116
Views
1
CrossRef citations to date
0
Altmetric
Review Article

Current translational and clinical practices in hematopoietic cell and gene therapy

&
Pages 775-790 | Received 12 Apr 2012, Accepted 14 May 2012, Published online: 16 Jul 2012

References

  • Drolet BC, Lorenzi NM. Translational research: understanding the continuum from bench to bedside. Translation Res J Lab Clin Med. 2011;157:1–5.
  • Zerhouni EA. Clinical research at a crossroads: the NIH roadmap. J Invest Med. 2006;54:171–3.
  • Westfall JM, Mold J, Fagnan L. Practice-based research: ‘Blue Highways’ on the NIH roadmap. J Am Med Assoc. 2007;297:403–6.
  • Silvestri F, Banavali S, Baccarani M, Preisler HD. The CD34 hemopoietic progenitor cell associated antigen: biology and clinical applications. Haematologica. 1992;77:265–73.
  • Bensinger W, Appelbaum F, Rowley S, Storb R, Sanders J, Lilleby K, . Factors that influence collection and engraftment of autologous peripheral-blood stem cells. J Clin Oncol. 1995;13:2547–55.
  • Naldini L. Ex vivo gene transfer and correction for cell-based therapies. Nat Rev Genet. 2011;12:301–15.
  • Riviere I, Dunbar CE, Sadelain M. Hematopoietic stem cell engineering at a crossroads. Blood. 2012;119:1107–16.
  • Butturini A, Bortin MM, Gale RP. Graft-versus-leukemia following bone marrow transplantation. Bone Marrow Transplant. 1987;2:233–42.
  • Zaia JA. Prevention and management of CMV-related problems after hematopoietic stem cell transplantation. Bone Marrow Transplant. 2002;29:633–8.
  • Aguilar LK, Rooney CM, Heslop HE. Lymphoproliferative disorders involving Epstein–Barr virus after hemopoietic stem cell transplantation. Curr Opin Oncol. 1999;11:96–101.
  • Howard DS, Phillips IG, Reece DE, Munn RK, Henslee-Downey J, Pittard M, . Adenovirus infections in hematopoietic stem cell transplant recipients. Clin Infect Dis. 1999;29:1494–501.
  • Greenberg P, Goodrich J, Riddell S. Adoptive immunotherapy of human cytomegalovirus infection: potential role in protection from disease progression. Transplant Proc. 1991;23:97–101.
  • Riddell SR, Watanabe KS, Goodrich JM, Li CR, Agha ME, Greenberg PD. Restoration of viral immunity in immunodeficient humans by the adoptive transfer of T cell clones. Science. 1992;257:238–41.
  • Walter EA, Greenberg PD, Gilbert MJ, Finch RJ, Watanabe KS, Thomas ED, . Reconstitution of cellular immunity against cytomegalovirus in recipients of allogeneic bone marrow by transfer of T-cell clones from the donor. N Engl J Med. 1995;333:1038–44.
  • Heslop HE, Ng CY, Li C, Smith CA, Loftin SK, Krance RA, . Long-term restoration of immunity against Epstein– Barr virus infection by adoptive transfer of gene-modified virus-specific T lymphocytes. Nat Med. 1996;2:551–5.
  • Gottschalk S, Edwards OL, Sili U, Huls MH, Goltsova T, Davis AR, . Generating CTLs against the subdominant Epstein–Barr virus LMP1 antigen for the adoptive immunotherapy of EBV-associated malignancies. Blood. 2003;101: 1905–12.
  • Leen AM, Sili U, Savoldo B, Jewell AM, Piedra PA, Brenner MK, . Fiber-modified adenoviruses generate subgroup cross-reactive, adenovirus-specific cytotoxic T lymphocytes for therapeutic applications. Blood. 2004;103:1011–9.
  • O'Reilly RJ, Doubrovina E, Trivedi D, Hasan A, Kollen W, Koehne G. Adoptive transfer of antigen-specific T-cells of donor type for immunotherapy of viral infections following allogeneic hematopoietic cell transplants. Immunol Res. 2007;38:237–50.
  • Becker C, Pohla H, Frankenberger B, Schuler T, Assenmacher M, Schendel DJ, . Adoptive tumor therapy with T lymphocytes enriched through an IFN-gamma capture assay. Nat Med. 2001;7:1159–62.
  • Chatziandreou I, Gilmour KC, McNicol AM, Costabile M, Sinclair J, Cubitt D, . Capture and generation of adenovirus specific T cells for adoptive immunotherapy. Br J Haematol. 2007;136:117–26.
  • Kerns HM, Ryu BY, Stirling BV, Sather BD, Astrakhan A, Humblet-Baron S, . B cell-specific lentiviral gene therapy leads to sustained B-cell functional recovery in a murine model of X-linked agammaglobulinemia. Blood. 2010;115: 2146–55.
  • Leen AM, Myers GD, Sili U, Huls MH, Weiss H, Leung KS, . Monoculture-derived T lymphocytes specific for multiple viruses expand and produce clinically relevant effects in immunocompromised individuals. Nat Med. 2006;12: 1160–6.
  • Leen AM, Christin A, Myers GD, Liu H, Cruz CR, Hanley PJ, . Cytotoxic T lymphocyte therapy with donor T cells prevents and treats adenovirus and Epstein–Barr virus infections after haploidentical and matched unrelated stem cell transplantation. Blood. 2009;114:4283–92.
  • Hanley PJ, Shaffer DR, Cruz CR, Ku S, Tzou B, Liu H, . Expansion of T cells targeting multiple antigens of cytomegalovirus, Epstein–Barr virus and adenovirus to provide broad antiviral specificity after stem cell transplantation. Cytotherapy. 2011;13:976–86.
  • Heslop HE, Slobod KS, Pule MA, Hale GA, Rousseau A, Smith CA, . Long-term outcome of EBV-specific T-cell infusions to prevent or treat EBV-related lymphoproliferative disease in transplant recipients. Blood. 2010;115:925–35.
