Skip to main content
Cancer Immunology, Immunotherapy : CII logoLink to Cancer Immunology, Immunotherapy : CII
. 2011 Apr 26;60(6):757–769. doi: 10.1007/s00262-011-1022-6

Clinical evaluation of cellular immunotherapy in acute myeloid leukaemia

Evelien L J Smits 1,, Cindy Lee 2,3, Nicola Hardwick 4, Suzanne Brooks 3, Viggo F I Van Tendeloo 1, Kim Orchard 2, Barbara-ann Guinn 3,4
PMCID: PMC11029703  PMID: 21519825

Abstract

Immunotherapy is currently under active investigation as an adjuvant therapy to improve the overall survival of patients with acute myeloid leukaemia (AML) by eliminating residual leukaemic cells following standard therapy. The graft-versus-leukaemia effect observed following allogeneic haematopoietic stem cell transplantation has already demonstrated the significant role of immune cells in controlling AML, paving the way to further exploitation of this effect in optimized immunotherapy protocols. In this review, we discuss the current state of cellular immunotherapy as adjuvant therapy for AML, with a particular focus on new strategies and recently published results of preclinical and clinical studies. Therapeutic vaccines that are being tested in AML include whole tumour cells as an autologous source of multiple leukaemia-associated antigens (LAA) and autologous dendritic cells loaded with LAA as effective antigen-presenting cells. Furthermore, adoptive transfer of cytotoxic T cells or natural killer cells is under active investigation. Results from phase I and II trials are promising and support further investigation into the potential of cellular immunotherapeutic strategies to prevent or fight relapse in AML patients.

Keywords: Acute leukaemia, Immunotherapy, Cellular therapy, AML, Whole cell vaccine, Adoptive transfer

Introduction

The prognosis for patients with acute myeloid leukaemia (AML) has been steadily improving, in particular for younger patients with an overall survival rate of 60% [1]. However, relapses still occur, and for older patients (>60 years), the outlook remains dismal with less than 10% attaining long-term survival (estimated survival is only 20% at 2 years) [2, 3].

The advent of haematopoietic stem cell transplantation (HSCT) and donor lymphocyte infusions (DLI) has led to a shift in the paradigm of AML therapy [4, 5]. HSCT offers the advantage of dose intensification, administering high-dose chemotherapy with or without total body irradiation, in an attempt to further reduce tumour burden [6]. Furthermore, non-autologous HSCT can result in an immunologic graft-versus-leukaemia (GVL) effect, where leukaemic cells are destroyed by allogeneic transplant cells [7]. However, HSCT is associated with a significant treatment-related mortality and morbidity, including graft-versus-host-disease (GVHD) and complications such as veno-occlusive disease and infections. HSCT is also associated with an elevated risk of developing secondary malignancies such as solid cancers [6, 7].

More recently, treatment has shifted to the use of non-myeloablative and reduced-intensity conditioning regimens, which utilize administration of chemotherapy and/or radiation at reduced doses, sufficient to allow donor stem cell engraftment and to enable eradication of residual leukaemia cells by allogeneic effector immune cells within the graft [8]. Patients are frequently given DLI after treatment to boost this GVL effect. Non-myeloablative transplants have allowed extension of the upper age limit for allogeneic transplantation and reduced toxicity with the use of less intensive conditioning regimens. Unfortunately, this approach is associated with a significant relapse rate and therefore mostly restricted in use to clinical trials for selected patients [8].

The majority of AML patients undergoing HSCT receive donor stem cells from siblings or unrelated donors matched for MHC molecules [6]. When transplantation is performed with a mismatched family member or unrelated donors, detrimental alloreactive responses towards the mismatched MHC molecules can occur, resulting in GVHD. Even with complete matching for MHC molecules, T cell reactivity to minor histocompatibility antigens (mHAgs) due to polymorphisms in common cellular proteins can occur, which also leads to GVHD. In order to reduce the severity of GVHD and its mortality and morbidity, grafts can be manipulated to be depleted of T cells prior to transplantation [9]. This approach suffers from increased relapse rates due to the loss of the beneficial associated GVL effect, and patients continue to die from their disease. Therefore, the need for alternative therapies remains acute to fight relapse. Targeted anti-leukaemia immunotherapy would offer the desired combination of GVL effects with minimal toxicity due to the preferential targeting of AML cells. Recent findings of the group of Fuchs open the gate to a broader use of immunotherapy for patients with advanced haematological malignancies. The authors observed that short-course high-dose cyclophosphamide administered post-transplantation was effective in preventing acute and chronic GVHD in patients after HLA-matched or partially mismatched allo-HSCT [1012]. In this way, immunosuppressive treatment could potentially be limited after transplantation, paving the way to use immunotherapy strategies early after transplantation, a train of thought that is also followed by others [13]. In this review, we describe preclinical and clinical studies testing cellular immunotherapies for AML, with special focus on recent new developments in this area. Other clinical trials comprising interleukin (IL)-2 administration, monoclonal antibody therapy and peptide-based vaccination have already extensively been reviewed elsewhere [1418].

Cellular therapy to treat AML

In the last decade, research efforts have been broadening the array of cell therapy approaches which can be used as adjuvant therapy in AML. Cellular therapy comprises the administration of therapeutic cellular vaccines (tumour or DC vaccines) to induce specific immune responses in vivo, as well as adoptive transfer of effector cells to exert anti-tumour immune reactivity (T cells or NK cells) in patients.

NK cells can sense the altered expression of MHC molecules and stress markers on malignant cells through the expression of different NK cell receptors [19]. Furthermore, abnormal expression of normal cellular proteins or expression of mutated proteins in malignant cells give rise to ‘tumour proteins’, fragments of which can be presented on the cell surface, called tumour-associated antigens (TAA) [20]. By binding of TAA to its T cell receptor (TCR), an activated effector T cell is able to recognize and destroy malignant cells expressing these TAA.

It is generally believed that the presence of a large tumour load would overrule the capacity of the immune system to eradicate the malignant cells. Therefore, cellular immunotherapy is particularly being examined in AML patients as an adjuvant therapy following standard therapy to destroy residual leukaemic cells [14, 21]. Ideally, AML immunotherapy would induce an immune response that clears minimal residual disease (MRD), while sparing normal tissue, and would generate immunological memory that protects against disease recurrence. After chemotherapy, T cells are reduced in AML patients [22]. Lymphocyte counts in AML patients during and within 1 month after induction chemotherapy are inversely correlated with relapse rate and overall survival [23, 24], pointing to the importance of immune surveillance, and more specifically lymphocytes, in preventing relapse [16]. Results of active immunization strategies (described below) support the hypothesis that the immune system of AML patients in remission can be boosted to attack AML cells. Examples of high-potential leukaemia-associated antigens (LAAs) are Wilms’ tumour protein (WT1), proteinase 3 (PR3) and the receptor for hyaluronic acid-mediated motility (RHAMM/CD168). The immunogenicity of WT1, PR3 and RHAMM has been shown by the presence of TAA-specific cytotoxic T lymphocytes (CTL) in AML patients [2527]. Furthermore, in vitro generated or ex vivo isolated T cells specific for WT1, PR3 or RHAMM were able to kill primary myeloid leukaemic cells [2630]. An in-depth description of LAAs relevant to AML has been published elsewhere [3137].

