GET THE APP

Adoptive Immunotherapy for Acute Myeloid Leukemia: From Allogenei
Journal of Leukemia

Journal of Leukemia
Open Access

ISSN: 2329-6917

+44 1300 500008

Review Article - (2014) Volume 2, Issue 2

Adoptive Immunotherapy for Acute Myeloid Leukemia: From Allogeneic Hematopoietic Cell Transplantation to CAR T Cells

Merav Bar1,2*
1Division of Clinical Research, Fred Hutchinson Cancer Research Center, USA
2Department of Medicine, University of Washington, Seattle, WA, USA
*Corresponding Author: Merav Bar, Fred Hutchinson Cancer Research Centre, PO Box 19024 Seattle, WA 98109-1024, USA, Tel: +1 206-667-4971 Email:

Abstract

Allogeneic Hematopoietic Cell Transplantation (allo-HCT) has improved outcome of patients with high risk Acute Myeloid Leukemia (AML), but it harbors a risk of morbidity and mortality due to damage to normal cells and tissues by the high intensity conditioning or due to Graft Versus Host Disease (GVHD). As a major component of the curative potential of allo-HCT is derived from the response of the donor immunity against the malignant cells through the so called Graft Versus Leukemia effect (GVL), novel adoptive immunotherapy strategies have been developed to generate immune response against the leukemic cells, while sparing GVHD.

Keywords: Graft Versus Host Disease (GVHD); Hematopoiesis; Leukemic cells

Allogeneic Hematopoietic Cell Transplantation

The principle of anti-leukemic adoptive immunotherapy, as coined by Georges Mathé in 1965, is an activity of allogeneic immunologically competent cells against the host’s leukemic cells [1]. The best known model and the most commonly used method of anti-leukemic adoptive immunotherapy is allogeneic Hematopoietic Cell Transplantation (allo-HCT). The initial goal of allo-HCT was to eradicate leukemic cells by high doses of chemo or radiation therapy, which also destroys normal hematopoiesis, and then rescue the patient with a healthy hematopoietic progenitor cells from an allogeneic donor. However, it is now clear that the potential curative effect of allogeneic HCT is derived not only from the intense cytotoxic therapy, but also from the response of the donor’s immune cells against the leukemic cells, through a so called graft versus leukemia (GVL) effect.

The evidence to support GVL effect of allo-HCT are: (i) lower relapse rate among recipients of HLA-identical sibling transplants than recipients of syngeneic transplants [2-5]; (ii) decrease relapse rate among patients who develop Graft Versus Host Disease (GVHD) after allo-HCT [2,6]; (iii) decreased GVHD, but increased relapse rate after T cell depleted allo-HCT [7,8]; (iv) anti-leukemic effect and curative potential of Donor Lymphocyte Infusion (DLI) in patients with relapse leukemia after transplant [9,10]; and (v) effectiveness of nonmyeloablative HCT [11].

GVL is mediated through the immunologic activity of donor T cells and Natural Killer (NK) cells recognizing Leukemia-Associated Antigens (LAA), minor histocompetability antigens (mHag), and in case of mismatch or haploidentical transplants also major histocompetability antigens that are expressed by the malignant cells [12,13].

Donor immunity may contribute to the cure of the leukemia by allo-HCT, but is also the cause of GVHD, mediated by donor T cell recognizing antigens expressing by normal cells. GVHD is a major cause of morbidity and mortality after transplant, and it was recently demonstrated that the beneficial effect of GVL may be outweighed by the morbidity and mortality caused by GVHD [11]. Thus, therapies that provide potent GVL while sparing GVHD are needed.

