primates. Blood 2003; 102: 4329–4335.

    Article  CAS  Google Scholar 

  29. Batchelder CA, Duru N, Lee CI, Baker CA, Swainson L, McCune JM et al. Myeloid-lymphoid ontogeny in the rhesus monkey (Macaca mulatta. Anat Rec (Hoboken) 2014; 297: 1392–1406.

    Article  CAS  Google Scholar 

  30. Parsons TJ, Power C, Manor O . Fetal and early life growth and body mass index from birth to early adulthood in 1958 British cohort: longitudinal study. BMJ 2001; 323: 1331–1335.

    Article  CAS  Google Scholar 

  31. Nelson WE . Growth and development in the infant and child. In: Nelson WE (ed), Textbook of Pediatrics, 8th edn. W.B Saunders Company: Philadelphia, 1964, p 21.

    Google Scholar 

  32. Nelson WE . Growth and development in the infant and the child. In: Nelson WE (ed), Textbook of Pediatrics. W.B. Saunders Company: Philadelphia, 1964, p 24.

    Google Scholar 

  33. Lee EK, Hu C, Bhargava R, Rozengurt N, Stout D, Grody WW et al. Long-term survival of the juvenile lethal arginase-deficient mouse with AAV gene therapy. Mol Ther 2012; 20: 1844–1851.

    Article  CAS  Google Scholar 

  34. Wang L, Bell P, Lin J, Calcedo R, Tarantal AF, Wilson JM . AAV8-mediated hepatic gene transfer in infant rhesus monkeys (Macaca mulatta. Mol Ther 2011; 19: 2012–2020.

    Article  CAS  Google Scholar 

  35. Coppoletta JM, Wolbach SB . Body length and organ weights of infants and children: a study of the body length and normal weights of the more important vital organs of the body between birth and twelve years of age. Am J Pathol 1933; 9: 55–70.

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Garza KM, Agersborg SS, Baker E, Tung KS . Persistence of physiological self antigen is required for the regulation of self tolerance. J Immunol 2000; 164: 3982–3989.

    Article  CAS  Google Scholar 

  37. Jamieson BD, Ahmed R . T-cell tolerance: exposure to virus in utero does not cause a permanent deletion of specific T cells. Proc Natl Acad Sci USA 1988; 85: 2265–2268.

    Article  CAS  Google Scholar 

  38. Guibert J, Benachi A, Grebille AG, Ernault P, Zorn JR, Costa JM . Kinetics of SRY gene appearance in maternal serum: detection by real time PCR in early pregnancy after assisted reproductive technique. Hum Reprod 2003; 18: 1733–1736.

    Article  CAS  Google Scholar 

  39. Mays LE, Wilson JM . The complex and evolving story of T cell activation to AAV vector-encoded transgene products. Mol Ther 2011; 19: 16–27.

    Article  CAS  Google Scholar 

  40. Lock M, Alvira M, Vandenberghe LH, Samanta A, Toelen J, Debyser Z et al. Rapid, simple, and versatile manufacturing of recombinant adeno-associated viral vectors at scale. Hum Gene Ther 2010; 21: 1259–1271.

    Article  CAS  Google Scholar 

  41. Tarantal A . Ultrasound imaging in rhesus and long-tailed macaques: Reproductive and research applications. In: Wolfe-Coote S (ed), The Laboratory Primate, 1st edn. Academic Press, 2005, pp 317–351.

    Google Scholar 

  42. Tarantal AF, McDonald RJ, Jimenez DF, Lee CC, O'Shea CE, Leapley AC et al. Intrapulmonary and intramyocardial gene transfer in rhesus monkeys (Macaca mulatta: safety and efficiency of HIV-1-derived lentiviral vectors for fetal gene delivery. Mol Ther 2005; 12: 87–98.

    Article  CAS  Google Scholar 

  43. Calcedo R, Vandenberghe LH, Gao G, Lin J, Wilson JM . Worldwide epidemiology of neutralizing antibodies to adeno-associated viruses. J Infect Dis 2009; 199: 381–390.

    Article  Google Scholar 

Download references

Acknowledgements

This study was supported by the National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH) Center for Fetal Monkey Gene Transfer for Heart, Lung, and Blood Diseases (#HL85794; AFT); the Primate Center base operating grant (#OD011107) (AFT); NIH grants #NS071076, #NS071076-04S1, and #HD057555 (GSL); NIH grant AI108826-01 (GSL and AFT); and NIGMS Medical Genetics NIH T32 GM008243 and the Society of University Surgeons (DST). We thank Agustin Vega-Crespo and Sergio Duarte for their assistance with the fluorescence microscopy, Julie Johnston for helpful discussions and Roberto Calcedo for helpful discussions and for performing the neutralizing antibody assay.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to G S Lipshutz.

Ethics declarations

Competing interests

The authors declare no conflict of interest.

Additional information

Supplementary Information accompanies this paper on Gene Therapy website

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tai, D., Hu, C., Lee, C. et al. Development of operational immunologic tolerance with neonatal gene transfer in nonhuman primates: preliminary studies. Gene Ther 22, 923–930 (2015). https://doi.org/10.1038/gt.2015.65

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/gt.2015.65

This article is cited by

Search

Quick links