Whole tumour cells

Autologous modified whole tumour cells are attractive accessory cells for the in vivo or in vitro stimulation of AML-specific immune responses directed against multiple antigens, thereby decreasing the risk of generating escape mutants. Furthermore, random mutations in tumour cells can generate unique antigens in each individual, providing a rationale for customized immunotherapy approaches. A disadvantage of whole tumour cell approaches is that along with TAA expression, they also contain a wide range of normal self-proteins not linked to the malignant phenotype. Generally, tolerance to self-antigens is tightly controlled, but the aim of cancer immunotherapy is to break tolerance to TAAs and therefore, the danger of inducing autoimmunity to normal tissues should not be discounted [38]. It is hoped that as tumour cells express self-antigens considered as TAA to a higher extent compared to normal tissues, they will be more susceptible to TAA-specific CTL recognition. Focusing on WT1 as a TAA, WT1-specific CTL could possibly also recognize normal hematopoietic progenitor cells expressing WT1 in the bone marrow, causing leucocytopaenia. Until now, leucocytopaenia was only detected in patients with myelodysplastic syndrome (MDS) following peptide vaccination and not in AML [39, 40]. In MDS, the hematopoietic stem cells are transformed and so it is believed that the observed leucocytopaenia is an indicator of the effectiveness of the vaccine [39].

In AML, expression of MHC molecules on primary tumour cells allows them to officiate as accessory cells [41], presenting a range of patient-specific TAA to T cells. However, their antigen-presenting potential may be hampered by insufficient expression of costimulatory molecules, high expression of apoptosis-inducing molecules and/or constitutive secretion of immunosuppressive cytokines [42]. Not only the antigen-presenting capacity, but also the susceptibility of AML cells for immune-mediated cell death is affected by these factors, as well as by heterogenous constitutive chemokine secretion and chemokine responsiveness of primary AML cells [43, 44]. In order to make therapy outcome more predictable, heterogeneity of the expression of immunomodulatory and apoptosis-related molecules by primary AML cells should be reduced (e.g. by genetic modification, vide infra) or should be taken into account to define subsets of patients that are likely to benefit from immunotherapy. Another important parameter to reduce heterogeneity is the choice of culture medium, because it influences the constitutive cytokine secretion of in vitro cultured primary AML cells [45, 46]. It was shown that these cells can be cultured in standardized serum-free conditions [45, 46], thereby excluding the risk of adverse immune reactions against serum-associated molecules.

In order to avoid transfusion of viable, proliferating AML cells when using them as accessory cells, primary AML cells can be γ-irradiated. Bruserud and Ulvestad [47] investigated whether γ-irradiated AML cells could function as accessory cells during anti-CD3-stimulated T cell activation in vitro. The authors showed an irradiation dose-dependent reduction in the secretion of the proinflammatory cytokines IL-1β, IL-6 and TNF-α by primary AML cells. Although highly proliferative T cell responses were not always associated with high levels of cytokine secretion, an irradiation dose-dependent reduction in anti-CD3-stimulated T cell proliferation was observed. A dose of 50 Gy was considered by the authors to be both safe and to conserve the accessory cell function of primary AML cells.

In one of the earliest vaccination trials using autologous whole AML cells, primary cells were frozen in the presence of autologous serum and gamma-irradiated with 100 Gy after thawing [48]. Four AML patients were included in the clinical trial following achievement of chemotherapy-induced remission, receiving intradermal and subcutaneous injections in all four limbs. In vitro analysis of peripheral blood effector cells collected from the patients pre- and post-vaccination showed that immunotherapy increased the activity of patient-derived lymphocytes against autologous leukaemic cells [48]. Later, the protocol was adjusted by co-injecting irradiated autologous AML cells with Bacillus Calmette-Guerin (BCG), resulting in prolonged remission and longer survival after first relapse, compared to non-vaccinated AML patients [49], indicating a clinical effect of the vaccine. No data are available to allow correlation of clinical effects to specific characteristics of the AML cells. A positive outcome was also reported in the study of Zhang et al. [50], where autologous whole cell vaccines comprising Mitomycin C-treated autologous AML cells mixed with IL-2, GM-CSF and IL-6 were administered subcutaneously in the limb of 25 AML patients with relapsed or refractory disease. Partial responses were observed in five patients and CRs seen in four patients were associated with a reduction in serum IL-10 post-vaccination. Efficacy of the vaccine was associated with expression of CD80, which was exclusively expressed on AML cells of some patients with AML FAB M4 and M5.

Genetic modification can be used to reduce heterogeneity and to increase immunogenicity of tumour cells from a wide range of patients by conferring expression of immunostimulatory molecules, including cytokines and costimulatory molecules [51]. Early mouse in vivo studies on the transfer of cytokine or costimulatory genes into murine AML cells or human leukaemia cell lines demonstrated reduced tumourigenicity of the transfected cells [5255]. Furthermore, vaccinated mice developed protective immunity against rechallenge with wild-type cells and MRD could be eliminated [5356].

Building on these promising results, several gene transfer methods were tested to modify human primary AML cells, trying to overcome variable levels and/or low persistence of transgene expression. These methods include plasmid transfection [57], nucleofection [58] and transduction with vectors from retroviruses [59, 60], adenoviruses [6063], herpes simplex viruses [64] and/or adeno-associated viruses [65, 66]. Despite technical obstacles, expression of immunostimulatory molecules by primary AML cells consistently resulted in enhanced T cell stimulation in vitro, mostly shown in allogeneic assays [14]. An efficient method for the genetic modification of human primary AML cells is the use of lentiviral vectors [6769]. In contrast to classical retroviral vectors, HIV-1-based lentiviral vectors readily transduce mitotic as well as a number of post-mitotic targets [70]. As lentiviral vectors do not encode viral proteins [71], they are suitable for immune gene therapy strategies. A report on the modification of human primary AML cells using lentiviral vectors to express B7.1 and GM-CSF in primary AML cells showed promising levels of transgene expression, but rather low proliferation of autologous T cells [67]. Studies on a fusagene lentiviral vector [72] encoding CD80 and IL-2 resulted in high levels of autologous T cell stimulation, NK cell activation and lysis of targeted AML cells [7375]. A phase I clinical trial has now been started in poor prognosis AML patients using autologous whole cell vaccines modified with the fusagene lentiviral vector to express CD80 and IL-2 [74]. Furthermore, a phase II clinical study was recently conducted using whole cell vaccines consisting of gamma-irradiated autologous AML cells mixed with K562 cells secreting GM-CSF in pre- and post-transplantation immunotherapy protocols [76]. Tumour-specific immune responses were detected, as well as reductions in MRD as measured by WT1 transcript levels [76]. These results are promising, despite the controversy on the use of whole tumour cell vaccines and GM-CSF as an immune-adjuvant since the halted phase III trial on GM-CSF-secreting allogeneic cells in prostate cancer [77].