  • Peggs KS, Verfuerth S, Pizzey A, Khan N, Guiver M, Moss PA, . Adoptive cellular therapy for early cytomegalovirus infection after allogeneic stem-cell transplantation with virus-specific T-cell lines. Lancet. 2003;362:1375–7.
  • Zandvliet ML, van Liempt E, Jedema I, Veltrop-Duits LA, Willemze R, Guchelaar HJ, . Co-ordinated isolation of CD8(+) and CD4(+) T cells recognizing a broad repertoire of cytomegalovirus pp65 and IE1 epitopes for highly specific adoptive immunotherapy. Cytotherapy. 2010;12:933–44.
  • Cruz CR, Hanley PJ, Liu H, Torrano V, Lin YF, Arce JA, . Adverse events following infusion of T cells for adoptive immunotherapy: a 10-year experience. Cytotherapy. 2010; 12:743–9.
  • Sili U, Leen AM, Vera JF, Gee AP, Huls H, Heslop HE, . Production of good manufacturing practice-grade cytotoxic T lymphocytes specific for Epstein–Barr virus, cytomegalovirus and adenovirus to prevent or treat viral infections post-allogeneic hematopoietic stem cell transplant. Cytotherapy. 2012;14:7–11.
  • Rosenberg SA, Aebersold P, Cornetta K, Kasid A, Morgan RA, Moen R, . Gene transfer into humans: immunotherapy of patients with advanced melanoma, using tumor-infiltrating lymphocytes modified by retroviral gene transduction. N Engl J Med. 1990;323:570–8.
  • Dudley ME, Wunderlich JR, Shelton TE, Even J, Rosenberg SA. Generation of tumor-infiltrating lymphocyte cultures for use in adoptive transfer therapy for melanoma patients. J Immunother. 2003;26:332–42.
  • Dudley ME, Yang JC, Sherry R, Hughes MS, Royal R, Kammula U, . Adoptive cell therapy for patients with metastatic melanoma: evaluation of intensive myeloablative chemoradiation preparative regimens. J Clin Oncol. 2008;26: 5233–9.
  • Rosenberg SA, Dudley ME. Adoptive cell therapy for the treatment of patients with metastatic melanoma. Curr Opin Immunol. 2009;21:233–40.
  • Rosenberg SA, Yang JC, Sherry RM, Kammula US, Hughes MS, Phan GQ, . Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin Cancer Res. 2011; 17:4550–7.
  • Gajewski TF, Meng Y, Blank C, Brown I, Kacha A, Kline J, . Immune resistance orchestrated by the tumor microenvironment. Immunol Rev. 2006;213:131–45.
  • Gattinoni L, Finkelstein SE, Klebanoff CA, Antony PA, Palmer DC, Spiess PJ, . Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells. J Exp Med. 2005; 202:907–12.
  • Kim PS, Ahmed R. Features of responding T cells in cancer and chronic infection. Curr Opin Immunol. 2010;22: 223–30.
  • Baitsch L, Baumgaertner P, Devevre E, Raghav SK, Legat A, Barba L, . Exhaustion of tumor-specific CD8 T cells in metastases from melanoma patients. J Clin Invest. 2011;121:2350–60.
  • Ohta T, Kikuchi H, Ogata H, Iwata K, Yoshida K, Katayama K, . Effect of anti-CD3 antibody on the generation of interleukin-2-activated lymphocytes from tumor tissues of gastrointestinal cancer. Cancer. 1992;70:741–8.
  • Baxevanis CN, Dedoussis GV, Papadopoulos NG, Missitzis I, Stathopoulos GP, Papamichail M. Tumor specific cytolysis by tumor infiltrating lymphocytes in breast cancer. Cancer. 1994;74:1275–82.
  • Kass R, Bellone S, Palmieri M, Cane S, Bignotti E, Henry-Tillman R, . Restoration of tumor-specific HLA class I restricted cytotoxicity in tumor infiltrating lymphocytes of advanced breast cancer patients by in vitro stimulation with tumor antigen-pulsed autologous dendritic cells. Breast Cancer Res Treat. 2003;80:275–85.
  • Santin AD, Bellone S, Palmieri M, Bossini B, Cane S, Bignotti E, . Restoration of tumor specific human leukocyte antigen class I-restricted cytotoxicity by dendritic cell stimulation of tumor infiltrating lymphocytes in patients with advanced ovarian cancer. Int J Gynecol Cancer. 2004; 14:64–75.
  • Turcotte S, Rosenberg SA. Immunotherapy for metastatic solid cancers. Adv Surg. 2011;45:341–60.
  • Semino C, Martini L, Queirolo P, Cangemi G, Costa R, Alloisio A, . Adoptive immunotherapy of advanced solid tumors: an eight year clinical experience. Anticancer Res. 1999;19:5645–9.
  • Porter DL, Levine BL, Bunin N, Stadtmauer EA, Luger SM, Goldstein S, . A phase 1 trial of donor lymphocyte infusions expanded and activated ex vivo via CD3/CD28 costimulation. Blood. 2006;107:1325–31.
  • Rapoport AP, Aqui NA, Stadtmauer EA, Vogl DT, Fang HB, Cai L, . Combination immunotherapy using adoptive T-cell transfer and tumor antigen vaccination on the basis of hTERT and survivin after ASCT for myeloma. Blood. 2011;117:788–97.
  • Clay TM, Custer MC, Sachs J, Hwu P, Rosenberg SA, Nishimura MI. Efficient transfer of a tumor antigen-reactive TCR to human peripheral blood lymphocytes confers anti-tumor reactivity. J Immunol. 1999;163:507–13.
  • Fujio K, Misaki Y, Setoguchi K, Morita S, Kawahata K, Kato I, . Functional reconstitution of class II MHC-restricted T cell immunity mediated by retroviral transfer of the alpha beta TCR complex. J Immunol. 2000;165:528–32.
