Skip to main content

Adoptive T Cell Transfer

  • Chapter
  • First Online:
Cancer Immunotherapy

Abstract

The clinical use of adoptively-transferred T or NK lymphocytes with anti-tumor activity is gaining in popularity as reports of success accumulate. High specific activity with minimal off target effects contribute to a class of therapy with minimal toxicity that is transformative for cancer patients, who with their physicians have come to accept severe short and long term toxicities as the cost of a frequently small hope of cure or extension of life. However, the successes of cell therapies are as yet in the minority and scientists are developing and testing strategies to improve the function and persistence of adoptively transferred lymphocytes in the face of multiple, potent immune evasion strategies used by tumors and their accessory cells to coexist with an intact immune response. These strategies include combination with chemotherapies and/or radiation and genetic modifications that improve T cell function and tumor targeting. However many of these strategies come with a price and re-introduce toxicities including death in rare cases. This chapter will outline the development of tumor-specific lymphocyte therapies, highlighting successes and difficulties and discussing potential ways forward in this exciting field.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 129.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 169.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 169.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Arstila TP et al (1999) A direct estimate of the human alphabeta T cell receptor diversity. Science 286:958–961

    Article  PubMed  CAS  Google Scholar 

  2. Penn I, Hammond W, Brettschneider L, Starzl TE (1969) Malignant lymphomas in transplantation patients. Transplant Proc 1:106–112

    PubMed  CAS  Google Scholar 

  3. Shapiro RS et al (1988) Epstein-Barr virus associated B cell lymphoproliferative disorders following bone marrow transplantation. Blood 71:1234–1243

    PubMed  CAS  Google Scholar 

  4. Swerdlow SH, Webber SA, Chadbum A, & Ferry,J. Post transplant lymphoproliferative disorders in Classification of Tumours of Hematopoietic and Lymphoid Tissues (eds. Swerdlow SH et al.) 342–349 (International Agency for Research on Cancer, Lyon, France, 2008).

    Google Scholar 

  5. Thomas JA, Crawford DH (1989) Epstein-Barr virus associated B-cell lymphomas in AIDS and after organ transplantation. Lancet 1:1075–1076

    Article  PubMed  CAS  Google Scholar 

  6. Doubrovina E et al (2012) Adoptive immunotherapy with unselected or EBV-specific T cells for biopsy proven EBV+ lymphomas after allogeneic hematopoietic cell transplants. Blood 119:2644–2656

    Article  PubMed  CAS  Google Scholar 

  7. Landais E, Saulquin X, Bonneville M, Houssaint E (2005) Long-term MHC class II presentation of the EBV lytic protein BHRF1 by EBV latently infected b cells following capture of BHRF1 antigen. J Immunol 175:7939–7946

    PubMed  CAS  Google Scholar 

  8. Martorelli D et al (2008) Spontaneous T cell responses to Epstein-Barr virus-encoded BARF1 protein and derived peptides in patients with nasopharyngeal carcinoma: bases for improved immunotherapy. Int J Cancer 123:1100–1107

    Article  PubMed  CAS  Google Scholar 

  9. Kelly GL et al (2009) An Epstein-Barr virus anti-apoptotic protein constitutively expressed in transformed cells and implicated in burkitt lymphomagenesis: the Wp/BHRF1 link. PLoS Pathog 5:e141

    Article  Google Scholar 

  10. Rickinson AB & Kieff E Epstein-Barr Virus in Fields Virology (eds. Knipe DM & Howley PM) 2575–2628 (Lippincott Williams & Williams, Philadelphia, 2001).

    Google Scholar 

  11. Rooney CM et al (1995) Use of gene-modified virus-specific T lymphocytes to control Epstein-Barr virus-related lymphoproliferation. Lancet 345:9–13

    Article  PubMed  CAS  Google Scholar 

  12. Heslop HE et al (1996) Long-term restoration of immunity against Epstein-Barr virus infection by adoptive transfer of gene-modified virus-specific T lymphocytes. Nat Med 2:551–555

    Article  PubMed  CAS  Google Scholar 

  13. Rooney CM et al (1998) Infusion of cytotoxic T cells for the prevention and treatment of Epstein-Barr virus-induced lymphoma in allogeneic transplant recipients. Blood 92:1549–1555