In addition to gene transfer, Smits et al. [78] showed that the immunogenicity of AML cells can also be increased by transfecting AML cells with the Toll-like receptor (TLR)3 ligand polyinosinic polycytidylic acid [poly(I:C)]. Toll-like receptor ligands are pathogen-associated molecular patterns, small motifs that are conserved within a group of micro-organisms and can activate immune cells [79]. By loading cells with the non-coding synthetic dsRNA poly(I:C), viral infection was mimicked. The pleiotropic response of primary AML cells and AML cell lines to poly(I:C) transfection included apoptosis, increased expression of costimulatory and MHC molecules, and production of proinflammatory cytokines. Strikingly, in response to electroporation with poly(I:C) all AML cell samples produced type I interferons, known for their anti-tumoural effects [80, 81]. Focusing on dendritic cells (DCs), coculture with apoptotic poly(I:C)-loaded AML cells induced maturation and enhanced their Th1-inducing capacity, which is important for tumour immunotherapy [78]. Furthermore, poly(I:C)-loaded AML cells could activate both the helper and the cytotoxic function of NK cells, resulting in NK cell-derived IFN-γ and killing of AML cells, respectively [82, 83]. Also, the TLR7/8 ligand resiquimod increased the immunogenicity of AML cells and activated IFN-γ secretion by NK cells [84], making TLR ligands attractive molecules to be incorporated in future AML immunotherapy strategies.

If an autologous AML vaccine is used to activate T cells, an important aspect to consider is the in vivo attraction of T cells by the vaccine, as well as recruitment of T cells to the AML compartment. Bruserud et al. [44, 85] showed that only a minority of patients had diminished in vitro chemotaxis of normal lymphocytes towards primary AML cells due to absence of chemokine release by the AML cells. In the broad constitutive chemokine release profile of the other primary AML cells, three chemokine clusters could be identified. Although samples from all clusters had a similar ability to attract normal T cells in vitro (predominantly CD4+ T cells), it is expected that different chemokine release profiles will contribute to the heterogeneity in the outcome of antileukaemic immunotherapy, since there was a significant variation between patients in intravascular and extravascular levels of the T cell chemotactic chemokines CXCL10, CCL5 and CCL17 [43].

The influence of irradiation and genetic modification on chemokine release profiles requires further investigation. Depending on which protocol was used, cytokine-mediated in vitro induction of a DC phenotype in primary human AML cells was associated with a significant increase in CCL17 and CCL22 release, pivotal for chemotaxis of normal T cells [86]. These data provide evidence that modification of primary AML cells influences the secretion of T cell attracting chemokines.

Failure of immune responses to eliminate leukaemic cells can be due to T cell dysfunction and to increased suppressor activity mediated by regulatory T (Treg) cells. Le Dieu et al. [87] showed that T cells from newly diagnosed AML patients had an abnormal genotype and a decreased ability to form functional immune synapses with AML cells. Further research is required to investigate if this is also the case for AML patients in remission as primary target group for immunotherapy. Elevated levels of Treg cells with high levels of suppressor activity were detected both in AML patients that were newly diagnosed and in patients in complete remission [88]. These factors can potentially block positive outcome of vaccination, either using modified tumour cells or DCs.

Dendritic cells

DCs are immune cells located in many normal tissues. Being professional antigen-presenting cells, they are specially adapted to direct the immune response to induce immune activation or tolerance [89]. They are known to be the most potent stimulators of naïve and resting T cells and therefore are promising vehicles for the in vivo delivery of tumour antigens (in the form of a ‘DC vaccine’) or through their in vitro generation of TAA-specific T cells [90, 91]. In order to obtain sufficient DCs for immunotherapeutic purposes, DCs are often cultured in vitro from a starting population of positively selected CD14 monocytes isolated from peripheral blood. The cells are differentiated under the influence of cytokines into ‘monocyte-derived DC’ [9294]. These immature DC are then amenable to loading with TAA. In AML, another attractive strategy is to differentiate the myeloid leukaemia cells themselves into AML-derived DC to circumvent the need for loading with TAAs [95, 96]. Only a few phase I clinical trials have been performed to date using AML-DC and have reported that the DC vaccine is well tolerated and able to induce immunological responses albeit with limited feasibility in some AML patients in terms of obtaining sufficient AML-DC vaccines and so far without evidence of clinical benefit [97100]. Currently, protocols are being optimized in order to increase the success rate of generating AML-DC [101, 102] and to increase the cytolytic capacity of T cells primed with AML-DC [103, 104].

For monocyte- or bone marrow-derived DC, two key issues are choice of TAA and method of TAA loading. Until now, preclinical and clinical studies have described loading of human DC with PML-RARα, survivin or WT1 using peptide pulsing or mRNA electroporation [105107]. Prior identification of LAA is not required when DC are fed with AML cell lysates and/or apoptotic AML cells, or when AML cell-DC fusion hybrids are generated [108114]. Irrespective of the antigen loading method, these studies have reported DC to be potent activators of autologous or allogeneic HLA-matched CTL in vitro, resulting in cytotoxic responses directed against primary AML cells and providing a rationale for clinical evaluation of DC vaccines in AML.

The primary focus of AML immunotherapy remains the prevention of relapse, and an attractive strategy is to take monocytes from AML patients in remission to generate monocyte-derived DC. Clinical-grade DC with the desired phenotype could be generated in sufficient numbers when starting with monocytes from AML patients in remission [115]. In a recent phase I/II study, DC were loaded with the LAA WT1 by mRNA electroporation. The DC vaccine was well tolerated, and five out of ten AML patients obtained molecular remission following administration of autologous WT1-loaded DC [107]. Furthermore, two of the patients in partial remission achieved CR instead of the expected relapse [107], supporting further clinical evaluation of this DC vaccine. In a phase I clinical study by Lee et al. [112], autologous monocyte-derived DC vaccines were well tolerated in two AML patients in relapse. The DC were loaded with LAA using AML lysates. Despite the detection of immunological responses, no clinical responses were observed, in terms of a lowered percentage of AML cells in the bone marrow [112], pointing to the importance of the choice of antigen and/or time of administration.

Adoptive transfer of T cells

Methods of immunotherapy described so far can be classed as active immunization or vaccination, where the introduction of immunogenic material aims to elicit activation and expansion of effector cells in the endogenous immune repertoire. A disadvantage of this approach is the requirement of the patient for some degree of immune competence. An alternative is passive immunization or adoptive transfer, where transfer of functional immune effector cells confers immunity and could possibly overcome lymphocyte deficiencies in AML patients [116121].

DLIs comprise the transfer of donor lymphocytes to leukaemia patients in relapse following HSCT [5]. Although DLIs have been successful in the treatment of relapsed CML [122], the induced remission in AML or accelerated phase CML patients has not been as long-lasting, probably due to tumour growth exceeding the capabilities of the immune response [123125]. Adoptively transferred T cells in DLI can exert anti-leukaemia responses, resulting in a so-called GVL effect. However, GVHD can also be initiated if the allogeneic T cells react against non-leukaemic cells, thereby causing damage to normal tissues [125, 126]. In order to reduce GVHD and to increase the GVL effect, donor lymphocytes can first be manipulated ex vivo to switch off alloreactive effector T cells and to enrich leukaemia-specific T cells. Anergy can be induced in donor alloreactive T cells by costimulatory molecule blockade during coculture with host peripheral blood mononuclear cells [127130]. In order to avoid possible bystander effects or reversion of anergy in vivo, selective allodepletion is being examined. Donor T cells are cocultured with host antigen-presenting cells, followed by elimination of activated T cells using antibodies targeting activation-induced markers such as CD25, CD69, CD71, CD95, CD134, CD137 and/or HLA-DR [131142]. Results of clinical trials using allodepletion point out that this promising approach needs optimization in order to further reduce transplant-related morbidities [136, 138].