  • Cooper LJ, Kalos M, Lewinsohn DA, Riddell SR, Greenberg PD. Transfer of specificity for human immunodeficiency virus type 1 into primary human T lymphocytes by introduction of T-cell receptor genes. J Virol. 2000;74:8207–12.
  • Morgan RA, Dudley ME, Rosenberg SA. Adoptive cell therapy: genetic modification to redirect effector cell specificity. Cancer J. 2010;16:336–41.
  • Eshhar Z, Waks T, Gross G, Schindler DG. Specific activation and targeting of cytotoxic lymphocytes through chimeric single chains consisting of antibody-binding domains and the gamma or zeta subunits of the immunoglobulin and T-cell receptors. Proc Natl Acad Sci USA. 1993;90:720–4.
  • McKeever U, Khandekar S, Newcomb J, Naylor J, Gregory P, Brauer P, . Immunization with soluble BDC 2.5 T cell receptor-immunoglobulin chimeric protein: antibody specificity and protection of nonobese diabetic mice against adoptive transfer of diabetes by maternal immunization. J Exp Med. 1996;184:1755–68.
  • Brentjens R, Yeh R, Bernal Y, Riviere I, Sadelain M. Treatment of chronic lymphocytic leukemia with genetically targeted autologous T cells: case report of an unforeseen adverse event in a phase I clinical trial. Mol Ther. 2010;18:666–8.
  • Peinert S, Kershaw MH, Prince HM. Chimeric T cells for adoptive immunotherapy of cancer: using what have we learned to plan for the future. Immunotherapy. 2009;1: 905–12.
  • Levine BL. T lymphocyte engineering ex vivo for cancer and infectious disease. Expert Opin Biol Ther. 2008;8:475–89.
  • Park JR, Digiusto DL, Slovak M, Wright C, Naranjo A, Wagner J, . Adoptive transfer of chimeric antigen receptor re-directed cytolytic T lymphocyte clones in patients with neuroblastoma. Mol Ther. 2007;15:825–33.
  • Kohn DB, Dotti G, Brentjens R, Savoldo B, Jensen M, Cooper LJ, . CARs on track in the clinic. Mol Ther. 2011;19:432–8.
  • Berger C, Flowers ME, Warren EH, Riddell SR. Analysis of transgene-specific immune responses that limit the in vivo persistence of adoptively transferred HSV-TK-modified donor T cells after allogeneic hematopoietic cell transplantation. Blood. 2006;107:2294–302.
  • Jensen MC, Popplewell L, Cooper LJ, DiGiusto D, Kalos M, Ostberg JR, . Antitransgene rejection responses contribute to attenuated persistence of adoptively transferred CD20/CD19-specific chimeric antigen receptor redirected T cells in humans. Biol Blood Marrow Transplant. 2010;16:1245–56.
  • Hernandez-Chacon JA, Li Y, Wu RC, Bernatchez C, Wang Y, Weber JS, . Costimulation through the CD137/4-1BB pathway protects human melanoma tumor-infiltrating lymphocytes from activation-induced cell death and enhances antitumor effector function. J Immunother. 2011;34:236–50.
  • Hombach AA, Abken H. Costimulation by chimeric antigen receptors revisited the T cell antitumor response benefits from combined CD28-OX40 signalling. Int J Cancer. 2011;129:2935–44.
  • Kowolik CM, Topp MS, Gonzalez S, Pfeiffer T, Olivares S, Gonzalez N, . CD28 costimulation provided through a CD19-specific chimeric antigen receptor enhances in vivo persistence and antitumor efficacy of adoptively transferred T cells. Cancer Res. 2006;66:10995–1004.
  • Milone MC, Fish JD, Carpenito C, Carroll RG, Binder GK, Teachey D, . Chimeric receptors containing CD137 signal transduction domains mediate enhanced survival of T cells and increased antileukemic efficacy in vivo. Mol Ther. 2009;17:1453–64.
  • Song DG, Ye Q, Carpenito C, Poussin M, Wang LP, Ji C, . In vivo persistence, tumor localization, and antitumor activity of CAR-engineered T cells is enhanced by costimulatory signaling through CD137 (4-1BB). Cancer Res. 2011; 71:4617–27.
  • Cooper LJ, Al-Kadhimi Z, DiGiusto D, Kalos M, Colcher D, Raubitschek A, . Development and application of CD19-specific T cells for adoptive immunotherapy of B cell malignancies. Blood Cells Mol Dis. 2004;33:83–9.
  • Kochenderfer JN, Feldman SA, Zhao Y, Xu H, Black MA, Morgan RA, . Construction and preclinical evaluation of an anti-CD19 chimeric antigen receptor. J Immunother. 2009;32:689–702.
  • Kochenderfer JN, Yu Z, Frasheri D, Restifo NP, Rosenberg SA. Adoptive transfer of syngeneic T cells transduced with a chimeric antigen receptor that recognizes murine CD19 can eradicate lymphoma and normal B cells. Blood. 2010;116: 3875–86.
  • Singh H, Figliola MJ, Dawson MJ, Huls H, Olivares S, Switzer K, . Reprogramming CD19-specific T cells with IL-21 signaling can improve adoptive immunotherapy of B-lineage malignancies. Cancer Res. 2011;71:3516–27.
  • Terakura S, Yamamoto TN, Gardner RA, Turtle CJ, Jensen MC, Riddell SR. Generation of CD19-chimeric antigen receptor modified CD8 + T cells derived from virus-specific central memory T cells. Blood. 2012;119:72–82.
  • Hollyman D, Stefanski J, Przybylowski M, Bartido S, Borquez-Ojeda O, Taylor C, . Manufacturing validation of biologically functional T cells targeted to CD19 antigen for autologous adoptive cell therapy. J Immunother. 2009;32:169–80.