    PubMed  CAS  Google Scholar 

  14. Heslop HE, Slobod KS, Pule MA, Hale GA, Rousseau A, Smith CA, Bollard CM, Liu H, Wu MF, Rochester RJ, Amrolia PJ, Hurwitz JL, Brenner MK, Rooney CM: Long-term outcome of EBV-specific T-cell infusions to prevent or treat EBV-related lymphoproliferative disease in transplant recipients. Blood 2010, 115:925–935. Plenary Paper. PMCID: PMC2817637

    Google Scholar 

  15. O'Reilly RJ et al (1998) Adoptive immunotherapy for Epstein-Barr virus-associated lymphoproliferative disorders complicating marrow allografts. Springer Semin Immunopathol 20:455–491

    Article  PubMed  Google Scholar 

  16. Gustafsson A et al (2000) Epstein-Barr virus (EBV) load in bone marrow transplant recipients at risk to develop posttransplant lymphoproliferative disease: prophylactic infusion of EBV-specific cytotoxic T cells. Blood 95:807–814

    PubMed  CAS  Google Scholar 

  17. Haque T et al (2007) Allogeneic cytotoxic T cell therapy for EBV-positive posttransplantation lymphoproliferative disease: results of a phase 2 multicenter clinical trial. Blood 110:1123–1131

    Article  PubMed  CAS  Google Scholar 

  18. Fujita Y et al (2008) Exploiting cytokine secretion to rapidly produce multivirus-specific T cells for adoptive immunotherapy. J Immunother 31:665–674

    Article  PubMed  CAS  Google Scholar 

  19. Moosmann A et al (2010) Effective and long-term control of EBV PTLD after transfer of peptide-selected T cells. Blood 115:2960–2970

    Article  PubMed  CAS  Google Scholar 

  20. Bodinier M et al (2000) Efficient detection and immunomagnetic sorting of specific T cells using multimers of MHC class I and peptide with reduced CD8 binding. Nat Med 6:707–710

    Article  PubMed  CAS  Google Scholar 

  21. Gerdemann U et al (2009) Nucleofection of DCs to generate Multivirus-specific T cells for prevention or treatment of viral infections in the immunocompromised host. Mol Ther 17:1616–1625

    Article  PubMed  CAS  Google Scholar 

  22. Mackinnon S, Thomson K, Verfuerth S, Peggs K, Lowdell M (2008) Adoptive cellular therapy for cytomegalovirus infection following allogeneic stem cell transplantation using virus-specific T cells. Blood Cells Mol Dis 40:63–67

    Article  PubMed  CAS  Google Scholar 

  23. Cobbold M et al (2005) Adoptive transfer of cytomegalovirus-specific CTL to stem cell transplant patients after selection by HLA-peptide tetramers. J Exp Med 202:379–386

    Article  PubMed  CAS  Google Scholar 

  24. Chan MK, Huang DW, Ho YH, Lee JC (1989) Detection of Epstein-Barr virus-associated antigen in fine needle aspiration smears from cervical lymph nodes in the diagnosis of nasopharyngeal carcinoma. Acta Cytol 33:351–354

    PubMed  CAS  Google Scholar 

  25. Pallesen G, Hamilton-Dutoit SJ, Rowe M, Young LS (1991) Expression of Epstein-Barr virus latent gene products in tumour cells of Hodgkin's disease. Lancet 337:320–322

    Article  PubMed  CAS  Google Scholar 

  26. Hislop AD, Taylor GS, Sauce D, Rickinson AB (2007) Cellular responses to viral infection in humans: lessons from Epstein-Barr virus. Annu Rev Immunol 25:587–617

    Article  PubMed  CAS  Google Scholar 

  27. Decaussin G, Sbih-Lammali F, de Turenne-Tessier M, Bouguermouh A, Ooka T (2000) Expression of BARF1 gene encoded by Epstein-Barr virus in nasopharyngeal carcinoma biopsies. Cancer Res 60:5584–5588

    PubMed  CAS  Google Scholar 

  28. Steigerwald-Mullen PM, Klein G, Kurilla MG, Masucci MG (1995) Inhibition of antigen processing by the internal repeat region of the Epstein-Barr virus nuclear antigen-1. Nature 375:685–688

    Article  PubMed  Google Scholar 

  29. Voo KS et al (2004) Evidence for the presentation of major histocompatibility complex class I-restricted Epstein-Barr virus nuclear antigen 1 peptides to CD8+ T lymphocytes. J Exp Med 199:459–470

    Article  PubMed  CAS  Google Scholar 

  30. Bollard CM et al (2004) Cytotoxic T Lymphocyte Therapy for Epstein-Barr Virus+Hodgkin's Disease. J Exp Med 200:1623–1633