In a phase I/II trial, enrichment of leukaemia-specific donor T cells was performed ex vivo using leukaemia-derived host antigen-presenting cells. These T cells were administered to fight AML relapse after allo-HSCT as alternative treatment for DLI [143]. In one AML patient, stable disease was detected 8 weeks after T cell infusion, while two other patients had no clinical response. The authors concluded that further optimization was required, because the procedure to generate leukaemia-reactive T cells took so long to move from bench to bedside that leukaemia patients with rapidly expanding disease often succumbed before therapy administration was possible [143]. In a study by the group of Quesenberry [144], a more rapid protocol was developed for refractory AML patients, where haploidentical T cells were administered following a minimal preparative treatment of 100 cGy (nonengrafting haploidentical transplantation with minimal myeloablation). In the group of 13 AML patients, 5 durable complete responses and 4 partial responses were seen, showing that long-term response can be found in transient chimerism. Before infusion, cells were primed with granulocyte colony-stimulating factor (G-CSF) to minimize GVHD by polarizing T cells to a Th2 phenotype.

The generation of CD4+ or CD8+ T cells with anti-leukaemia reactivity for adoptive transfer in AML is also scrutinized by others [28, 29, 145156]. Until now, clinical efficacy of adoptively transferred T cells has been shown mainly in the allograft setting [157]. Nevertheless, transfer of autologous T cells is an interesting concept to separate GVL reactions from GVHD. Currently, genetic engineering of T cells (described in depth by Vera et al. [158]) is being investigated in an attempt to overcome the difficulty of generating high numbers of leukaemia-reactive autologous T cells in vitro. A promising strategy is the isolation of genes encoding α and β T cell receptor (TCR) chains from LAA-specific T cell clones with high avidity, using them to redirect the specificity of primary T cells. Both WT1-specific and HA-1/2-specific TCR genes have been actively investigated for their anti-leukaemia reactivity in preclinical experiments following transduction of T cells. Explorative clinical trials are now being planned [157, 159]. Toxicity of this approach will depend on distribution of the target antigen in normal tissues. For WT1, specific attention should therefore be paid to the hematopoietic system.

Also, chimeric antigen receptors (CARs) can redirect T cell specificity. By transducing T cells with CARs, antigen specificity of an antibody (single-chain variable fragment) is coupled to the lytic capacity of T cells (transmembrane and intracytoplasmatic structure of CD3ζ) [158]. Advantages are the MHC independency and the extended range of tumour surface antigens that can be recognized by T cells. If CARs are linked to endodomains of the costimulatory molecules 4-1BB and CD28, T cells can obtain a higher activation status. For AML, CAR targeting the Lewis Y antigen had promising effects in preclinical experiments, paving the way for a phase I clinical trial [160]. In vivo toxicity of this approach, related to the potential of CAR-modified T cells to damage normal tissues, is a specific concern that was recently fed by reports on cholestase development (renal cell carcinoma patient [161]) and renal and respiratory failure (B-cell chronic lymphocytic leukaemia patient [162]).

Important characteristics of T cells that determine the success of adoptive transfer are the capacities to survive in vivo and to home to the sites with residual leukaemic cells. Although not all signals that attract T cells to cancer cell sites have been characterized yet, it is clear that expression of adhesion molecules and chemokine receptors on T cells will determine their capacity to migrate. Important chemokines for T cell attraction that can be secreted by AML cells are CXCL10, CCL5, CCL17 and CCL22 [43, 44, 85, 86]. Also, marrow stromal cells can contribute by secreting SDF1 to attract T cells that express the chemokine receptor CXCR4 [163]. In order to attract T cells to the bone marrow, the chemotactic gradient that is determined by the difference between chemokine concentrations in the bone marrow and in the serum must be in favour of the bone marrow [85]. In this regard, Olsnes et al. [43] showed that serum levels of CCL5, CCL17 and CXCL10 vary between AML patients and that interaction between AML cells and the stromal cells can raise the release of CCL17 and CXCL10, supporting the attraction of T cells to the AML sites. Homing capacities could be augmented by genetic transfer of chemokine receptors into T cells before adoptive transfer [158, 164].

Genetic engineering could also be used to increase the survival and functionality of adoptively transferred T cells by codelivery of genes encoding cytokines, costimulatory molecules and/or telomerase reverse transcriptase [158]. On the other hand, suicide genes could be introduced if transferred T cells can potentially harm normal tissues. Examples of suicide genes include thymidine kinase from herpes simplex virus I and Fas [158].

Adoptive transfer of NK cells

NK cells have the ability to kill autologous and allogeneic tumour cells [165, 166]. NK cell-mediated alloreactivity is mediated by killer immunoglobulin-like receptors (KIR), expressed by NK cells and recognizing subgroups of HLA class I molecules. Mismatches in binding of donor KIR with recipient HLA class I molecules triggers donor NK cells to kill recipient mismatched cells [167]. This mechanism has been shown to contribute substantially to GVL effects of HSCT with HLA-haplo-identical transplants from siblings. In this way, NK cell-mediated alloreactive responses can reduce relapse risk, while enhancing engraftment and protecting against GVHD, resulting in improved outcome for AML patients [165, 168170].

Aiming at increasing the clinical efficacy of allo-reactive NK cells, research is now focusing on the expansion of NK cells in vivo and ex vivo [165]. In a phase I study, enriched NK cell fractions from haplo-identical donors were transferred after activation with IL-2 and expanded in vivo following an intensive preparative regimen, associated with high levels of IL-15. Three out of four AML patients achieved CR following KIR ligand-mismatched NK cell transplantation [171]. In a recent phase I study, haplo-identical KIR-mismatched NK cells were transplanted in ten children with AML in combination with IL-2 following low-dose immunosuppression [172]. The purified cell product was injected without ex vivo expansion or activation. Significant in vivo expansion of NK cells could be detected, and all ten patients remained in remission during follow-up which ranged from 18 to 39 months.

By expanding NK cells ex vivo, AML patients can receive transplantations with high numbers of purified NK cell fractions. Currently, protocols are being optimized for ex vivo expansion of NK cells able to kill leukaemic cells [173, 174] and phase I clinical trials are ongoing [173, 175]. An initial trial involving one child with AML showed feasibility of NK cell isolation and expansion for clinical applications [175].

It has not been specified yet which chemokines are involved in the attraction of adoptively transferred NK cells to the bone marrow of AML patients. Nevertheless, NK cells express several chemokine receptors, so different chemokines might be involved [176178]. Immunocompetent cells, such as T cells and NK cells, might secrete factors that contribute to cancer growth and/or angiogenesis [177]. Furthermore, investigators should be aware of possible toxic effects of adoptive transfer, e.g. on hematopoiesis. Mesenchyal stromal cells (MSC) are important in the support of hematopoiesis and infusion of MSC is currently being investigated to aid hematopoietic recovery following allo-HSCT [179, 180]. Götherström et al. [181] recently showed that human IL-2-activated NK cells could kill foetal and adult MSC, as well as autologous MSC, so caution will be needed when transferring activated NK cells. On the other hand, the importance of NK cells in controlling AML is illustrated by the fact that relapse in AML patients following allo-HSCT is associated with decreased numbers of NK cells 1 year post-transplant [182]. Therefore, adoptive transfer of NK cells has the potential to become an important future cell therapy strategy to prevent AML relapse.

