35
Views
1
CrossRef citations to date
0
Altmetric
REVIEW

Preparing clinical grade Ag-specific T cells for adoptive immunotherapy trials

, PhD & , PhD , MD
Pages 613-629 | Published online: 01 Sep 2009

References

  • Powell DJ, Jr, Dudley ME, Hogan KA, et al. Adoptive transfer of vaccine-induced peripheral blood mononuclear cells to patients with metastatic melanoma following lymphodepletion. J Immunol 2006; 177: 6527–39
  • Kershaw MH, Westwood JA, Parker LL, et al. A phase I study on adoptive immunotherapy using gene-modified T-cells for ovarian cancer. Clin Cancer Res 2006; 12: 6106–15
  • Zhou J, Dudley ME, Rosenberg SA, et al. Selective growth, in vitro and in vivo, of individual T cell clones from tumor-infiltrating lymphocytes obtained from patients with melanoma. J Immunol 2004; 173: 7622–29
  • Hami LS, Green C, Leshinsky N, et al. GMP production and testing of Xcellerated T-cells for the treatment of patients with CLL. Cytotherapy 2004; 6: 554–62
  • Rapoport AP, Stadtmauer EA, Aqui N, et al. Restoration of immunity in lymphopenic individuals with cancer by vaccination and adoptive T-cell transfer. Nat Med 2005; 11: 1230–7
  • Zhou J, Dudley ME, Rosenberg SA, et al. Persistence of multiple tumor-specific T-cell clones is associated with complete tumor regression in a melanoma patient receiving adoptive cell transfer therapy. J Immunother 2005; 28: 53–62
  • Dudley ME, Wunderlich Jr, Yang JC, et al. Adoptive cell transfer therapy following non-myeloablative but lymphodepleting chemotherapy for the treatment of patients with refractory metastatic melanoma. J Clin Oncol 2005; 23: 2346–57
  • Rosenberg SA, Dudley ME. Cancer regression in patients with metastatic melanoma after the transfer of autologous antitumor lymphocytes. Proc Natl Acad Sci USA 2004; 101(Suppl 2)14639–45
  • Dudley ME, Wunderlich J, Nishimura MI, et al. Adoptive transfer of cloned melanoma-reactive T lymphocytes for the treatment of patients with metastatic melanoma. J Immunother 2001; 24: 363–73
  • Dudley ME, Wunderlich JR, Robbins PF, et al. Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes. Science 2002; 298: 850–4
  • Powell DJ, Jr, Dudley ME, Robbins PF. Transition of late-stage effector T-cells to CD27+ CD28+ tumor-reactive effector memory T-cells in humans after adoptive cell transfer therapy. Blood 2005; 105: 241–50
  • Gottschalk S, Heslop HE, Rooney CE. Adoptive immunotherapy for EBV-associated malignancies. Leuk Lymphoma 2005; 46: 1–10
  • Perruccio K, Tosti A, Burchielli E, et al. Transferring functional immune responses to pathogens after haploidentical hematopoietic transplantation. Blood 2005; 106: 4397–406
  • Cobbold M, Khan N, Pourgheysali B, et al. Adoptive transfer of cytomegalovirus-specific CTL to stem cell transplant patients after selection by HLA-peptide tetramers. J Exp Med 2005; 202: 379–86
  • Rauser G, Einsele H, Sinzger C, et al. Rapid generation of combined CMV-specific CD4+ and CD8+ T-cell lines for adoptive transfer into recipients of allogeneic stem cell transplants. Blood 2004; 103: 3565–72
  • Rooney CM, Smith CA, Ng CY, et al. Infusion of cytotoxic T-cells for the prevention and treatment of Epstein–Barr virus-induced lymphoma in allogeneic transplant recipients. Blood 1998; 92: 1549–55
  • Comoli P, Labirio M, Basso S, et al. Infusion of autologous Epstein–Barr virus (EBV)-specific cytotoxic T-cells for prevention of EBV-related lymphoproliferative disorder in solid organ transplant recipients with evidence of active virus replication. Blood 2002; 99: 2592–8