  • Brentjens RJ, Riviere I, Park JH, Davila ML, Wang X, Stefanski J, . Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood. 2011;118:4817–28.
  • Porter DL, Levine BL, Kalos M, Bagg A, June CH. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med. 2011;365:725–33.
  • Gattinoni L, Klebanoff CA, Palmer DC, Wrzesinski C, Kerstann K, Yu Z, . Acquisition of full effector function in vitro paradoxically impairs the in vivo antitumor efficacy of adoptively transferred CD8+ T cells. J Clin Invest. 2005; 115:1616–26.
  • Hinrichs CS, Borman ZA, Cassard L, Gattinoni L, Spolski R, Yu Z, . Adoptively transferred effector cells derived from naive rather than central memory CD8+ T cells mediate superior antitumor immunity. Proc Natl Acad Sci USA. 2009;106:17469–74.
  • Hinrichs CS, Borman ZA, Gattinoni L, Yu Z, Burns WR, Huang J, . Human effector CD8+ T cells derived from naive rather than memory subsets possess superior traits for adoptive immunotherapy. Blood. 2011;117:808–14.
  • Lan KL, Ou-Yang F, Yen SH, Shih HL, Lan KH. Cationic liposome coupled endostatin gene for treatment of peritoneal colon cancer. Clin Exp Metastasis. 2010;27: 307–18.
  • Berger C, Jensen MC, Lansdorp PM, Gough M, Elliott C, Riddell SR. Adoptive transfer of effector CD8+ T cells derived from central memory cells establishes persistent T cell memory in primates. J Clin Invest. 2008;118:294–305.
  • Turtle CJ, Swanson HM, Fujii N, Estey EH, Riddell SR. A distinct subset of self-renewing human memory CD8+ T cells survives cytotoxic chemotherapy. Immunity. 2009;31: 834–44.
  • Gennery AR, Slatter MA, Grandin L, Taupin P, Cant AJ, Veys P, . Transplantation of hematopoietic stem cells and long-term survival for primary immunodeficiencies in Europe: entering a new century, do we do better? J Allergy Clin Immunol. 2010;126:602–10.
  • Szabolcs P, Cavazzana-Calvo M, Fischer A, Veys P. Bone marrow transplantation for primary immunodeficiency diseases. Pediatr Clin North Am. 2010;57:207–37.
  • Prasad VK, Kurtzberg J. Cord blood and bone marrow transplantation in inherited metabolic diseases: scientific basis, current status and future directions. Br J Haematol. 2010;148:356–72.
  • Hutter G, Zaia JA. Allogeneic haematopoietic stem cell transplantation in patients with human immunodeficiency virus: the experiences of more than 25 years. Clin Exp Immunol. 2011;163:284–95.
  • Bordignon C, Notarangelo LD, Nobili N, Ferrari G, Casorati G, Panina P, . Gene therapy in peripheral blood lymphocytes and bone marrow for ADA-immunodeficient patients. Science. 1995;270:470–5.
  • Aiuti A, Slavin S, Aker M, Ficara F, Deola S, Mortellaro A, . Correction of ADA-SCID by stem cell gene therapy combined with nonmyeloablative conditioning. Science. 2002;296:2410–3.
  • Kohn DB, Hershfield MS, Carbonaro D, Shigeoka A, Brooks J, Smogorzewska EM, . T lymphocytes with a normal ADA gene accumulate after transplantation of transduced autologous umbilical cord blood CD34+ cells in ADA-deficient SCID neonates. Nat Med. 1998;4:775–80.
  • Aiuti A, Cattaneo F, Galimberti S, Benninghoff U, Cassani B, Callegaro L, . Gene therapy for immunodeficiency due to adenosine deaminase deficiency. N Engl J Med. 2009;360:447–58.
  • Ljungman P, Bregni M, Brune M, Cornelissen J, de Witte T, Dini G, . Allogeneic and autologous transplantation for haematological diseases, solid tumours and immune disorders: current practice in Europe 2009. Bone Marrow Transplant. 2010;45:219–34.
  • Hacein-Bey-Abina S, Garrigue A, Wang GP, Soulier J, Lim A, Morillon E, . Insertional oncogenesis in 4 patients after retrovirus-mediated gene therapy of SCID-X1. J Clin Invest. 2008;118:3132–42.
  • Howe SJ, Mansour MR, Schwarzwaelder K, Bartholomae C, Hubank M, Kempski H, . Insertional mutagenesis combined with acquired somatic mutations causes leukemogenesis following gene therapy of SCID-X1 patients. J Clin Invest. 2008;118:3143–50.
  • Gaspar HB, Cooray S, Gilmour KC, Parsley KL, Adams S, Howe SJ, . Long-term persistence of a polyclonal T cell repertoire after gene therapy for X-linked severe combined immunodeficiency. Sci Transl Med. 2011;3:97ra79.
  • Thrasher AJ, Hacein-Bey-Abina S, Gaspar HB, Blanche S, Davies EG, Parsley K, . Failure of SCID-X1 gene therapy in older patients. Blood. 2005;105:4255–7.
  • Boztug K, Schmidt M, Schwarzer A, Banerjee PP, Diez IA, Dewey RA, . Stem-cell gene therapy for the Wiskott–Aldrich syndrome. N Engl J Med. 2010;363: 1918–27.
  • Stein S, Ott MG, Schultze-Strasser S, Jauch A, Burwinkel B, Kinner A, . Genomic instability and myelodysplasia with monosomy 7 consequent to EVI1 activation after gene therapy for chronic granulomatous disease. Nat Med. 2010;16:198–204.
  • Ott MG, Schmidt M, Schwarzwaelder K, Stein S, Siler U, Koehl U, . Correction of X-linked chronic granulomatous disease by gene therapy, augmented by insertional activation of MDS1-EVI1, PRDM16 or SETBP1. Nat Med. 2006; 12:401–9.