Conclusions

Knowledge has been gained in recent years on the importance of disease status when applying cancer immunotherapy. Early studies focused on the treatment of advanced disease with a significant tumour bulk. By further understanding the mechanisms of tumour-induced immunosuppression, it is now clear that the immune system may not be able to effectively induce regression of large tumour burdens [183]. Reduced disease loads or immunotherapy treatments in combination with conventional tumour burden-reducing induction therapies may provide more favourable settings to apply therapeutic immunotherapy. In AML, immunotherapy is predominantly investigated to prevent relapse following standard therapy. Ideally, multiple antigenic epitopes and potent adjuvants would be incorporated to induce both innate and adaptive immune responses in order to break AML-mediated immunosuppressive mechanisms and immunological tolerance. Recently, promising progress has been made in the development of cellular immunotherapeutic protocols to prevent or to fight AML relapse.

Therapeutic vaccination using whole tumour cell or DC vaccines and adoptive transfer of NK cells with anti-leukaemic activity have been investigated in phase I or II clinical trials. Positive results associated with the application of these methods include decreased levels of the biomarker WT1, induction of remission (molecular and haematological) and/or increased relapse-free survival. These approaches are now being further evaluated in larger cohorts. Because of the observed GVL effect following DLI and the positive results with adoptively transferred T cells in metastatic melanoma [184], the adoptive transfer of leukaemia-specific T cells is now considered to be worthy of investigation. New and ongoing clinical trials will reveal whether this technique could provide a benefit in AML patients, with genetic engineering of T cells giving a boost to this research area.

Until now, not much attention has been paid to combination strategies. It is important to investigate whether the anti-leukaemic effects of cellular immunotherapeutic approaches could be further increased through the combined administration with immunomodulatory molecules, such as TLR ligands or ipilimumab. Toll-like receptor ligands can activate multiple cells involved in anti-tumour responses simultaneously [185]. Ipilimumab is an antibody currently under active investigation in clinical trials to overcome CTLA-4-mediated immunosuppression [186]. Recent insights have underscored the role of chemokines in the immunoregulatory network formed by immunocompetent cells, bone marrow stromal cells and AML cells [177], making these mediators and their receptors also attractive targets for immunomodulation. Due to the complexity of immunity and the need to overcome anergy to self-antigen-expressing tumour cells, likely personalized therapies and combination strategies are required to further improve the curative potential of cellular therapies, ultimately resulting in improved survival of AML patients.

Acknowledgments

E.S. is postdoctoral researcher of the Research Foundation Flanders (FWO-Vlaanderen), C.L. was funded by EBMT, N.H. and B.G. were funded by Leukaemia & Lymphoma Research and S.B. by Cancer Research U.K. This work was supported in part by research grants of the FWO-Vlaanderen (G.0082.08), the Belgian Foundation against Cancer, and the Methusalem program of the Flemish Government.

Abbreviations

AML

Acute myeloid leukaemia

CR

Complete remission

CTL

Cytotoxic T lymphocyte

DC

Dendritic cell

DLI

Donor leucocyte infusion

FAB

French-American-British

GVHD

Graft-versus-host-disease

GVL

Graft-versus-leukaemia

HLA

Human leucocyte antigen

HSCT

Haematopoietic stem cell transplantation

LAA

Leukaemia-associated antigen

LSA

Leukaemia-specific antigen

mHAg

Minor histocompatibility antigen

MHC

Major histocompatibility complex

MRD

Minimal residual disease

NK

Natural killer

poly(I:C)