NK cell-mediated anti-leukemic adoptive immunotherapy

Clinical observations have shown that donor NK cells may contribute to GVL activity in HLA-mismatched allogeneic HCT. NK cells display a number of activating and inhibitory receptors that interact with a wide variety of ligands on target cells, and the function of the NK cell depends on the net effect of the activating and inhibitory receptors [14]. Among the NK cells inhibitory receptors are Killer Immunoglobulin Receptors (KIRs) that recognize certain groups of HLA class I molecules on the target cell. It has been demonstrated that donor NK cells expressing KIRs that are mismatched with the recipient HLA class I and therefore are not inhibited by the HLA molecules of the recipient, so called alloreactive NK cells, are associated with decreased relapse and decreased GVHD rates after T cell-depleted HLA-mismatched or HLA-matched HCT in patients with Acute Myeloid Leukemia (AML) [15-18]. A number of early stage clinical trials have demonstrated that NK cells can be safely infused in AML patients following immunosuppressive chemotherapy and, in some cases, clinical responses without GVHD have been observed [19-21]. The results of these clinical trials demonstrate that NK cell-based therapy, which may provide anti-leukemic effect without GVHD, is a potential strategy to consolidate leukemiaremission in high-risk AML, without the need of allo-HCT.

T Cell Mediated Anti-Leukemic Adoptive Immunotherapy

A potential approach to generate GVL is by identifying and expending donor T cells that recognize LAA or mHags expressing on the malignant cells. Cytotoxic T lymphocyte (CTL) clones specific for hematopoietic restricted mHags have been shown to have anti- leukemic activity when used to treat patients with relapse leukemia after allo-HCT [22]. T cell clones specific for LAA as WT1, CD45, and PR1 have been generated to target leukemic cells [23-25], and recently Greenberg and colleagues reported results from a clinical trial using ex vivo expanded HLA-A*0201-restricted WT1-specific donor-derived CD8 CTL clones for treatment of relapsed or high-risk leukemia after allo-HCT [26]. This clinical trial demonstrated long term persistence and potential anti-leukemic activity of donor CTL clones specific for WT1. However, isolation and ex vivo expansion of high avidity mHag or LAA-specific T cell clones is challenging process, as it relies on the individual donor T cell repertoire and may result in products with variable quality and wide range of avidity, persistence, and function in vivo.

A potential way to circumvent the challenge of isolation and expansion of antigen-specific high avidity T cells is by using gene transfer technologies to genetically engineer T cell to express a unique high-affinity T cell receptor (TCR). The first successful TCR gene transfer to human peripheral blood lymphocytes conferring antitumor reactivity was reported by Clay and colleagues in 1999, using a TCR specific for an HLA-A2-restricted epitope of the MART-1 antigen, which is highly expressed by malignant melanomas [27]. Since then, several studies have demonstrated that transfer of a tumor antigen-specific TCR into T cells has successfully led to generation of an antigen-specific T cell population [28-41]. However there are a number of limitations to this approach:

1. The TCR recognizes a peptide fragment of its target antigen only when the peptide is presented by an HLA Class I molecule on the surface of the target cell. Therefore, antigen specific T cells are restricted to specific HLA allele, requires generation of patient-specific product. To date, most TCR gene transfer studies have focused on TCRs restricted to HLA-A2, as it is the most common HLA class I, present in ~50% of Caucasians [42,43].

2. Mispairing of the transduced and the endogenous TCR α and β chains that may lead to generation of non-specific TCR. A potential way to overcome this difficulty is insertion of point mutations into the constant regions of the transduced TCR α and β chains to create complementary cystein residues that form an inter-chain disulfide bond, which promotes preferential pairing between the transduced TCR chains and significantly reduces mispairing [44,45].

3. Downregulation of HLA class I expression by some leukemic cells have been reported [46], which may prevent recognition of the antigenic target by the TCR, and thus inhibits the antileukemic function of the T cell.