  • Scholler J, Brady TL, Binder-Scholl G, Hwang WT, Plesa G, Hege KM, . Decade-long safety and function of retroviral-modified chimeric antigen receptor T cells. Sci Transl Med. 2012;4:132ra53.
  • Cartier N, Hacein-Bey-Abina S, Bartholomae CC, Veres G, Schmidt M, Kutschera I, . Hematopoietic stem cell gene therapy with a lentiviral vector in X-linked adrenoleukodystrophy. Science. 2009;326:818–23.
  • Biffi A, Bartolomae CC, Cesana D, Cartier N, Aubourg P, Ranzani M, . Lentiviral vector common integration sites in preclinical models and a clinical trial reflect a benign integration bias and not oncogenic selection. Blood. 2011;117: 5332–9.
  • Cavazzana-Calvo M, Payen E, Negre O, Wang G, Hehir K, Fusil F, . Transfusion independence and HMGA2 activation after gene therapy of human beta-thalassaemia. Nature. 2010;467:318–22.
  • Nathwani AC, Tuddenham EG, Rangarajan S, Rosales C, McIntosh J, Linch DC, . Adenovirus-associated virus vector-mediated gene transfer in hemophilia B. N Engl J Med. 2011;365:2357–65.
  • Roy K, Stein L, Kaushal S. Ocular gene therapy: an evaluation of recombinant adeno-associated virus-mediated gene therapy interventions for the treatment of ocular disease. Hum Gene Ther. 2010;21:915–27.
  • DiGiusto DL, Krishnan A, Li L, Li H, Li S, Rao A, . RNA-based gene therapy for HIV with lentiviral vector-modified CD34(+) cells in patients undergoing transplantation for AIDS-related lymphoma. Sci Transl Med. 2010; 36ra43.
  • Krishnan A, Molina A, Zaia J, Smith D, Vasquez D, Kogut N, . Durable remissions with autologous stem cell transplantation for high-risk HIV-associated lymphomas. Blood. 2005;105:874–8.
  • Holt N, Wang J, Kim K, Friedman G, Wang X, Taupin V, . Human hematopoietic stem/progenitor cells modified by zinc-finger nucleases targeted to CCR5 control HIV-1 in vivo. Nat Biotechnol. 2010;28:839–47.
  • Podsakoff GM, Engel BC, Carbonaro DA, Choi C, Smogorzewska EM, Bauer G, . Selective survival of peripheral blood lymphocytes in children with HIV-1 following delivery of an anti-HIV gene to bone marrow CD34(+) cells. Mol Ther. 2005;12:77–86.
  • Zinno F, Landi F, Scerpa MC, Aureli V, Lanti A, Ceccarelli S, . Processing of hematopoietic stem cells from peripheral blood before cryopreservation: use of a closed automated system. Transfusion. 2011;51:2656–63.
  • Janssen WE, Ribickas A, Meyer LV, Smilee RC. Large-scale Ficoll gradient separations using a commercially available, effectively closed, system. Cytotherapy. 2010;12:418–24.
  • Aktas M, Radke TF, Strauer BE, Wernet P, Kogler G. Separation of adult bone marrow mononuclear cells using the automated closed separation system Sepax. Cytotherapy. 2008;10:203–11.
  • Powell DJ Jr, Brennan AL, Zheng Z, Huynh H, Cotte J, Levine BL. Efficient clinical-scale enrichment of lymphocytes for use in adoptive immunotherapy using a modified counterflow centrifugal elutriation program. Cytotherapy. 2009;11: 923–35.
  • Perseghin P, D'Amico G, Dander E, Gaipa G, Dassi M, Biagi E, . Isolation of monocytes from leukapheretic products for large-scale GMP-grade generation of cytomegalovirus-specific T-cell lines by means of an automated elutriation device. Transfusion. 2008;48:1644–9.
  • Kim S, Kim HO, Baek EJ, Choi Y, Kim HS, Lee MG. Monocyte enrichment from leukapheresis products by using the Elutra cell separator. Transfusion. 2007;47:2290–6.
  • Gonzalez-Vicent M, Perez A, Abad L, Sevilla J, Ramirez M, Diaz MA. Graft manipulation and reduced-intensity conditioning for allogeneic hematopoietic stem cell transplantation from mismatched unrelated and mismatched/haploidentical related donors in pediatric leukemia patients. J Pediatr Hematol Oncol. 2010;32:85–90.
  • Devine SM, Carter S, Soiffer RJ, Pasquini MC, Hari PN, Stein A, . Low risk of chronic graft-versus-host disease and relapse associated with T cell-depleted peripheral blood stem cell transplantation for acute myelogenous leukemia in first remission: results of the blood and marrow transplant clinical trials network protocol 0303. Biol Blood Marrow Transplant. 2011;17:1343–51.
  • Prieto PA, Durflinger KH, Wunderlich JR, Rosenberg SA, Dudley ME. Enrichment of CD8+ cells from melanoma tumor-infiltrating lymphocyte cultures reveals tumor reactivity for use in adoptive cell therapy. J Immunother. 2010;33: 547–56.
  • Feuchtinger T, Opherk K, Bethge WA, Topp MS, Schuster FR, Weissinger EM, . Adoptive transfer of pp65-specific T cells for the treatment of chemorefractory cytomegalovirus disease or reactivation after haploidentical and matched unrelated stem cell transplantation. Blood. 2010;116:4360–7.
  • Di Nicola M, Zappasodi R, Carlo-Stella C, Mortarini R, Pupa SM, Magni M, . Vaccination with autologous tumor-loaded dendritic cells induces clinical and immunologic responses in indolent B-cell lymphoma patients with relapsed and measurable disease: a pilot study. Blood. 2009;113:18–27.
  • Hardy NM, Fellowes V, Rose JJ, Odom J, Pittaluga S, Steinberg SM, . Costimulated tumor-infiltrating lymphocytes are a feasible and safe alternative donor cell therapy for relapse after allogeneic stem cell transplantation. Blood. 2012;119:2956–2959.