  • Walter EA, Grenberg PD, Gilbert MJ, et al. 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
  • Rossig C, Bollard CM, Calonge MJ, et al. Epstein–Barr virus-specific human T lymphocytes expressing antitumor chimeric T-cell receptors: potential for improved immunotherapy. Blood 2002; 99: 2009–16
  • Rizzuto GA, Wolchok JD. Persistence makes perfect: the benefits of IL-2 in adoptive immunotherapy. Cytotherapy 2005; 7: 391–92
  • Atkins MB. Cytokine-based therapy and biochemotherapy for advanced melanoma. Clin Cancer Res 2006; 12: S2353–8
  • Yee C, Thompson JA, Byrd D, et al. Adoptive T cell therapy using antigen-specific CD8+ T cell clones for the treatment of patients with metastatic melanoma: in vivo persistence, migration, and antitumor effect of transferred T-cells. Proc Natl Acad Sci USA 2002; 99: 16168–73
  • Singh H, Serrano LM, Pfeiffer T, et al. Combining adoptive cellular and immunocytokine therapies to improve treatment of B-lineage malignancy. Cancer Res 2007; 67: 2872–80
  • Wherry EJ, Teichgraber V, Becker TC, et al. Lineage relationship and protective immunity of memory CD8 T cell subsets. Nat Immunol 2003; 4: 225–34
  • Klebanoff CA, Gattinoni L, Torabi-Parizi P, et al. 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
  • Liu Z, Fan H, Wu Y, et al. Potent in vivo anti-tumor activity of isolated CD62L(low) lymph node cells sensitized in vivo with tumor lysate-pulsed DC-based vaccines. Cytotherapy 2005; 7: 353–62
  • Zhang Y, Joe G, Hexner E, et al. Host-reactive CD8+ memory stem cells in graft-versus-host disease. Nat Med 2005; 11: 1299–305
  • Zhang Y, Joe G, Hexner E, et al. Alloreactive memory T-cells are responsible for the persistence of graft-versus-host disease. J Immunol 2005; 174: 3051–8
  • Berger, C, Jensen, MC, Riddell, SR. Abstract 866. Blood 2006;108:
  • Cooper LJ, Kalos M, Lewinsohn DA, et al. 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, Wunderlich JR, et al. Cancer regression in patients after transfer of genetically engineered lymphocytes. Science 2006; 314: 126–9
  • van der Veken LT, Hoogeboom M, de Paus RA, et al. HLA class II restricted T-cell receptor gene transfer generates CD4+ T-cells with helper activity as well as cytotoxic capacity. Gene Ther 2005; 12: 1686–95
  • Scholten KB, Schreurs MW, Ruizendaal JJ, et al. Preservation and redirection of HPV16E7-specific T cell receptors for immunotherapy of cervical cancer. Clin Immunol 2005; 114: 119–29
  • Roszkowski JJ, Lyons GE, Kast WM, et al. Simultaneous generation of CD8+ and CD4+ melanoma-reactive T-cells by retroviral-mediated transfer of a single T-cell receptor. Cancer Res 2005; 65: 1570–6
  • Kahlon KS, Brown C, Cooper LJ, et al. Specific recognition and killing of glioblastoma multiforme by interleukin 13-zetakine redirected cytolytic T-cells. Cancer Res 2004; 64: 9160–6
  • Jensen MC, Cooper LJ, Wu AM, et al. Engineered CD20-specific primary human cytotoxic T lymphocytes for targeting B-cell malignancy. Cytotherapy 2003; 5: 131–8
  • Cooper LJ, Topp MS, Serrano M, et al. T-cell clones can be rendered specific for CD19: toward the selective augmentation of the graft-versus-B-lineage leukemia effect. Blood 2003; 101: 1637–44
  • Ahmad M, Rees RC, Ali SA. Escape from immunotherapy: possible mechanisms that influence tumor regression/progression. Cancer Immunol Immunother 2004; 53: 844–54
  • Willemsen RA, Weijtens ME, Ronteltap C, et al. Grafting primary human T lymphocytes with cancer-specific chimeric single chain and two chain TCR. Gene Ther 2000; 7: 1369–77