4. Chimeric Antigen Receptor (CAR) is another approach to T cell gene therapy. CAR is a genetically engineered molecule composed of an antigen-specific extracellular binding domain, fused to a transmembrane domain and intracellular T cellspecific signalling domain. The CARs are transduced into T cells, which become “CAR T cells” with specific target based on the CAR binding domain. Upon CAR binding to an antigen on the cell surface of a target cell, the CAR T cell induces apoptosis of the target cell using the same mechanisms of ordinary T cell. In contrast to a TCR, which recognizes a peptide fragment of an antigen only when the peptide is presented by an HLA Class I molecule on the surface of target cells, a CAR molecule recognizes an intact cell surface antigen, and it is HLA independent. Thus, the same CAR molecule may be used in a broad range of patients with different HLA types. CARs can target any cell surface antigen including carbohydrate and lipid moieties, which increases the repertoire of potential targets for CAR-base therapy. However CARs target only cell surface antigens, therefore, CAR T cells cannot target intracellular mutated or over expressed proteins.

Since the CAR concept was first introduced by Eshhar and colleagues in 1989 [47], CAR T cells have been generated to target several antigens on malignant cells [48-64], and a number of clinical trials are currently evaluating the use of CAR T cells, originated from allogeneic donors or patients, for treatment of different malignancies. In haematological malignancies there have been promising reports of using CAR-modified anti-CD19 T cells for treatment of B-cell malignancies [65-70]. CAR T cell targeting AML antigens as CD33 and CD123 have been reported [71,72], and recently early phase small clinical trial has demonstrated persistence and potential anti-leukemic effect of CAR T cell against the LeY antigen in AML [73].

Conclusions

Adoptive immunotherapy has the potential to provide long-term survival and even cure in patients with leukemia. The well-established allogeneic hematopoietic cell transplantation has improved outcome of patient with high risk AML, however it harbors the risk of morbidity and mortality due to damage to normal cells and tissues by the high intensity conditioning or due to graft versus host disease. Novel approaches using NK cells and T cells have been developed to provide more direct anti-leukemic therapy while sparing the risk of toxicity to normal tissues. However several obstacles need to be overcome in order to improve in vivo survival and antileukemic function of the transferred cells. Additionally, reliable large scale manufacturing of the therapeutic cells is essential to allow large clinical trials to evaluate the efficacy of various novel strategies before these therapies can be adopted for routine use to treat AML patients.