    Article  PubMed  CAS  Google Scholar 

  31. Chua D et al (2001) Adoptive transfer of autologous Epstein-Barr virus-specific cytotoxic T cells for nasopharyngeal carcinoma. Int J Cancer 94(1):73–80

    Google Scholar 

  32. Straathof KC et al (2005) Treatment of nasopharyngeal carcinoma with Epstein-Barr virus-specific T lymphocytes. Blood 105:1898–1904

    Article  PubMed  CAS  Google Scholar 

  33. Louis CU et al (2009) Enhancing the in vivo expansion of adoptively transferred EBV-specific CTL with lymphodepleting CD45 monoclonal antibodies in NPC patients. Blood 113:2442–2450

    Article  PubMed  CAS  Google Scholar 

  34. Bollard CM et al (2007) Complete responses of relapsed lymphoma following genetic modification of tumor-antigen presenting cells and T-lymphocyte transfer. Blood 110:2838–2845

    Article  PubMed  CAS  Google Scholar 

  35. Bollard,C.M. et al. Complete tumor responses in lymphoma patients receiving autologous cytotoxic T lymphocytes targeting Epstein Barr virus (EBV) latent membrane proteins. American Society of Hematology 53rd Annual Meeting 2011. 2011. Ref Type: Abstract

    Google Scholar 

  36. Clemente CG et al (1996) Prognostic value of tumor infiltrating lymphocytes in the vertical growth phase of primary cutaneous melanoma. Cancer 77:1303–1310

    Article  PubMed  CAS  Google Scholar 

  37. Naito Y et al (1998) CD8+ T cells infiltrated within cancer cell nests as a prognostic factor in human colorectal cancer. Cancer Res 58:3491–3494

    PubMed  CAS  Google Scholar 

  38. Sato E et al (2005) Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc Natl Acad Sci USA 102:18538–18543

    Article  PubMed  CAS  Google Scholar 

  39. Muul LM, Spiess PJ, Director EP, Rosenberg SA (1987) Identification of specific cytolytic immune responses against autologous tumor in humans bearing malignant melanoma. J Immunol 138:989–995

    PubMed  CAS  Google Scholar 

  40. Rosenberg SA et al (1994) Treatment of patients with metastatic melanoma with autologous tumor-infiltrating lymphocytes and interleukin 2. J Natl Cancer Inst 86:1159–1166

    Article  PubMed  CAS  Google Scholar 

  41. Dudley ME, Wunderlich JR, Shelton TE, Even J, Rosenberg SA (2003) Generation of tumor-infiltrating lymphocyte cultures for use in adoptive transfer therapy for melanoma patients. J Immunother 26:332–342

    Article  PubMed  Google Scholar 

  42. Rosenberg SA, Dudley ME (2009) Adoptive cell therapy for the treatment of patients with metastatic melanoma. Curr Opin Immunol 21:233–240

    Article  PubMed  CAS  Google Scholar 

  43. Wrzesinski C et al (2010) Increased intensity lymphodepletion enhances tumor treatment efficacy of adoptively transferred tumor-specific T cells. J Immunother 33:1–7

    Article  PubMed  Google Scholar 

  44. Freedman RS et al (1994) Intraperitoneal adoptive immunotherapy of ovarian carcinoma with tumor-infiltrating lymphocytes and low-dose recombinant interleukin-2: a pilot trial. J Immunother Emphasis Tumor Immunol 16:198–210

    Article  PubMed  CAS  Google Scholar 

  45. Fujita K et al (1995) Prolonged disease-free period in patients with advanced epithelial ovarian cancer after adoptive transfer of tumor-infiltrating lymphocytes. Clin Cancer Res 1:501–507

    PubMed  CAS  Google Scholar 

  46. Chekmasova AA, Brentjens RJ (2010) Adoptive T cell immunotherapy strategies for the treatment of patients with ovarian cancer. Discov Med 9:62–70

    PubMed  Google Scholar 

  47. Tran KQ et al (2008) Minimally cultured tumor-infiltrating lymphocytes display optimal characteristics for adoptive cell therapy. J Immunother 31:742–751

    Article  PubMed  CAS  Google Scholar 

  48. Yanagi Y et al (1984) A human T cell-specific cDNA clone encodes a protein having extensive homology to immunoglobulin chains. Nature 308:145–149