  • Kuball J, Dossett ML, Wolfl M, et al. Facilitating matched pairing and expression of TCR chains introduced into human T cells. Blood 2007; 109: 2331–8
  • Cohen CJ, Li YF, El-Gamil M, et al. Enhanced antitumor activity of T cells engineered to express T-cell receptors with a second disulfide bond. Cancer Res 2007; 67: 3898–903
  • Cohen CJ, Zhao Y, Zheng Z, et al. Enhanced antitumor activity of murine–human hybrid T-cell receptor (TCR) in human lymphocytes is associated with improved pairing and TCR/CD3 stability. Cancer Res 2006; 66: 8878–86
  • Cooper LJ, Kalos M, DiGiusto D, et al. T cell genetic modification for re-directed tumor recognition. Cancer Chemother Biol Resp Modifiers 2005; 22: 293–324
  • Jensen MC, Popplewell L, DiGiusto D, et al. A first-in-human clinical trial of adoptive therapy using CD19-specific chimeric antigen receptor re-directed T cells for recurrent/refractory follicular lymphoma. Mol Ther 2007; 15: S142
  • Kershaw MH, Westwood JA, Parker LL, et al. A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer. Clin Cancer Res 2006; 12: 6106–15
  • Park JR, Digiusto DL, Slovak M, et al. Adoptive transfer of chimeric antigen receptor re-directed cytolytic T lymphocyte clones in patients with neuroblastoma. Mol Ther 2007; 15: 825–33
  • Lamers CH, Sleijfer S, Vulto AG, et al. Treatment of metastatic renal cell carcinoma with autologous T-lymphocytes genetically retargeted against carbonic anhydrase IX: first clinical experience. J Clin Oncol 2006; 24: e20–2
  • Lamers, CH, Langeveld, SC, Groot-van Ruijven, CM, , et al. Gene-modified T cells for adoptive immunotherapy of renal cell cancer maintain transgene-specific immune functions in vivo. Cancer Immunol Immunother 2007, May 4; [Epub ahead of print].
  • Walker RE, Bechtel CM, Natarajan V, et al. Long-term in vivo survival of receptor-modified syngeneic T cells in patients with human immunodeficiency virus infection. Blood 2000; 96: 467–74
  • Mitsuyasu RT, Anton PA, Deeks SG, et al. Prolonged survival and tissue trafficking following adoptive transfer of CD4zeta gene-modified autologous CD4(+) and CD8(+) T cells in human immunodeficiency virus-infected subjects. Blood 2000; 96: 785–93
  • Eshhar Z, Waks T, Gross G, et al. 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
  • Peggs KS, Allison JP. Co-stimulatory pathways in lymphocyte regulation: the immunoglobulin superfamily. Br J Haematol 2005; 130: 809–24
  • Altvater B, Pscherer S, Landmejer S, et al. CD28 co-stimulation via tumour-specific chimaeric receptors induces an incomplete activation response in Epstein–Barr virus-specific effector memory T-cells. Clin Exp Immunol 2006; 144: 447–57
  • Maher J, Brentjens RJ, Gunset G, et al. Human T-lymphocyte cytotoxicity and proliferation directed by a single chimeric TCRzeta/CD28 receptor. Nat Biotechnol 2002; 20: 70–5
  • Loskog A, Giandomenico V, Rossig C, et al. Addition of the CD28 signaling domain to chimeric T-cell receptors enhances chimeric T-cell resistance to T regulatory cells. Leukemia 2006; 20: 1819–28
  • Kowolik CM, Topp MS, Gonzalez S, et al. 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
  • Imai C, Mihara K, Andreansky M, et al. Chimeric receptors with 4-1BB signaling capacity provoke potent cytotoxicity against acute lymphoblastic leukemia. Leukemia 2004; 18: 676–84
  • Westwood JA, Smyth MJ, Teng MW, et al. Adoptive transfer of T-cells modified with a humanized chimeric receptor gene inhibits growth of Lewis-Y-expressing tumors in mice. Proc Natl Acad Sci USA 2005; 102: 19051–6