References

  1. Mathé G, Amiel JL, Schwarzenberg L, Cattan A, Schneider M (1965) Adoptive immunotherapy of acute leukemia: experimental and clinical results. Cancer Res 25: 1525-1531.
  2. Weiden PL, Sullivan KM, Flournoy N, Storb R, Thomas ED (1981) Antileukemic effect of chronic graft-versus-host disease: contribution to improved survival after allogeneic marrow transplantation. N Engl J Med 304: 1529-1533.
  3. Gale RP, Horowitz MM, Ash RC, Champlin RE, Goldman JM, et al. (1994) Identical-twin bone marrow transplants for leukemia. Ann Intern Med 120: 646-652.
  4. Horowitz MM, Gale RP, Sondel PM, Goldman JM, Kersey J, et al. (1990) Graft-versus-leukemia reactions after bone marrow transplantation. Blood 75: 555-562.
  5. Sullivan KM, Fefer A, Witherspoon R, Storb R, Buckner CD, et al. (1987) Graft-versus-leukemia in man: relationship of acute and chronic graft-versus-host disease to relapse of acute leukemia following allogeneic bone marrow transplantation. ProgClinBiol Res 244: 391-399.
  6. Sullivan KM, Weiden PL, Storb R, Witherspoon RP, Fefer A, et al. (1989) Influence of acute and chronic graft-versus-host disease on relapse and survival after bone marrow transplantation from HLA-identical siblings as treatment of acute and chronic leukemia. Blood 73: 1720-1728.
  7. Weiden PL, Flournoy N, Thomas ED, Prentice R, Fefer A, et al. (1979) Antileukemic effect of graft-versus-host disease in human recipients of allogeneic-marrow grafts. N Engl J Med 300: 1068-1073.
  8. Martin PJ, Hansen JA, Buckner CD, Sanders JE, Deeg HJ, et al. (1985) Effects of in vitro depletion of T cells in HLA-identical allogeneic marrow grafts. Blood 66: 664-672.
  9. Marmont AM, Horowitz MM, Gale RP, Sobocinski K, Ash RC, et al. (1991) T-cell depletion of HLA-identical transplants in leukemia. Blood 78: 2120-2130.
  10. Kolb HJ1, Mittermüller J, Clemm C, Holler E, Ledderose G, et al. (1990) Donor leukocyte transfusions for treatment of recurrent chronic myelogenousleukemia in marrow transplant patients.Blood 76: 2462-2465.
  11. Bar M, Sandmaier BM, Inamoto Y, Bruno B, Hari P, et al. (2013) Donor lymphocyte infusion for relapsed hematological malignancies after allogeneic hematopoietic cell transplantation: prognostic relevance of the initial CD3+ T cell dose. Biol Blood Marrow Transplant 19: 949-957.
  12. Storb R, Gyurkocza B, Storer BE, Sorror ML, Blume K, et al. (2013) Graft-versus-host disease and graft-versus-tumor effects after allogeneic hematopoietic cell transplantation. J ClinOncol 31: 1530-1538.
  13. Kolb HJ (2008) Graft-versus-leukemia effects of transplantation and donor lymphocytes. Blood 112: 4371-4383.
  14. Velardi A (2012) Natural killer cell alloreactivity 10 years later. CurrOpinHematol 19: 421-426.
  15. Farag SS, Fehniger TA, Ruggeri L, Velardi A, Caligiuri MA (2002) Natural killer cell receptors: new biology and insights into the graft-versus-leukemia effect. Blood 100: 1935-1947.
  16. Ruggeri L, Capanni M, Urbani E, Perruccio K, Shlomchik WD, et al. (2002) Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants. Science 295: 2097-2100.
  17. Moroi K, Sato T (1975) Comparison between procaine and isocarboxazid metabolism in vitro by a liver microsomal amidase-esterase. BiochemPharmacol 24: 1517-1521.
  18. Ruggeri L, Mancusi A, Capanni M, Urbani E, Carotti A, et al. (2007) Donor natural killer cell allorecognition of missing self in haploidentical hematopoietic transplantation for acute myeloid leukemia: challenging its predictive value. Blood 110: 433-440.
  19. Kalos M, Levine BL, Porter DL, Katz S, Grupp SA, et al. (2011) T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. SciTransl Med 3: 95ra73.