Polyinosinic polycytidylic acid

PR3

Proteinase 3

RHAMM

Receptor for hyaluronic acid-mediated motility

SSX2IP

Synovial sarcoma X breakpoint 2-interacting protein

TAA

Tumour-associated antigen

TLR

Toll-like receptor

WHO

World Health Organization

WT1

Wilms’ tumour protein

References

  • 1.Rubnitz JE, Gibson B, Smith FO. Acute myeloid leukemia. Hematol Oncol Clin North Am. 2010;24:35–63. doi: 10.1016/j.hoc.2009.11.008. [DOI] [PubMed] [Google Scholar]
  • 2.Stone RM, O’Donnell MR, Sekeres MA (2004) Acute myeloid leukemia. Hematol Am Soc Hematol Educ Program 98–117 [DOI] [PubMed]
  • 3.O’Donnell MR, Abboud CN, Altman J, et al. Acute myeloid leukemia. J Natl Compr Canc Netw. 2010;9:280–317. doi: 10.6004/jnccn.2011.0027. [DOI] [PubMed] [Google Scholar]
  • 4.Hamadani M, Awan FT, Elder P, et al. Allogeneic hematopoietic stem cell transplantation for peripheral T cell lymphomas; evidence of graft-versus-T cell lymphoma effect. Biol Blood Marrow Transplant. 2008;14:480–483. doi: 10.1016/j.bbmt.2008.01.002. [DOI] [PubMed] [Google Scholar]
  • 5.Soiffer RJ. Donor lymphocyte infusions for acute myeloid leukaemia. Best Pract Res Clin Haematol. 2008;21:455–466. doi: 10.1016/j.beha.2008.07.009. [DOI] [PubMed] [Google Scholar]
  • 6.Hamadani M, Awan FT, Copelan EA. Hematopoietic stem cell transplantation in adults with acute myeloid leukemia. Biol Blood Marrow Transplant. 2008;14:556–567. doi: 10.1016/j.bbmt.2008.02.019. [DOI] [PubMed] [Google Scholar]
  • 7.Lodewyck T, Cornelissen JJ. Allogeneic stem cell transplantation in acute myeloid leukemia: a risk-adapted approach. Blood Rev. 2008;22:293–302. doi: 10.1016/j.blre.2008.03.008. [DOI] [PubMed] [Google Scholar]
  • 8.Storb R. Reduced-intensity conditioning transplantation in myeloid malignancies. Curr Opin Oncol. 2009;21:S3–S5. doi: 10.1097/01.cco.0000357467.45843.ba. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Marmont AM, Horowitz MM, Gale RP, et al. T-cell depletion of HLA-identical transplants in leukemia. Blood. 1991;78:2120–2130. [PubMed] [Google Scholar]
  • 10.O’Donnell PV, Luznik L, Jones RJ, et al. Nonmyeloablative bone marrow transplantation from partially HLA-mismatched related donors using posttransplantation cyclophosphamide. Biol Blood Marrow Transplant. 2002;8:377–386. doi: 10.1053/bbmt.2002.v8.pm12171484. [DOI] [PubMed] [Google Scholar]
  • 11.Luznik L, Bolanos-Meade J, Zahurak M, et al. High-dose cyclophosphamide as single-agent, short-course prophylaxis of graft-versus-host disease. Blood. 2010;115:3224–3230. doi: 10.1182/blood-2009-11-251595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Luznik L, O’Donnell PV, Symons HJ, et al. HLA-haploidentical bone marrow transplantation for hematologic malignancies using nonmyeloablative conditioning and high-dose, posttransplantation cyclophosphamide. Biol Blood Marrow Transplant. 2008;14:641–650. doi: 10.1016/j.bbmt.2008.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Rezvani K, Barrett AJ. Characterizing and optimizing immune responses to leukaemia antigens after allogeneic stem cell transplantation. Best Pract Res Clin Haematol. 2008;21:437–453. doi: 10.1016/j.beha.2008.07.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Smits EL, Berneman ZN, Van Tendeloo VF. Immunotherapy of acute myeloid leukemia: current approaches. Oncologist. 2009;14:240–252. doi: 10.1634/theoncologist.2008-0165. [DOI] [PubMed] [Google Scholar]
  • 15.Schmitt M, Casalegno-Garduno R, Xu X, et al. Peptide vaccines for patients with acute myeloid leukemia. Expert Rev Vaccines. 2009;8:1415–1425. doi: 10.1586/erv.09.90. [DOI] [PubMed] [Google Scholar]
  • 16.Barrett AJ, Le Blanc K. Immunotherapy prospects for acute myeloid leukaemia. Clin Exp Immunol. 2010;161:223–232. doi: 10.1111/j.1365-2249.2010.04197.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Liseth K, Ersvaer E, Hervig T, et al. Combination of intensive chemotherapy and anticancer vaccines in the treatment of human malignancies: the hematological experience. J Biomed Biotechnol. 2010;2010:692097. doi: 10.1155/2010/692097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Thoren FB, Romero AI, Brune M, et al. Histamine dihydrochloride and low-dose interleukin-2 as post-consolidation immunotherapy in acute myeloid leukemia. Expert Opin Biol Ther. 2009;9:1217–1223. doi: 10.1517/14712590903130566. [DOI] [PubMed] [Google Scholar]
  • 19.Pegram HJ, Andrews DM, Smyth MJ, et al. Activating and inhibitory receptors of natural killer cells. Immunol Cell Biol. 2011;89:216–224. doi: 10.1038/icb.2010.78. [DOI] [PubMed] [Google Scholar]
  • 20.Cheever MA, Allison JP, Ferris AS, et al. The prioritization of cancer antigens: a national cancer institute pilot project for the acceleration of translational research. Clin Cancer Res. 2009;15:5323–5337. doi: 10.1158/1078-0432.CCR-09-0737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Chan L, Hardwick NR, Guinn BA, et al. An immune edited tumour versus a tumour edited immune system: Prospects for immune therapy of acute myeloid leukaemia. Cancer Immunol Immunother. 2006;55:1017–1024. doi: 10.1007/s00262-006-0129-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Bruserud O, Ulvestad E, Berentsen S, et al. T-lymphocyte functions in acute leukaemia patients with severe chemotherapy-induced cytopenia: characterization of clonogenic T-cell proliferation. Scand J Immunol. 1998;47:54–62. doi: 10.1046/j.1365-3083.1998.00254.x. [DOI] [PubMed] [Google Scholar]
  • 23.Behl D, Porrata LF, Markovic SN, et al. Absolute lymphocyte count recovery after induction chemotherapy predicts superior survival in acute myelogenous leukemia. Leukemia. 2006;20:29–34. doi: 10.1038/sj.leu.2404032. [DOI] [PubMed] [Google Scholar]
  • 24.Barrett AJ, Savani BN. Does chemotherapy modify the immune surveillance of hematological malignancies? Leukemia. 2009;23:53–58. doi: 10.1038/leu.2008.273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Scheibenbogen C, Letsch A, Thiel E, et al. CD8 T-cell responses to Wilms tumor gene product WT1 and proteinase 3 in patients with acute myeloid leukemia. Blood. 2002;100:2132–2137. doi: 10.1182/blood-2002-01-0163. [DOI] [PubMed] [Google Scholar]
  • 26.Greiner J, Li L, Ringhoffer M, Barth TF, et al. Identification and characterization of epitopes of the receptor for hyaluronic acid-mediated motility (RHAMM/CD168) recognized by CD8+ T cells of HLA-A2-positive patients with acute myeloid leukemia. Blood. 2005;106:938–945. doi: 10.1182/blood-2004-12-4787. [DOI] [PubMed] [Google Scholar]
  • 27.Rezvani K, Brenchley JM, Price DA, et al. T-cell responses directed against multiple HLA-A*0201-restricted epitopes derived from Wilms’ tumor 1 protein in patients with leukemia and healthy donors: identification, quantification, and characterization. Clin Cancer Res. 2005;11:8799–8807. doi: 10.1158/1078-0432.CCR-05-1314. [DOI] [PubMed] [Google Scholar]
  • 28.Oka Y, Elisseeva OA, Tsuboi A, et al. Human cytotoxic T-lymphocyte responses specific for peptides of the wild-type Wilms’ tumor gene (WT1) product. Immunogenetics. 2000;51:99–107. doi: 10.1007/s002510050018. [DOI] [PubMed] [Google Scholar]
  • 29.Ohminami H, Yasukawa M, Fujita S. HLA class I-restricted lysis of leukemia cells by a CD8(+) cytotoxic T-lymphocyte clone specific for WT1 peptide. Blood. 2000;95:286–293. [PubMed] [Google Scholar]