  • Berger C, Flowers ME, Warren EH, et al. 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
  • Kornblau SM, Aycox PG, Stephens C, et al. Control of graft-versus-host disease with maintenance of the graft-versus-leukemia effect in a murine allogeneic transplant model using retrovirally transduced murine suicidal lymphocytes. Exp Hematol 2007; 35: 842–53
  • Traversari C, Marktel S, Magnani Z, et al. The potential immunogenicity of the TK suicide gene does not prevent full clinical benefit associated with the use of TK-transduced donor lymphocytes in HSCT for hematologic malignancies. Blood 2007; 109: 4708–15
  • Ciceri F, Bonini C, Marktel S, et al. Antitumor effects of HSV-TK-engineered donor lymphocytes after allogeneic stem-cell transplantation. Blood 2007; 109: 4698–707
  • Straathof KC, Pule MA, Yotnda P, et al. An inducible caspase 9 safety switch for T-cell therapy. Blood 2005; 105: 4247–54
  • Sato T, Neschadim A, Konrad M, et al. Engineered human tmpk/AZT as a novel enzyme/prodrug axis for suicide gene therapy. Mol Ther 2007; 15: 962–70
  • Hacein-Bey-Abina S, Von Kalle C, Schmidt M, et al. LMO2-associated clonal T cell proliferation in two patients after gene therapy for SCID-X1. Science 2003; 302: 415–19
  • Ivics Z, Hackett PB, Plasterk RH, Izsvak Z. Molecular reconstruction of sleeping beauty, a Tc1-like transposon from fish, and its transposition in human cells. Cell 1997; 91: 501–10
  • Geurts AM, Hackett CS, Bell JB, et al. Structure-based prediction of insertion-site preferences of transposons into chromosomes. Nucleic Acids Res 2006; 34: 2803–11
  • Frommolt R, Rohrbach F, Theobald M. Sleeping Beauty transposon system: future trend in T-cell-based gene therapies?. Future Oncol 2006; 2: 345–9
  • Huang X, Wilber AC, Bao L, et al. Stable gene transfer and expression in human primary T cells by the Sleeping Beauty transposon system. Blood 2006; 107: 483–91
  • Hackett PB. Integrating DNA vectors for gene therapy. Mol Ther 2007; 15: 10–2
  • Lemarie C, Calmels B, Malenfant C, et al. Clinical experience with the delivery of thawed and washed autologous blood cells, with an automated closed fluid management device: CytoMate. Transfusion 2005; 45: 737–42
  • Lemarie C, Sugaye R, Kaur I, et al. Purification of monocytes from cryopreserved mobilized apheresis products by elutriation with the Elutra device. J Immunol Methods 2007; 318: 30–6
  • Huntenburg CC, Kunkel LA, Schneidkraut MJ. CD34+ cell engraftment, ex vivo expansion, and malignant cell depletion following immunomagnetic selection. J Hematother 1998; 07: 175–83
  • McNiece IK, Stoney GB, Kern BP, et al. CD34+ cell selection from frozen cord blood products using the Isolex 300i and CliniMACS CD34 selection devices. J Hematother 1998; 7: 457–61
  • Orchard PJ, Blazer BR, Burger S, et al. 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
  • Watts MJ, Somervaille TC, Ings SJ, et al. Variable product purity and functional capacity after CD34 selection: a direct comparison of the CliniMACS (v2.1) and Isolex 300i (v2.5) clinical scale devices. Br J Haematol 2002; 118: 117–23
  • Williams SF, Lee WJ, Bender JG, et al. Selection and expansion of peripheral blood CD34+ cells in autologous stem cell transplantation for breast cancer. Blood 1996; 87: 1687–91
  • Jayasinghe SM, Wunderlich J, McKee A, et al. Sterile and disposable fluidic subsystem suitable for clinical high speed fluorescence-activated cell sorting. Cytometry B Clin Cytom 2006; 70: 344–54