  20. Hsu KC, Keever-Taylor CA, Wilton A, Pinto C, Heller G, et al. (2005) Improved outcome in HLA-identical sibling hematopoietic stem-cell transplantation for acute myelogenousleukemia predicted by KIR and HLA genotypes. Blood 105: 4878-4884.
  21. Miller JS, Soignier Y, Panoskaltsis-Mortari A, McNearney SA, Yun GH, et al. (2005) Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood 105: 3051-3057.
  22. Rubnitz JE1, Inaba H, Ribeiro RC, Pounds S, Rooney B, et al. (2010) NKAML: a pilot study to determine the safety and feasibility of haploidentical natural killer cell transplantation in childhood acute myeloid leukemia. J ClinOncol 28: 955-959.
  23. Curti A, Ruggeri L, D'Addio A, Bontadini A, Dan E, et al. (2011) Successful transfer of alloreactivehaploidentical KIR ligand-mismatched natural killer cells after infusion in elderly high risk acute myeloid leukemia patients. Blood 118: 3273-3279.
  24. Warren EH, Fujii N, Akatsuka Y, Chaney CN, Mito JK, et al. (2010) Therapy of relapsed leukemia after allogeneic hematopoietic cell transplantation with T cells specific for minor histocompatibility antigens. Blood 115: 3869-3878.
  25. Guo Y, Niiya H, Azuma T, Uchida N, Yakushijin Y, et al. (2005) Direct recognition and lysis of leukemia cells by WT1-specific CD4+ T lymphocytes in an HLA class II-restricted manner. Blood 106: 1415-1418.
  26. Amrolia PJ, Reid SD, Gao L, Schultheis B, Dotti G, et al. (2003) Allorestricted cytotoxic T cells specific for human CD45 show potent antileukemic activity. Blood 101: 1007-1014.
  27. Ma Q, Wang C, Jones D, Quintanilla KE, Li D, et al. (2010) Adoptive transfer of PR1 cytotoxic T lymphocytes associated with reduced leukemia burden in a mouse acute myeloid leukemiaxenograft model. Cytotherapy 12: 1056-1062.
  28. Chapuis AG, Ragnarsson GB, Nguyen HN, Chaney CN, Pufnock JS, et al. (2013) Transferred WT1-reactive CD8+ T cells can mediate antileukemic activity and persist in post-transplant patients. SciTransl Med 5: 174ra27.
  29. Clay TM, Custer MC, Sachs J, Hwu P, Rosenberg SA, et al. (1999) Efficient transfer of a tumor antigen-reactive TCR to human peripheral blood lymphocytes confers anti-tumor reactivity. J Immunol 163: 507-513.
  30. Stanislawski T, Voss RH, Lotz C, Sadovnikova E, Willemsen RA, et al. (2001) Circumventing tolerance to a human MDM2-derived tumor antigen by TCR gene transfer. Nat Immunol 2: 962-970.
  31. Morgan RA, Dudley ME, Yu YY, Zheng Z, Robbins PF, et al. (2003) High efficiency TCR gene transfer into primary human lymphocytes affords avid recognition of melanoma tumor antigen glycoprotein 100 and does not alter the recognition of autologous melanoma antigens. J Immunol 171: 3287-3295.
  32. Jolly RD, Thompson KG, Winchester BG (1975) Bovine mannosidosis--a model lysosomal storage disease. Birth Defects Orig Artic Ser 11: 273-278.
  33. Schaft N, Willemsen RA, de Vries J, Lankiewicz B, Essers BW, et al. (2003) Peptide fine specificity of anti-glycoprotein 100 CTL is preserved following transfer of engineered TCR alpha beta genes into primary human T lymphocytes. J Immunol 170: 2186-2194.
  34. Marniemi J, Parkki MG (1975) Radiochemical assay of glutathione S-epoxide transferase and its enhancement by phenobarbital in rat liver in vivo. BiochemPharmacol 24: 1569-1572.
  35. Zhao Y, Zheng Z, Robbins PF, Khong HT, Rosenberg SA, et al. (2005) Primary human lymphocytes transduced with NY-ESO-1 antigen-specific TCR genes recognize and kill diverse human tumor cell lines. J Immunol 174: 4415-4423.
  36. Cohen CJ, Zheng Z, Bray R, Zhao Y, Sherman LA, et al. (2005) Recognition of fresh human tumor by human peripheral blood lymphocytes transduced with a bicistronic retroviral vector encoding a murine anti-p53 TCR. J Immunol 175: 5799-5808.