  • Brunstein CG, Miller JS, Cao Q, McKenna DH, Hippen KL, Curtsinger J, . Infusion of ex vivo expanded T regulatory cells in adults transplanted with umbilical cord blood: safety profile and detection kinetics. Blood. 2011;117:1061–70.
  • Rapoport AP, Stadtmauer EA, Aqui N, Vogl D, Chew A, Fang HB, . Rapid immune recovery and graft-versus-host disease-like engraftment syndrome following adoptive transfer of costimulated autologous T cells. Clin Cancer Res. 2009;15:4499–507.
  • Bernstein WB, Cox JH, Aronson NE, Tracy L, Schlienger K, Ratto-Kim S, . Immune reconstitution following autologous transfers of CD3/CD28 stimulated CD4(+) T cells to HIV-infected persons. Clin Immunol. 2004;111: 262–74.
  • Orchard PJ, Blazar BR, Burger S, Levine B, Basso L, Nelson DM, . Clinical-scale selection of anti-CD3/CD28-activated T cells after transduction with a retroviral vector expressing herpes simplex virus thymidine kinase and truncated nerve growth factor receptor. Hum Gene Ther. 2002;13:979–88.
  • Notta F, Doulatov S, Laurenti E, Poeppl A, Jurisica I, Dick JE. Isolation of single human hematopoietic stem cells capable of long-term multilineage engraftment. Science. 2011;333: 218–21.
  • Klebanoff CA, Gattinoni L, Torabi-Parizi P, Kerstann K, Cardones AR, Finkelstein SE, . Central memory self/tumor-reactive CD8+ T cells confer superior antitumor immunity compared with effector memory T cells. Proc Natl Acad Sci USA. 2005;102:9571–6.
  • Gattinoni L, Lugli E, Ji Y, Pos Z, Paulos CM, Quigley MF, . A human memory T cell subset with stem cell-like properties. Nat Med. 2011;17:1290–7.
  • Spanholtz J, Preijers F, Tordoir M, Trilsbeek C, Paardekooper J, de Witte T, . Clinical-grade generation of active NK cells from cord blood hematopoietic progenitor cells for immunotherapy using a closed-system culture process. PLoS One. 2011;6:e20740.
  • Sensebe L. Clinical grade production of mesenchymal stem cells. Biomed Mater Eng. 2008;18:S3–10.
  • Tran CA, Burton L, Russom D, Wagner JR, Jensen MC, Forman SJ, . Manufacturing of large numbers of patient-specific T cells for adoptive immunotherapy: an approach to improving product safety, composition, and production capacity. J Immunother. 2007;30:644–54.
  • Vera JF, Brenner LJ, Gerdemann U, Ngo MC, Sili U, Liu H, . Accelerated production of antigen-specific T cells for preclinical and clinical applications using gas-permeable rapid expansion cultureware (G-Rex). J Immunother. 2010; 33:305–15.
  • Sadeghi A, Pauler L, Anneren C, Friberg A, Brandhorst D, Korsgren O, . Large-scale bioreactor expansion of tumor-infiltrating lymphocytes. J Immunol Methods. 2011; 364:94–100.
  • Hami LS, Green C, Leshinsky N, Markham E, Miller K, Craig S. GMP production and testing of Xcellerated T cells for the treatment of patients with CLL. Cytotherapy. 2004;6:554–62.
  • Delaney C, Heimfeld S, Brashem-Stein C, Voorhies H, Manger RL, Bernstein ID. Notch-mediated expansion of human cord blood progenitor cells capable of rapid myeloid reconstitution. Nat Med. 2010;16:232–6.
  • Choi B, Chun E, Kim SY, Kim M, Lee KY, Kim SJ. Notch-induced hIL-6 production facilitates the maintenance of self-renewal of hCD34+ cord blood cells through the activation of Jak-PI3K-STAT3 pathway. Am J Pathol. 2012;180: 351–64.
  • Auvray C, Delahaye A, Pflumio F, Haddad R, Amsellem S, Miri-Nezhad A, . HOXC4 homeoprotein efficiently expands human hematopoietic stem cells and triggers similar molecular alterations as HOXB4. Haematologica. 2012;97: 168–78.
  • Watts KL, Nelson V, Wood BL, Trobridge GD, Beard BC, Humphries RK, . Hematopoietic stem cell expansion facilitates multilineage engraftment in a nonhuman primate cord blood transplantation model. Exp Hematol. 2011;40:187–196.
  • Boitano AE, Wang J, Romeo R, Bouchez LC, Parker AE, Sutton SE, . Aryl hydrocarbon receptor antagonists promote the expansion of human hematopoietic stem cells. Science. 2010;329:1345–8.
  • 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: 4329–35.
  • Mezquita P, Beard BC, Kiem HP. NOD/SCID repopulating cells contribute only to short-term repopulation in the baboon. Gene Ther. 2008;15:1460–2.
  • Kohn DB. Lentiviral vectors ready for prime-time. Nat Biotechnol. 2007;25:65–6.
  • Papanikolaou E, Anagnou NP. Major challenges for gene therapy of thalassemia and sickle cell disease. Curr Gene Ther. 2010;10:404–12.
  • Dropulic B. Lentiviral vectors: their molecular design, safety, and use in laboratory and preclinical research. Hum Gene Ther. 2011;22:649–57.
  • Klages N, Zufferey R, Trono D. A stable system for the high-titer production of multiply attenuated lentiviral vectors. Mol Ther. 2000;2:170–6.
  • Farson D, Witt R, McGuinness R, Dull T, Kelly M, Song J, . A new-generation stable inducible packaging cell line for lentiviral vectors. Hum Gene Ther. 2001;12:981–97.
  • Xu K, Ma H, McCown TJ, Verma IM, Kafri T. Generation of a stable cell line producing high-titer self-inactivating lentiviral vectors. Mol Ther. 2001;3:97–104.