  • Lennartz K, Lu M, Flasshove M, Moritz T, Kirstein U. Improving the biosafety of cell sorting by adaptation of a cell sorting system to a biosafety cabinet. Cytometry A 2005; 66: 119–127
  • Oberyszyn AS, Robertson FM. Novel rapid method for visualization of extent and location of aerosol contamination during high-speed sorting of potentially biohazardous samples. Cytometry 2001; 43: 217–22
  • Perfetto SP, Ambrozak DR, Roederer M, Koup RA. Viable infectious cell sorting in a BSL-3 facility. Methods Mol Biol 2004; 263: 419–24
  • Lennartz K, Lu M, Flasshove M, et al. Improving the biosafety of cell sorting by adaptation of a cell sorting system to a biosafety cabinet. Cytometry A 2005; 66: 119–27
  • Gentry T, Foster S, Winstead L, et al. Simultaneous isolation of human BM hematopoietic, endothelial and mesenchymal progenitor cells by flow sorting based on aldehyde dehydrogenase activity: implications for cell therapy. Cytotherapy 2007; 9: 259–74
  • Nagano M, Yamashita T, Hamada H, et al. Identification of functional endothelial progenitor cells suitable for the treatment of ischemic tissue using human umbilical cord blood. Blood 2007; 110: 151–60
  • Storms RW, Green PD, Safford KM, et al. Distinct hematopoietic progenitor compartments are delineated by the expression of aldehyde dehydrogenase and CD34. Blood 2005; 106: 95–102
  • Hess DA, Wirthlin L, Craft TP, et al. Selection based on CD133 and high aldehyde dehydrogenase activity isolates long-term reconstituting human hematopoietic stem cells. Blood 2006; 107: 2162–9
  • Riddell SR, Watanabe KS, Goodrich JM, et al. Restoration of viral immunity in immunodeficient humans by the adoptive transfer of T cell clones. Science 1992; 257: 238–41
  • Cooper LJ, Ausubel L, Gutierrez M, et al. Manufacturing of gene-modified cytotoxic T lymphocytes for autologous cellular therapy for lymphoma. Cytotherapy 2006; 8: 105–17
  • Numbenjapon T, Serrano LM, Singh H, et al. Characterization of an artificial antigen-presenting cell to propagate cytolytic CD19-specific T-cells. Leukemia 2006; 20: 1889–92
  • Oelke M, Maus MV, Didiano D, et al. Ex vivo induction and expansion of antigen-specific cytotoxic T-cells by HLA-Ig-coated artificial antigen-presenting cells. Nat Med 2003; 9: 619–24
  • Maus MV, Riley JL, Kwok WW, et al. HLA tetramer-based artificial antigen-presenting cells for stimulation of CD4+ T-cells. Clin Immunol 2003; 106: 16–22
  • Thomas AK, Maus MV, Shalaby WS, et al. A cell-based artificial antigen-presenting cell coated with anti-CD3 and CD28 antibodies enables rapid expansion and long-term growth of CD4 T lymphocytes. Clin Immunol 2002; 105: 259–72
  • Maus MV, Thomas AK, Leonard DG, et al. Ex vivo expansion of polyclonal and antigen-specific cytotoxic T lymphocytes by artificial APCs expressing ligands for the T-cell receptor, CD28 and 4-1BB. Nat Biotechnol 2002; 20: 143–8
  • Garlie NK, LeFever AV, Siebenlist RE, et al. T-cells coactivated with immobilized anti-CD3 and anti-CD28 as potential immunotherapy for cancer. J Immunother 1999; 22: 336–45
  • Levine BL, Cotte J, Small CC, et al. Large-scale production of CD4+ T-cells from HIV-1-infected donors after CD3/CD28 costimulation. J Hematother 1998; 7: 437–48
  • Trickett AE, Kwan YL, Cameron B, et al. Ex vivo expansion of functional T lymphocytes from HIV-infected individuals. J Immunol Methods 2002; 262: 71–83
  • Bernstein WB, Cox JH, Aronson NE, et al. Immune reconstitution following autologous transfers of CD3/CD28 stimulated CD4(+) T-cells to HIV-infected persons. Clin Immunol 2004; 111: 262–74