  37. Parkhurst MR, Joo J, Riley JP, Yu Z, Li Y, et al. (2009) Characterization of genetically modified T-cell receptors that recognize the CEA:691-699 peptide in the context of HLA-A2.1 on human colorectal cancer cells. Clin Cancer Res 15: 169-180.
  38. Frankel TL, Burns WR, Peng PD, Yu Z, Chinnasamy D, et al. (2010) Both CD4 and CD8 T cells mediate equally effective in vivo tumor treatment when engineered with a highly avid TCR targeting tyrosinase. J Immunol 184: 5988-5998.
  39. Chinnasamy N, Wargo JA, Yu Z, Rao M, Frankel TL, et al. (2011) A TCR targeting the HLA-A*0201-restricted epitope of MAGE-A3 recognizes multiple epitopes of the MAGE-A antigen superfamily in several types of cancer. J Immunol 186: 685-696.
  40. Straetemans T, van Brakel M, van Steenbergen S, Broertjes M, Drexhage J, et al. (2012) TCR gene transfer: MAGE-C2/HLA-A2 and MAGE-A3/HLA-DP4 epitopes as melanoma-specific immune targets. ClinDevImmunol 2012: 586314.
  41. Hillerdal V, Nilsson B, Carlsson B, Eriksson F, Essand M (2012) T cells engineered with a T cell receptor against the prostate antigen TARP specifically kill HLA-A2+ prostate and breast cancer cells. ProcNatlAcadSci U S A 109: 15877-15881.
  42. Tsuji T, Yasukawa M, Matsuzaki J, Ohkuri T, Chamoto K, et al. (2005) Generation of tumor-specific, HLA class I-restricted human Th1 and Tc1 cells by cell engineering with tumor peptide-specific T-cell receptor genes. Blood 106: 470-476.
  43. Heemskerk MH, Hoogeboom M, Hagedoorn R, Kester MG, Willemze R, et al. (2004) Reprogramming of virus-specific T cells into leukemia-reactive T cells using T cell receptor gene transfer. J Exp Med 199: 885-894.
  44. Xue SA, Gao L, Hart D, Gillmore R, Qasim W, et al. (2005) Elimination of human leukemia cells in NOD/SCID mice by WT1-TCR gene-transduced human T cells. Blood 106: 3062-3067.
  45. Krausa P, Brywka M 3rd, Savage D, Hui KM, Bunce M, et al. (1995) Genetic polymorphism within HLA-A- 02: significant allelic variation revealed in different populations. Tissue Antigens 45: 223-231.
  46. Cohen CJ, Li YF, El-Gamil M, Robbins PF, Rosenberg SA, et al. (2007) Enhanced antitumor activity of T cells engineered to express T-cell receptors with a second disulfide bond. Cancer Res 67: 3898-3903.
  47. Schmoldt A, Benthe HF, Haberland G (1975) Digitoxin metabolism by rat liver microsomes. BiochemPharmacol 24: 1639-1641.
  48. Kuball J, Dossett ML, Wolfl M, Ho WY, Voss RH, et al. (2007) Facilitating matched pairing and expression of TCR chains introduced into human T cells. Blood 109: 2331-2338.
  49. Demanet C, Mulder A, Deneys V, Worsham MJ, Maes P, et al. (2004) Down-regulation of HLA-A and HLA-Bw6, but not HLA-Bw4, allospecificities in leukemic cells: an escape mechanism from CTL and NK attack? Blood 103: 3122-3130.
  50. Gross G, Waks T, Eshhar Z (1989) Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. ProcNatlAcadSci U S A 86: 10024-10028.
  51. Finney HM, Lawson AD, Bebbington CR, Weir AN (1998) Chimeric receptors providing both primary and costimulatorysignaling in T cells from a single gene product. J Immunol 161: 2791-2797.
  52. Brentjens RJ, Latouche JB, Santos E, Marti F, Gong MC, et al. (2003) Eradication of systemic B-cell tumors by genetically targeted human T lymphocytes co-stimulated by CD80 and interleukin-15. Nat Med 9: 279-286.
  53. Imai C, Mihara K, Andreansky M, Nicholson IC, Pui CH, et al. (2004) Chimeric receptors with 4-1BB signaling capacity provoke potent cytotoxicity against acute lymphoblastic leukemia. Leukemia 18: 676-684.
  54. Lamers CH, Sleijfer S, Vulto AG, Kruit WH, Kliffen M, et al. (2006) Treatment of metastatic renal cell carcinoma with autologous T-lymphocytes genetically retargeted against carbonic anhydrase IX: first clinical experience. J ClinOncol 24: e20-22.