    Article  PubMed  CAS  Google Scholar 

  49. Hedrick SM, Cohen DI, Nielsen EA, Davis MM (1984) Isolation of cDNA clones encoding T cell-specific membrane-associated proteins. Nature 308:149–153

    Article  PubMed  CAS  Google Scholar 

  50. Bunnell BA, Muul LM, Donahue RE, Blaese RM, Morgan RA (1995) High-efficiency retroviral-mediated gene transfer into human and nonhuman primate peripheral blood lymphocytes. Proc Natl Acad Sci USA 92:7739–7743

    Article  PubMed  CAS  Google Scholar 

  51. Morgan RA et al (2006) Cancer regression in patients after transfer of genetically engineered lymphocytes. Science 314:126–129

    Article  PubMed  CAS  Google Scholar 

  52. Johnson LA et al (2009) Gene therapy with human and mouse T cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen. Blood 114:535–546

    Article  PubMed  CAS  Google Scholar 

  53. Cooper LJ, Kalos M, Lewinsohn DA, Riddell SR, Greenberg PD (2000) Transfer of specificity for human immunodeficiency virus type 1 into primary human T lymphocytes by introduction of T cell receptor genes. J Virol 74:8207–8212

    Article  PubMed  CAS  Google Scholar 

  54. Rubinstein MP et al (2003) Transfer of TCR genes into mature T cells is accompanied by the maintenance of parental T cell avidity. J Immunol 170:1209–1217

    PubMed  CAS  Google Scholar 

  55. Voss RH et al (2006) Redirection of T cells by delivering a transgenic mouse-derived MDM2 tumor antigen-specific TCR and its humanized derivative is governed by the CD8 coreceptor and affects natural human TCR expression. Immunol Res 34:67–87

    Article  PubMed  CAS  Google Scholar 

  56. Kuball J et al (2007) Facilitating matched pairing and expression of TCR chains introduced into human T cells. Blood 109:2331–2338

    Article  PubMed  CAS  Google Scholar 

  57. Okamoto S et al (2009) Improved expression and reactivity of transduced tumor-specific TCRs in human lymphocytes by specific silencing of endogenous TCR. Cancer Res 69:9003–9011

    Article  PubMed  CAS  Google Scholar 

  58. Natali PG et al (1989) Selective changes in expression of HLA class I polymorphic determinants in human solid tumors. Proc Natl Acad Sci USA 86:6719–6723

    Article  PubMed  CAS  Google Scholar 

  59. Gross G, Gorochov G, Waks T, Eshhar Z (1989) Generation of effector T cells expressing chimeric T cell receptor with antibody type-specificity. Transplant Proc 21:127–130

    PubMed  CAS  Google Scholar 

  60. Eshhar Z, Waks T, Gross G, Schindler DG (1993) 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 90:720–724

    Article  PubMed  CAS  Google Scholar 

  61. Pule MA et al (2008) Virus-specific T cells engineered to coexpress tumor-specific receptors: persistence and antitumor activity in individuals with neuroblastoma. Nat Med 14:1264–1270

    Article  PubMed  CAS  Google Scholar 

  62. Schwartz RH (1992) Costimulation of T lymphocytes: the role of CD28, CTLA-4, and B7/BB1 in interleukin-2 production and immunotherapy. Cell 71:1065–1068

    Article  PubMed  CAS  Google Scholar 

  63. Kobata T, Agematsu K, Kameoka J, Schlossman SF, Morimoto C (1994) CD27 is a signal-transducing molecule involved in CD45RA+naive T cell costimulation. J Immunol 153:5422–5432

    PubMed  CAS  Google Scholar 

  64. Watts TH (2005) TNF/TNFR family members in costimulation of T cell responses. Annu Rev Immunol 23:23–68

    Article  PubMed  CAS  Google Scholar 

  65. Finney HM, Lawson AD, Bebbington CR, Weir AN (1998) Chimeric receptors providing both primary and costimulatory signaling in T cells from a single gene product. J Immunol 161:2791–2797

    PubMed  CAS  Google Scholar 

  66. Maher J, Brentjens RJ, Gunset G, Riviere I, Sadelain M (2002) Human T-lymphocyte cytotoxicity and proliferation directed by a single chimeric TCRzeta /CD28 receptor. Nat Biotechnol 20:70–75

    Article  PubMed  CAS  Google Scholar 

  67. Vera J et al (2006) T lymphocytes redirected against the kappa light chain of human immunoglobulin efficiently kill mature B lymphocyte-derived malignant cells. Blood 108:3890–3897