  • Laport GG, Levine BL, Stadtmauer EA, et al. Adoptive transfer of costimulated T-cells induces lymphocytosis in patients with relapsed/refractory non-Hodgkin lymphoma following CD34 + -selected hematopoietic cell transplantation. Blood 2003; 102: 2004–13
  • Porter DL, Levine BL, Bunin N, et al. A phase 1 trial of donor lymphocyte infusions expanded and activated ex vivo via CD3/CD28 costimulation. Blood 2006; 107: 1325–31
  • Santegoets SJ, Schreurs MW, Masterson AJ, et al. In vitro priming of tumor-specific cytotoxic T lymphocytes using allogeneic dendritic cells derived from the human MUTZ-3 cell line. Cancer Immunol Immunother 2006; 55: 1480–90
  • Michiels A, Breckpot K, Corthals J, et al. Induction of antigen-specific CD8+ cytotoxic T-cells by dendritic cells co-electroporated with a dsRNA analogue and tumor antigen mRNA. Gene Ther 2006; 13: 1027–36
  • Mackensen A, Meidenbauer N, Vogl S, et al. Phase I study of adoptive T-cell therapy using antigen-specific CD8+ T-cells for the treatment of patients with metastatic melanoma. J Clin Oncol 2006; 24: 5060–9
  • Trivedi D, Wiliams RY, O'Reilly RJ, et al. Generation of CMV-specific T lymphocytes using protein-spanning pools of pp65-derived overlapping pentadecapeptides for adoptive immunotherapy. Blood 2005; 105: 2793–801
  • Peng JC, Thomas R, Nielsen LK. Generation and maturation of dendritic cells for clinical application under serum-free conditions. J Immunother 2005; 28: 599–609
  • Elias M, van Zanten J, Hospers GA, et al. Closed system generation of dendritic cells from a single blood volume for clinical application in immunotherapy. J Clin Apher 2005; 20: 197–207
  • Adamson L, Palmborg A, Svensson A, et al. Development of a technology platform for large-scale clinical grade production of DC. Cytotherapy 2004; 6: 363–71
  • Sorg RV, Ozcan Z, Brefort T, et al. Clinical-scale generation of dendritic cells in a closed system. J Immunother 2003; 26: 374–83
  • Thurner B, Roder C, Dieckmann D, et al. Generation of large numbers of fully mature and stable dendritic cells from leukapheresis products for clinical application. J Immunol Methods 1999; 223: 1–15
  • Wu JY, Ernstoff MS, Hill JM, et al. Ex vivo expansion of non-MHC-restricted cytotoxic effector cells as adoptive immunotherapy for myeloma. Cytotherapy 2006; 8: 141–8
  • Klingemann HG, Martinson J. Ex vivo expansion of natural killer cells for clinical applications. Cytotherapy 2004; 6: 15–22
  • Tam YK, Martinson JA, Doligosa K, et al. Ex vivo expansion of the highly cytotoxic human natural killer-92 cell-line under current good manufacturing practice conditions for clinical adoptive cellular immunotherapy. Cytotherapy 2003; 5: 259–72
  • Robinson KL, Ayello J, Hughes R, et al. Ex vivo expansion, maturation, and activation of umbilical cord blood-derived T lymphocytes with IL-2, IL-12, anti-CD3, and IL-7. Potential for adoptive cellular immunotherapy post-umbilical cord blood transplantation. Exp Hematol 2002; 30: 245–51
  • Warncke M, Dodero A, Dierbach H, et al. Murine dendritic cells generated under serum-free conditions have a mature phenotype and efficiently induce primary immune responses. J Immunol Methods 2006; 310: 1–11
  • Sombroek CC, Stam AG, Masterson AJ, et al. Prostanoids play a major role in the primary tumor-induced inhibition of dendritic cell differentiation. J Immunol 2002; 168: 4333–43