  55. Till BG, Jensen MC, Wang J, Chen EY, Wood BL, et al. (2008) Adoptive immunotherapy for indolent non-Hodgkin lymphoma and mantle cell lymphoma using genetically modified autologous CD20-specific T cells. Blood 112: 2261-2271.
  56. Zhao Y, Wang QJ, Yang S, Kochenderfer JN, Zheng Z, et al. (2009) A herceptin-based chimeric antigen receptor with modified signaling domains leads to enhanced survival of transduced T lymphocytes and antitumor activity. J Immunol 183: 5563-5574.
  57. Morgan RA, Yang JC, Kitano M, Dudley ME, Laurencot CM, et al. (2010) Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. MolTher 18: 843-851.
  58. Krause A, Guo HF, Latouche JB, Tan C, Cheung NK, et al. (1998) Antigen-dependent CD28 signaling selectively enhances survival and proliferation in genetically modified activated human primary T lymphocytes. J Exp Med 188: 619-626.
  59. Pule MA, Savoldo B, Myers GD, Rossig C, Russell HV, et al. (2008) Virus-specific T cells engineered to coexpresstumor-specific receptors: persistence and antitumor activity in individuals with neuroblastoma. Nat Med 14: 1264-1270.
  60. Maher J, Brentjens RJ, Gunset G, Rivière I, Sadelain M (2002) Human T-lymphocyte cytotoxicity and proliferation directed by a single chimeric TCRzeta /CD28 receptor. Nat Biotechnol 20: 70-75.
  61. Katari UL, Keirnan JM, Worth AC, Hodges SE, Leen AM, et al. (2011) Engineered T cells for pancreatic cancer treatment. HPB (Oxford) 13: 643-650.
  62. Carpenito C, Milone MC, Hassan R, Simonet JC, Lakhal M, et al. (2009) Control of large, established tumorxenografts with genetically retargeted human T cells containing CD28 and CD137 domains. ProcNatlAcadSci U S A 106: 3360-3365.
  63. Lefkowitz RJ (1975) Identification of adenylatecyclase-coupled beta-adrenergic receptors with radiolabeled beta-adrenergic antagonists. BiochemPharmacol 24: 1651-1658.
  64. Park JR, Digiusto DL, Slovak M, Wright C, Naranjo A, et al. (2007) Adoptive transfer of chimeric antigen receptor re-directed cytolytic T lymphocyte clones in patients with neuroblastoma. MolTher 15: 825-833.
  65. Chinnasamy D, Yu Z, Theoret MR, Zhao Y, Shrimali RK, et al. (2010) Gene therapy using genetically modified lymphocytes targeting VEGFR-2 inhibits the growth of vascularized syngenictumors in mice. J Clin Invest 120: 3953-3968.
  66. Chekmasova AA, Rao TD, Nikhamin Y, Park KJ, Levine DA, et al. (2010) Successful eradication of established peritoneal ovarian tumors in SCID-Beige mice following adoptive transfer of T cells genetically targeted to the MUC16 antigen. Clin Cancer Res 16: 3594-3606.
  67. Kershaw MH, Westwood JA, Parker LL, Wang G, Eshhar Z, et al. (2006) A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer. Clin Cancer Res 12: 6106-6115.
  68. Song DG, Ye Q, Carpenito C, Poussin M, Wang LP, et al. (2011) In vivo persistence, tumor localization, and antitumor activity of CAR-engineered T cells is enhanced by costimulatorysignaling through CD137 (4-1BB). Cancer Res 71: 4617-4627.
  69. 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.
  70. Brentjens RJ, Rivière I, Park JH, Davila ML, Wang X, et al. (2011) Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood 118: 4817-4828.
  71. Grupp SA, Kalos M, Barrett D, Aplenc R, Porter DL, et al. (2013) Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med 368: 1509-1518.
  72. Mardiros A, Dos Santos C, McDonald T, Brown CE, Wang X, et al. (2013) T cells expressing CD123-specific chimeric antigen receptors exhibit specific cytolytic effector functions and antitumor effects against human acute myeloid leukemia. Blood 122: 3138-3148.
Citation: Bar M (2014) Adoptive Immunotherapy for Acute Myeloid Leukemia: From Allogeneic Hematopoietic Cell Transplantation to CAR T Cells. J Leuk (Los Angel) 2:134.

Copyright: © 2014 Bar M. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top