  • 30.Rezvani K. PR1 vaccination in myeloid malignancies. Expert Rev Vaccines. 2008;7:867–875. doi: 10.1586/14760584.7.7.867. [DOI] [PubMed] [Google Scholar]
  • 31.Greiner J, Schmitt M, Li L, et al. Expression of tumor-associated antigens in acute myeloid leukemia: Implications for specific immunotherapeutic approaches. Blood. 2006;108:4109–4117. doi: 10.1182/blood-2006-01-023127. [DOI] [PubMed] [Google Scholar]
  • 32.Greiner J, Dohner H, Schmitt M. Cancer vaccines for patients with acute myeloid leukemia–definition of leukemia-associated antigens and current clinical protocols targeting these antigens. Haematologica. 2006;91:1653–1661. [PubMed] [Google Scholar]
  • 33.Guinn BA, Tobal K, Mills KI. Comparison of the survival implications of tumour-associated versus cancer-testis antigen expression in acute myeloid leukaemia. Br J Haematol. 2007;136:510–512. doi: 10.1111/j.1365-2141.2006.06454.x. [DOI] [PubMed] [Google Scholar]
  • 34.Guinn BA, Mohamedali A, Mills KI, et al. Leukemia associated antigens: their dual role as biomarkers and immunotherapeutic targets for acute myeloid leukemia. Biomark Insights. 2007;2:69–79. [PMC free article] [PubMed] [Google Scholar]
  • 35.Greiner J, Bullinger L, Guinn BA, et al. Leukemia-associated antigens are critical for the proliferation of acute myeloid leukemia cells. Clin Cancer Res. 2008;14:7161–7166. doi: 10.1158/1078-0432.CCR-08-1102. [DOI] [PubMed] [Google Scholar]
  • 36.Greiner J, Schmitt M. Leukemia-associated antigens as target structures for a specific immunotherapy in chronic myeloid leukemia. Eur J Haematol. 2008;80:461–468. doi: 10.1111/j.1600-0609.2008.01053.x. [DOI] [PubMed] [Google Scholar]
  • 37.Guinn BA, Bullinger L, Thomas NS, et al. SSX2IP expression in acute myeloid leukaemia: an association with mitotic spindle failure in t(8;21), and cell cycle in t(15;17) patients. Br J Haematol. 2008;140:250–251. doi: 10.1111/j.1365-2141.2007.06892.x. [DOI] [PubMed] [Google Scholar]
  • 38.Yee C, Thompson JA, Roche P, et al. Melanocyte destruction after antigen-specific immunotherapy of melanoma: direct evidence of t cell-mediated vitiligo. J Exp Med. 2000;192:1637–1644. doi: 10.1084/jem.192.11.1637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Oka Y, Tsuboi A, Taguchi T, et al. Induction of WT1 (Wilms’ tumor gene)-specific cytotoxic T lymphocytes by WT1 peptide vaccine and the resultant cancer regression. Proc Natl Acad Sci USA. 2004;101:13885–13890. doi: 10.1073/pnas.0405884101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Oka Y, Sugiyama H. WT1 peptide vaccine, one of the most promising cancer vaccines: its present status and the future prospects. Immunotherapy. 2010;2:591–594. doi: 10.2217/imt.10.58. [DOI] [PubMed] [Google Scholar]
  • 41.Bruserud O, Ulvestad E. Acute myelogenous leukemia blasts as accessory cells during in vitro T lymphocyte activation. Cell Immunol. 2000;206:36–50. doi: 10.1006/cimm.2000.1725. [DOI] [PubMed] [Google Scholar]
  • 42.Bruserud O. Acute myelogenous leukemia blasts as accessory cells during T lymphocyte activation: possible implications for future therapeutic strategies. Leukemia. 1999;13:1175–1187. doi: 10.1038/sj.leu.2401452. [DOI] [PubMed] [Google Scholar]
  • 43.Olsnes AM, Motorin D, Ryningen A, et al. T lymphocyte chemotactic chemokines in acute myelogenous leukemia (AML): local release by native human AML blasts and systemic levels of CXCL10 (IP-10), CCL5 (RANTES) and CCL17 (TARC) Cancer Immunol Immunother. 2006;55:830–840. doi: 10.1007/s00262-005-0080-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Bruserud O, Ryningen A, Olsnes AM, et al. Subclassification of patients with acute myelogenous leukemia based on chemokine responsiveness and constitutive chemokine release by their leukemic cells. Haematologica. 2007;92:332–341. doi: 10.3324/haematol.10148. [DOI] [PubMed] [Google Scholar]
  • 45.Bruserud O, Frostad S, Foss B. In vitro culture of acute myelogenous leukemia blasts: a comparison of four different culture media. J Hematother. 1999;8:63–73. doi: 10.1089/106161299320587. [DOI] [PubMed] [Google Scholar]
  • 46.Bruserud O, Gjertsen BT, von Volkman HL. In vitro culture of human acute myelogenous leukemia (AML) cells in serum-free media: studies of native AML blasts and AML cell lines. J Hematother Stem Cell Res. 2000;9:923–932. doi: 10.1089/152581600750062372. [DOI] [PubMed] [Google Scholar]
  • 47.Bruserud O, Ulvestad E. Effects of gamma-irradiation on acute myelogenous leukemia blasts: in vitro studies of proliferation, constitutive cytokine secretion, and accessory cell function during T cell activation. J Hematother Stem Cell Res. 1999;8:431–441. doi: 10.1089/152581699320199. [DOI] [PubMed] [Google Scholar]
  • 48.Powles RL, Balchin LA, Fairley GH, et al. Recognition of leukaemia cells as foreign before and after autoimmunization. Br Med J. 1971;1:486–489. doi: 10.1136/bmj.1.5747.486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Powles RL, Russell J, Lister TA, et al. Immunotherapy for acute myelogenous leukaemia: a controlled clinical study 2 1/2 years after entry of the last patient. Br J Cancer. 1977;35:265–272. doi: 10.1038/bjc.1977.38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Zhang WG, Liu SH, Cao XM, et al. A phase-I clinical trial of active immunotherapy for acute leukemia using inactivated autologous leukemia cells mixed with IL-2, GM-CSF, and IL-6. Leuk Res. 2005;29:3–9. doi: 10.1016/j.leukres.2004.04.015. [DOI] [PubMed] [Google Scholar]
  • 51.Van Tendeloo VF, Van Broeckhoven C, Berneman ZN. Gene-based cancer vaccines: an ex vivo approach. Leukemia. 2001;15:545–558. doi: 10.1038/sj.leu.2402069. [DOI] [PubMed] [Google Scholar]
  • 52.Cignetti A, Guarini A, Carbone A, et al. Transduction of the IL2 gene into human acute leukemia cells: induction of tumor rejection without modifying cell proliferation and IL2 receptor expression. J Natl Cancer Inst. 1994;86:785–791. doi: 10.1093/jnci/86.10.785. [DOI] [PubMed] [Google Scholar]
  • 53.Matulonis U, Dosiou C, Freeman G, et al. B7–1 is superior to B7–2 costimulation in the induction and maintenance of T cell-mediated antileukemia immunity. Further evidence that B7–1 and B7–2 are functionally distinct. J Immunol. 1996;156:1126–1131. [PubMed] [Google Scholar]
  • 54.Nakazaki Y, Tani K, Lin ZT, et al. Vaccine effect of granulocyte-macrophage colony-stimulating factor or CD80 gene-transduced murine hematopoietic tumor cells and their cooperative enhancement of antitumor immunity. Gene Ther. 1998;5:1355–1362. doi: 10.1038/sj.gt.3300726. [DOI] [PubMed] [Google Scholar]
  • 55.Dunussi-Joannopoulos K, Dranoff G, Weinstein HJ, et al. Gene immunotherapy in murine acute myeloid leukemia: granulocyte-macrophage colony-stimulating factor tumor cell vaccines elicit more potent antitumor immunity compared with B7 family and other cytokine vaccines. Blood. 1998;91:222–230. [PubMed] [Google Scholar]
  • 56.Dunussi-Joannopoulos K, Weinstein HJ, Nickerson PW, et al. Irradiated B7–1 transduced primary acute myelogenous leukemia (AML) cells can be used as therapeutic vaccines in murine AML. Blood. 1996;87:2938–2946. [PubMed] [Google Scholar]