    Article  PubMed  CAS  Google Scholar 

  68. Kowolik CM et al (2006) CD28 costimulation provided through a CD19-specific chimeric antigen receptor enhances in vivo persistence and antitumor efficacy of adoptively transferred T cells. Cancer Res 66:10995–11004

    Article  PubMed  CAS  Google Scholar 

  69. Savoldo B et al (2011) CD28 costimulation improves expansion and persistence of chimeric antigen receptor-modified T cells in lymphoma patients. J Clin Invest 121:1822–1826

    Article  PubMed  CAS  Google Scholar 

  70. Pule MA et al (2005) A chimeric T cell antigen receptor that augments cytokine release and supports clonal expansion of primary human T cells. Mol Ther 12:933–941

    Article  PubMed  CAS  Google Scholar 

  71. Carpenito C et al (2009) Control of large, established tumor xenografts with genetically retargeted human T cells containing CD28 and CD137 domains. Proc Natl Acad Sci USA 106:3360–3365

    Article  PubMed  CAS  Google Scholar 

  72. Zhong XS, Matsushita M, Plotkin J, Riviere I, Sadelain M (2010) Chimeric antigen receptors combining 4-1BB and CD28 signaling domains augment PI3kinase/AKT/Bcl-XL activation and CD8+ T cell-mediated tumor eradication. Mol Ther 18:413–420

    Article  PubMed  CAS  Google Scholar 

  73. Kalos M et al (2011) T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci Transl Med 3:95ra73

    Article  PubMed  CAS  Google Scholar 

  74. Porter DL, Levine BL, Kalos M, Bagg A, June CH (2011) Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med 365:725–733

    Article  PubMed  CAS  Google Scholar 

  75. Savoldo B et al (2007) Epstein Barr virus specific cytotoxic T lymphocytes expressing the anti-CD30zeta artificial chimeric T cell receptor for immunotherapy of Hodgkin disease. Blood 110:2620–2630

    Article  PubMed  CAS  Google Scholar 

  76. Heslop HE (2010) Safer CARS. Mol Ther 18:661–662

    Article  PubMed  CAS  Google Scholar 

  77. Rossig C, Bollard CM, Nuchtern JG, Rooney CM, Brenner MK (2002) Epstein-Barr virus-specific human T lymphocytes expressing antitumor chimeric T cell receptors: potential for improved immunotherapy. Blood 99:2009–2016

    Article  PubMed  CAS  Google Scholar 

  78. Cooper LJ et al (2005) Enhanced antilymphoma efficacy of CD19-redirected influenza MP1-specific CTLs by cotransfer of T cells modified to present influenza MP1. Blood 105:1622–1631

    Article  PubMed  CAS  Google Scholar 

  79. Leen AM, Rooney CM, Foster AE (2007) Improving T cell therapy for cancer. Annu Rev Immunol 25:243–265

    Article  PubMed  CAS  Google Scholar 

  80. Di SA et al (2011) Inducible apoptosis as a safety switch for adoptive cell therapy. N Engl J Med 365:1673–1683

    Article  Google Scholar 

  81. Craddock JA et al (2010) Enhanced tumor trafficking of GD2 chimeric antigen receptor T cells by expression of the chemokine receptor CCR2b. J Immunother 33:780–788

    Article  PubMed  CAS  Google Scholar 

  82. Vera JF et al (2009) Genetic manipulation of tumor-specific cytotoxic T lymphocytes to restore responsiveness to IL-7. Mol Ther 17:880–888

    Article  PubMed  CAS  Google Scholar 

  83. Hoyos V et al (2010) Engineering CD19-specific T lymphocytes with interleukin-15 and a suicide gene to enhance their anti-lymphoma/leukemia effects and safety. Leukemia 24:1160–1170

    Article  PubMed  CAS  Google Scholar 

  84. Sun J et al (2010) T cells expressing constitutively active Akt resist multiple tumor-associated inhibitory mechanisms. Mol Ther 18:2006–2017

    Article  PubMed  CAS  Google Scholar 

  85. Bollard CM et al (2002) Adapting a transforming growth factor beta-related tumor protection strategy to enhance antitumor immunity. Blood 99:3179–3187

    Article  PubMed  CAS  Google Scholar 

  86. Cruz CR et al (2011) Improving T cell therapy for relapsed EBV-negative Hodgkin lymphoma by targeting upregulated MAGE-A4. Clin Cancer Res 17:7058–7066