  • Ni Y, Sun S, Oparaocha I, Humeau L, Davis B, Cohen R, . Generation of a packaging cell line for prolonged large-scale production of high-titer HIV-1-based lentiviral vector. J Gene Med. 2005;7:818–34.
  • Witting SR, Li LH, Jasti A, Allen C, Cornetta K, Brady J, . Efficient large volume lentiviral vector production using flow electroporation. Hum Gene Ther. Volume 23(2): 243–9.
  • Pacchia AL, Adelson ME, Kaul M, Ron Y, Dougherty JP. An inducible packaging cell system for safe, efficient lentiviral vector production in the absence of HIV-1 accessory proteins. Virology. 2001;282:77–86.
  • Broussau S, Jabbour N, Lachapelle G, Durocher Y, Tom R, Transfiguracion J, . Inducible packaging cells for large-scale production of lentiviral vectors in serum-free suspension culture. Mol Ther. 2008;16:500–7.
  • Stewart HJ, Leroux-Carlucci MA, Sion CJ, Mitrophanous KA, Radcliffe PA. Development of inducible EIAV-based lentiviral vector packaging and producer cell lines. Gene Ther. 2009;16:805–14.
  • Merten OW, Charrier S, Laroudie N, Fauchille S, Dugue C, Jenny C, . Large-scale manufacture and characterization of a lentiviral vector produced for clinical ex vivo gene therapy application. Hum Gene Ther. 2011;22:343–56.
  • Lombardo A, Genovese P, Beausejour CM, Colleoni S, Lee YL, Kim KA, . Gene editing in human stem cells using zinc finger nucleases and integrase-defective lentiviral vector delivery. Nat Biotechnol. 2007;25:1298–306.
  • Doyon Y, Vo TD, Mendel MC, Greenberg SG, Wang J, Xia DF, . Enhancing zinc-finger-nuclease activity with improved obligate heterodimeric architectures. Nat Methods. 2011;8:74–9.
  • Miller JC, Holmes MC, Wang J, Guschin DY, Lee YL, Rupniewski I, . An improved zinc-finger nuclease architecture for highly specific genome editing. Nat Biotechnol. 2007;25: 778–85.
  • Urnov FD, Miller JC, Lee YL, Beausejour CM, Rock JM, Augustus S, . Highly efficient endogenous human gene correction using designed zinc-finger nucleases. Nature. 2005;435:646–51.
  • Carroll D, Morton JJ, Beumer KJ, Segal DJ. Design, construction and in vitro testing of zinc finger nucleases. Nat Protoc. 2006;1:1329–41.
  • Porteus MH. Mammalian gene targeting with designed zinc finger nucleases. Mol Ther. 2006;13:438–46.
  • Kim YG, Cha J, Chandrasegaran S. Hybrid restriction enzymes: zinc finger fusions to Fok I cleavage domain. Proc Natl Acad Sci USA. 1996;93:1156–60.
  • Cannon P, June C. Chemokine receptor 5 knockout strategies. Curr Opin HIV AIDS. 2011;6:74–9.
  • Wilen CB, Wang J, Tilton JC, Miller JC, Kim KA, Rebar EJ, . Engineering HIV-resistant human CD4+ T cells with CXCR4-specific zinc-finger nucleases. PLoS Pathog. 2011;7: e1002020.
  • Sebastiano V, Maeder ML, Angstman JF, Haddad B, Khayter C, Yeo DT, . In situ genetic correction of the sickle cell anemia mutation in human induced pluripotent stem cells using engineered zinc finger nucleases. Stem Cells. 2011;29: 1717–26.
  • Li H, Haurigot V, Doyon Y, Li T, Wong SY, Bhagwat AS, . In vivo genome editing restores haemostasis in a mouse model of haemophilia. Nature. 2011;475:217–21.
  • Yusa K, Rashid ST, Strick-Marchand H, Varela I, Liu PQ, Paschon DE, . Targeted gene correction of alpha1-antitrypsin deficiency in induced pluripotent stem cells. Nature. 2011;478:391–4.
  • Ramalingam S, Kandavelou K, Rajenderan R, Chandrasegaran S. Creating designed zinc-finger nucleases with minimal cytotoxicity. J Mol Biol. 2011;405:630–41.
  • Cornu TI, Cathomen T. Quantification of zinc finger nuclease-associated toxicity. Methods Mol Biol. 2010;649:237–45.
  • Pruett-Miller SM, Reading DW, Porter SN, Porteus MH. Attenuation of zinc finger nuclease toxicity by small-molecule regulation of protein levels. PLoS Genet. 2009;5:e1000376.
  • Pattanayak V, Ramirez CL, Joung JK, Liu DR. Revealing off-target cleavage specificities of zinc-finger nucleases by in vitro selection. Nat Methods. 2011;8:765–70.
  • Li L, Piatek MJ, Atef A, Piatek A, Wibowo A, Fang X, . Rapid and highly efficient construction of TALE-based transcriptional regulators and nucleases for genome modification. Plant Mol Biol. 78:407–16.
  • Mussolino C, Morbitzer R, Lutge F, Dannemann N, Lahaye T, Cathomen T. A novel TALE nuclease scaffold enables high genome editing activity in combination with low toxicity. Nucleic Acids Res. 2011;39:9283–93.
  • Arnould S, Chames P, Perez C, Lacroix E, Duclert A, Epinat JC, . Engineering of large numbers of highly specific homing endonucleases that induce recombination on novel DNA targets. J Mol Biol. 2006;355:443–58.
  • Stoddard BL. Homing endonucleases: from microbial genetic invaders to reagents for targeted DNA modification. Structure. 2011;19:7–15.
  • Burnett JC, Rossi JJ, Tiemann K. Current progress of siRNA/shRNA therapeutics in clinical trials. Biotechnol J. 2011;6: 1130–46.