  • Upham JW, Holt BJ, Baron-Hay MJ, et al. Inhalant allergen-specific T-cell reactivity is detectable in close to 100% of atopic and normal individuals: covert responses are unmasked by serum-free medium. Clin Exp Allergy 1995; 25: 634–42
  • Carlens S, Gilljam M, Remberger M, et al. Ex vivo T lymphocyte expansion for retroviral transduction: influence of serum-free media on variations in cell expansion rates and lymphocyte subset distribution. Exp Hematol 2000; 28: 1137–46
  • Waldmann TA, Dubois S, Tagaya Y. Contrasting roles of IL-2 and IL-15 in the life and death of lymphocytes: implications for immunotherapy. Immunity 2001; 14: 105–10
  • Bulfone-PauS S, Bulanova E, Pohl T, et al. Death deflected: IL-15 inhibits TNF-alpha-mediated apoptosis in fibroblasts by TRAF2 recruitment to the IL-15Ralpha chain. FASEB J 1999; 13: 1575–85
  • Mueller YM, Bojczuk PM, Halstead ES, et al. IL-15 enhances survival and function of HIV-specific CD8+ T-cells. Blood 2003; 101: 1024–9
  • Mueller YM, Makar V, Bojczuk PM, et al. IL-15 enhances the function and inhibits CD95/Fas-induced apoptosis of human CD4+ and CD8+ effector-memory T-cells. Int Immunol 2003; 15: 49–58
  • Teague RM, Sather BD, Sacks JA, et al. Interleukin-15 rescues tolerant CD8+ T-cells for use in adoptive immunotherapy of established tumors. Nat Med 2006; 12: 335–41
  • Peggs KS, Verfuerth S, Pizzey A, et al. Adoptive cellular therapy for early cytomegalovirus infection after allogeneic stem-cell transplantation with virus-specific T-cell lines. Lancet 2003; 362: 1375–7
  • Leen AM, Myers GD, Sili U, et al. Monoculture-derived T lymphocytes specific for multiple viruses expand and produce clinically relevant effects in immunocompromised individuals. Nat Med 2006; 12: 1160–6
  • Straathof KC, Bollard CM, Popat U, et al. Treatment of nasopharyngeal carcinoma with Epstein–Barr virus-specific T lymphocytes. Blood 2005; 105: 1898–904
  • Office of Biotechnology Activities, published by National Institutes of Health, http://www4.od.nih.gov/oba/, Page Updated: 05/23/2006, page visited by me 09-21-2007.
  • Recombinant DNA Advisory Committee, National Institutes of Health, http://www4.od.nih.gov/oba/rac/aboutrdagt.htm, page visited by me 09-21-2007.
  • Tey SK, Brenner MK. The continuing contribution of gene marking to cell and gene therapy. Mol Ther 2007; 15: 666–76
  • Bonini, C, Bondanza, A, Perna, SK, , et al. The suicide gene therapy challenge: how to improve a successful gene therapy approach. Mol Ther. 2007;15:1248–1252.
  • Schroder AR, Shinn P, Chen H, et al. HIV-1 integration in the human genome favors active genes and local hotspots. Cell 2002; 110: 521–9
  • Wu X, Li Y, Crise B, Burgess SM. Transcription start regions in the human genome are favored targets for MLV integration. Science 2003; 300: 1749–51
  • Dudley ME, Wunderlich JR, Yang JC, et al. Adoptive cell transfer therapy following non-myeloablative but lymphodepleting chemotherapy for the treatment of patients with refractory metastatic melanoma. J Clin Oncol 2005; 23: 2346–57
  • Atkins MB, Kunkel L, Sznol M, et al. High-dose recombinant interleukin-2 therapy in patients with metastatic melanoma: long-term survival update. Cancer J Sci Am 2000; 6(Suppl 1)S11–14
  • Mackensen A, Meidenbauer N, Vogl S, et al. Phase I study of adoptive T-cell therapy using antigen-specific CD8+ T-cells for the treatment of patients with metastatic melanoma. J Clin Oncol 2006; 24: 5060–9
  • Miller JS, Soignier Y, Panoskaltsis-Mortari A, et al. Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood 2005; 105: 3051–7

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.