    Article  PubMed  CAS  Google Scholar 

  87. Robinson BW, Morstyn G (1987) Natural killer (NK)-resistant human lung cancer cells are lysed by recombinant interleukin-2-activated NK cells. Cell Immunol 106:215–222

    Article  PubMed  CAS  Google Scholar 

  88. Fujisaki H et al (2009) Expansion of highly cytotoxic human natural killer cells for cancer cell therapy. Cancer Res 69:4010–4017

    Article  PubMed  CAS  Google Scholar 

  89. Rosenberg SA et al (1985) Observations on the systemic administration of autologous lymphokine-activated killer cells and recombinant interleukin-2 to patients with metastatic cancer. N Engl J Med 313:1485–1492

    Article  PubMed  CAS  Google Scholar 

  90. Oldham RK (1983) Natural killer cells: artifact to reality: an odyssey in biology. Cancer Metastasis Rev 2:323–336

    Article  PubMed  CAS  Google Scholar 

  91. Caligiuri MA (2008) Human natural killer cells. Blood 112:461–469

    Article  PubMed  CAS  Google Scholar 

  92. Vivier E, Tomasello E, Baratin M, Walzer T, Ugolini S (2008) Functions of natural killer cells. Nat Immunol 9:503–510

    Article  PubMed  CAS  Google Scholar 

  93. Sutlu T, Alici E (2009) Natural killer cell-based immunotherapy in cancer: current insights and future prospects. J Intern Med 266:154–181

    Article  PubMed  CAS  Google Scholar 

  94. Ruggeri L, Mancusi A, Capanni M, Martelli MF, Velardi A (2005) Exploitation of alloreactive NK cells in adoptive immunotherapy of cancer. Curr Opin Immunol 17:211–217

    Article  PubMed  CAS  Google Scholar 

  95. Miller JS et al (2005) Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood 105:3051–3057

    Article  PubMed  CAS  Google Scholar 

  96. Burns LJ et al (2003) IL-2-based immunotherapy after autologous transplantation for lymphoma and breast cancer induces immune activation and cytokine release: a phase I/II trial. Bone Marrow Transplant 32:177–186

    Article  PubMed  CAS  Google Scholar 

  97. Shi J et al (2008) Infusion of haplo-identical killer immunoglobulin-like receptor ligand mismatched NK cells for relapsed myeloma in the setting of autologous stem cell transplantation. Br J Haematol 143:641–653

    Article  PubMed  Google Scholar 

  98. Dillman RO et al (2009) Intralesional lymphokine-activated killer cells as adjuvant therapy for primary glioblastoma. J Immunother 32:914–919

    Article  PubMed  Google Scholar 

  99. Cho D, Campana D (2009) Expansion and activation of natural killer cells for cancer immunotherapy. Korean J Lab Med 29:89–96

    Article  PubMed  CAS  Google Scholar 

  100. Nagashima S et al (1998) Stable transduction of the interleukin-2 gene into human natural killer cell lines and their phenotypic and functional characterization in vitro and in vivo. Blood 91:3850–3861

    PubMed  CAS  Google Scholar 

  101. Kershaw MH et al (2006) A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer. Clin Cancer Res 12(20 Pt 1):6106–6115

    Google Scholar 

  102. Lamers CH et al (2006) Treatment of metastatic renal cell carcinoma with autologous T-lymphocytes genetically retargeted against carbonic anhydrase IX: first clinical experience. J Clin Oncol 24(13):e20–22

    Article  PubMed  Google Scholar 

  103. Till BG et al (2008) Adoptive immunotherapy for indolent non-Hodgkin lymphoma and mantle cell lymphoma using genetically modified autologous CD20-specific T cells. Blood 112(6):2261–2271

    Article  PubMed  CAS  Google Scholar 

  104. Park JR (2007) Adoptive transfer of chimeric antigen receptor re-directed cytolytic T lymphocyte clones in patients with neuroblastoma. Mol ther 15(4):825–833

    Article  PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Cliona M. Rooney .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2013 Springer Science+Business Media New York

About this chapter

Cite this chapter

Shaffer, D.R., Cruz, C.R.Y., Rooney, C.M. (2013). Adoptive T Cell Transfer. In: Curiel, T. (eds) Cancer Immunotherapy. Springer, New York, NY. https://doi.org/10.1007/978-1-4614-4732-0_3

Download citation

Publish with us

Policies and ethics