  • Ehsani A, Saetrom P, Zhang J, Alluin J, Li H, Snove O Jr, . Rational design of micro-RNA-like bifunctional siRNAs targeting HIV and the HIV coreceptor CCR5. Mol Ther. 2010;18:796–802.
  • Chattopadhyay S, Ely A, Bloom K, Weinberg MS, Arbuthnot P. Inhibition of hepatitis B virus replication with linear DNA sequences expressing antiviral micro-RNA shuttles. Biochem Biophys Res Commun. 2009;389:484–9.
  • Shrivastava N, Srivastava A. RNA interference: an emerging generation of biologicals. Biotechnol J. 2008;3:339–53.
  • Izquierdo M. Short interfering RNAs as a tool for cancer gene therapy. Cancer Gene Ther. 2005;12:217–27.
  • Brown BD, Naldini L. Exploiting and antagonizing microRNA regulation for therapeutic and experimental applications. Nat Rev Genet. 2009;10:578–85.
  • Zhong YQ, Xia ZS, Fu YR, Zhu ZH. Knockdown of hTERT by SiRNA suppresses growth of Capan-2 human pancreatic cancer cell via the inhibition of expressions of Bcl-2 and COX-2. J Dig Dis. 2010;11:176–84.
  • Li MJ, Bauer G, Michienzi A, Yee JK, Lee NS, Kim J, . Inhibition of HIV-1 infection by lentiviral vectors expressing Pol III-promoted anti-HIV RNAs. Mol Ther. 2003;8:196–206.
  • Anderson J, Akkina R. Complete knockdown of CCR5 by lentiviral vector-expressed siRNAs and protection of transgenic macrophages against HIV-1 infection. Gene Ther. 2007;14:1287–97.
  • Kunze D, Wuttig D, Fuessel S, Kraemer K, Kotzsch M, Meye A, . Multitarget siRNA inhibition of antiapoptotic genes (XIAP, BCL2, BCL-X(L)) in bladder cancer cells. Anticancer Res. 2008;28:2259–63.
  • Brown BD, Gentner B, Cantore A, Colleoni S, Amendola M, Zingale A, . Endogenous microRNA can be broadly exploited to regulate transgene expression according to tissue, lineage and differentiation state. Nat Biotechnol. 2007;25:1457–67.
  • Ciceri F, Bonini C, Stanghellini MT, Bondanza A, Traversari C, Salomoni M, . Infusion of suicide-gene-engineered donor lymphocytes after family haploidentical haemopoietic stem-cell transplantation for leukaemia (the TK007 trial): a non-randomised phase I-II study. Lancet Oncol. 2009;10: 489–500.
  • Di Stasi A, Tey SK, Dotti G, Fujita Y, Kennedy-Nasser A, Martinez C, . Inducible apoptosis as a safety switch for adoptive cell therapy. N Engl J Med. 2011;365:1673–83.
  • Kaneko S, Mastaglio S, Bondanza A, Ponzoni M, Sanvito F, Aldrighetti L, . IL-7 and IL-15 allow the generation of suicide gene-modified alloreactive self-renewing central memory human T lymphocytes. Blood. 2009;113:1006–15.
  • Li L, Liu LN, Feller S, Allen C, Shivakumar R, Fratantoni J, . Expression of chimeric antigen receptors in natural killer cells with a regulatory-compliant non-viral method. Cancer Gene Ther. 2010;17:147–54.
  • Liu LN, Shivakumar R, Allen C, Fratantoni JC. Delivery of whole tumor lysate into dendritic cells for cancer vaccination. Methods Mol Biol. 2008;423:139–53.
  • Hackett PB, Largaespada DA, Cooper LJ. A transposon and transposase system for human application. Mol Ther. 2010;18:674–83.
  • McLachlan G, Davidson H, Holder E, Davies LA, Pringle IA, Sumner-Jones SG, . Pre-clinical evaluation of three non-viral gene transfer agents for cystic fibrosis after aerosol delivery to the ovine lung. Gene Ther. 2011;18:996–1005.
  • Fenske DB, Chonn A, Cullis PR. Liposomal nanomedicines: an emerging field. Toxicol Pathol. 2008;36:21–9.
  • Hosseinkhani H, Tabata Y. Self assembly of DNA nanoparticles with polycations for the delivery of genetic materials into cells. J Nanosci Nanotechnol. 2006;6:2320–8.
  • Orlando SJ, Santiago Y, DeKelver RC, Freyvert Y, Boydston EA, Moehle EA, . Zinc-finger nuclease-driven targeted integration into mammalian genomes using donors with limited chromosomal homology. Nucleic Acids Res. 2010;38:e152.
  • Benabdallah BF, Allard E, Yao S, Friedman G, Gregory PD, Eliopoulos N, . Targeted gene addition to human mesenchymal stromal cells as a cell-based plasma-soluble protein delivery platform. Cytotherapy. 2010;12:394–9.
  • Papapetrou EP, Lee G, Malani N, Setty M, Riviere I, Tirunagari LM, . Genomic safe harbors permit high beta-globin transgene expression in thalassemia induced pluripotent stem cells. Nat Biotechnol. 2011;29:73–8.
  • Gersbach CA, Gaj T, Gordley RM, Mercer AC, Barbas CF 3rd. Targeted plasmid integration into the human genome by an engineered zinc-finger recombinase. Nucleic Acids Res. 2011;39:7868–78.
  • Russom D, Ahmed A, Gonzalez N, Alvarnas J, DiGiusto D. Implementation of a configurable laboratory information management system for use in cellular process development and manufacturing. Cytotherapy. 2012;14:114–21.
  • Trounson A, Dewitt ND. Stem cell biology: towards the reality of cell therapeutics. Nature Cell Biology. 2012; 14:331.
  • Trounson A, DeWitt ND, Feigal EG. The alpha stem cell clinic: a model for evaluating and delivering stem cell-based therapies. Stem Cells Trans Med. 2012;1:9–14.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.