  • 57.Mutis T, Schrama E, Melief CJ, et al. CD80-Transfected acute myeloid leukemia cells induce primary allogeneic T-cell responses directed at patient specific minor histocompatibility antigens and leukemia-associated antigens. Blood. 1998;92:1677–1684. [PubMed] [Google Scholar]
  • 58.Schakowski F, Buttgereit P, Mazur M, et al. Novel non-viral method for transfection of primary leukemia cells and cell lines. Genet Vaccines Ther. 2004;2:1. doi: 10.1186/1479-0556-2-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Hirst WJ, Buggins A, Darling D, et al. Enhanced immune costimulatory activity of primary acute myeloid leukaemia blasts after retrovirus-mediated gene transfer of B7.1. Gene Ther. 1997;4:691–699. doi: 10.1038/sj.gt.3300437. [DOI] [PubMed] [Google Scholar]
  • 60.Roddie PH, Paterson T, Turner ML. Gene transfer to primary acute myeloid leukaemia blasts and myeloid leukaemia cell lines. Cytokines Cell Mol Ther. 2000;6:127–134. doi: 10.1080/mccm.6.3.127.134. [DOI] [PubMed] [Google Scholar]
  • 61.Wattel E, Vanrumbeke M, Abina MA, et al. Differential efficacy of adenoviral mediated gene transfer into cells from hematological cell lines and fresh hematological malignancies. Leukemia. 1996;10:171–174. [PubMed] [Google Scholar]
  • 62.Howard DS, Rizzierri DA, Grimes B, et al. Genetic manipulation of primitive leukemic and normal hematopoietic cells using a novel method of adenovirus-mediated gene transfer. Leukemia. 1999;13:1608–1616. doi: 10.1038/sj/leu/2401541. [DOI] [PubMed] [Google Scholar]
  • 63.Saudemont A, Corm S, Wickham T, et al. Induction of leukemia-specific CD8+ cytotoxic T cells with autologous myeloid leukemic cells maturated with a fiber-modified adenovirus encoding TNF-alpha. Mol Ther. 2005;11:950–959. doi: 10.1016/j.ymthe.2004.12.016. [DOI] [PubMed] [Google Scholar]
  • 64.Dilloo D, Rill D, Entwistle C, et al. A novel herpes vector for the high-efficiency transduction of normal and malignant human hematopoietic cells. Blood. 1997;89:119–127. [PubMed] [Google Scholar]
  • 65.Anderson R, Macdonald I, Corbett T, et al. Construction and biological characterization of an interleukin-12 fusion protein (Flexi-12): delivery to acute myeloid leukemic blasts using adeno-associated virus. Hum Gene Ther. 1997;8:1125–1135. doi: 10.1089/hum.1997.8.9-1125. [DOI] [PubMed] [Google Scholar]
  • 66.Gonzalez R, Vereecque R, Wickham TJ, et al. Increased gene transfer in acute myeloid leukemic cells by an adenovirus vector containing a modified fiber protein. Gene Ther. 1999;6:314–320. doi: 10.1038/sj.gt.3300836. [DOI] [PubMed] [Google Scholar]
  • 67.Stripecke R, Cardoso AA, Pepper KA, et al. Lentiviral vectors for efficient delivery of CD80 and granulocyte-macrophage- colony-stimulating factor in human acute lymphoblastic leukemia and acute myeloid leukemia cells to induce antileukemic immune responses. Blood. 2000;96:1317–1326. [PubMed] [Google Scholar]
  • 68.Biagi E, Bambacioni F, Gaipa G, et al. Efficient lentiviral transduction of primary human acute myelogenous and lymphoblastic leukemia cells. Haematologica. 2001;86:13–16. [PubMed] [Google Scholar]
  • 69.Koya RC, Kasahara N, Pullarkat V, et al. Transduction of acute myeloid leukemia cells with third generation self-inactivating lentiviral vectors expressing CD80 and GM-CSF: effects on proliferation, differentiation, and stimulation of allogeneic and autologous anti-leukemia immune responses. Leukemia. 2002;16:1645–1654. doi: 10.1038/sj.leu.2402582. [DOI] [PubMed] [Google Scholar]
  • 70.Heinzinger NK, Bukinsky MI, Haggerty SA, et al. The Vpr protein of human immunodeficiency virus type 1 influences nuclear localization of viral nucleic acids in nondividing host cells. Proc Natl Acad Sci USA. 1994;91:7311–7315. doi: 10.1073/pnas.91.15.7311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Koya RC, Weber JS, Kasahara N, et al. Making dendritic cells from the inside out: lentiviral vector-mediated gene delivery of granulocyte-macrophage colony-stimulating factor and interleukin 4 into CD14+ monocytes generates dendritic cells in vitro. Hum Gene Ther. 2004;15:733–748. doi: 10.1089/1043034041648381. [DOI] [PubMed] [Google Scholar]
  • 72.Gaken J, Jiang J, Daniel K, et al. Fusagene vectors: a novel strategy for the expression of multiple genes from a single cistron. Gene Ther. 2000;7:1979–1985. doi: 10.1038/sj.gt.3301341. [DOI] [PubMed] [Google Scholar]
  • 73.Chan L, Hardwick N, Darling D, et al. IL-2/B7.1 (CD80) fusagene transduction of AML blasts by a self-inactivating lentiviral vector stimulates T cell responses in vitro: a strategy to generate whole cell vaccines for AML. Mol Ther. 2005;11:120–131. doi: 10.1016/j.ymthe.2004.09.006. [DOI] [PubMed] [Google Scholar]
  • 74.Ingram W, Chan L, Guven H, et al. Human CD80/IL2 lentivirus-transduced acute myeloid leukaemia (AML) cells promote natural killer (NK) cell activation and cytolytic activity: implications for a phase I clinical study. Br J Haematol. 2009;145:749–760. doi: 10.1111/j.1365-2141.2009.07684.x. [DOI] [PubMed] [Google Scholar]
  • 75.Hardwick N, Chan L, Ingram W, et al. Lytic activity against primary AML cells is stimulated in vitro by an autologous whole cell vaccine expressing IL-2 and CD80. Cancer Immunol Immunother. 2010;59:379–388. doi: 10.1007/s00262-009-0756-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Borrello IM, Levitsky HI, Stock W, et al. Granulocyte-macrophage colony-stimulating factor (GM-CSF)-secreting cellular immunotherapy in combination with autologous stem cell transplantation (ASCT) as postremission therapy for acute myeloid leukemia (AML) Blood. 2009;114:1736–1745. doi: 10.1182/blood-2009-02-205278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Koos D, Josephs SF, Alexandrescu DT, et al. Tumor vaccines in 2010: need for integration. Cell Immunol. 2010;263:138–147. doi: 10.1016/j.cellimm.2010.03.019. [DOI] [PubMed] [Google Scholar]
  • 78.Smits EL, Ponsaerts P, Van de Velde AL, et al. Proinflammatory response of human leukemic cells to dsRNA transfection linked to activation of dendritic cells. Leukemia. 2007;21:1691–1699. doi: 10.1038/sj.leu.2404763. [DOI] [PubMed] [Google Scholar]
  • 79.Kawai T, Akira S. The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nat Immunol. 2010;11:373–384. doi: 10.1038/ni.1863. [DOI] [PubMed] [Google Scholar]
  • 80.Dunn GP, Koebel CM, Schreiber RD. Interferons, immunity and cancer immunoediting. Nat Rev Immunol. 2006;6:836–848. doi: 10.1038/nri1961. [DOI] [PubMed] [Google Scholar]
  • 81.Ferrantini M, Capone I, Belardelli F. Interferon-alpha and cancer: mechanisms of action and new perspectives of clinical use. Biochimie. 2007;89:884–893. doi: 10.1016/j.biochi.2007.04.006. [DOI] [PubMed] [Google Scholar]
  • 82.Lion E, Smits EL, Berneman ZN, et al. Acute myeloid leukemic cell lines loaded with synthetic dsRNA trigger IFN-gamma secretion by human NK cells. Leuk Res. 2009;33:539–546. doi: 10.1016/j.leukres.2008.08.020. [DOI] [PubMed] [Google Scholar]
  • 83.Lion E, Smits EL, Berneman ZN, et al. Quantification of IFN-gamma produced by human purified NK cells following tumor cell stimulation: comparison of three IFN-gamma assays. J Immunol Methods. 2009;350:89–96. doi: 10.1016/j.jim.2009.08.014. [DOI] [PubMed] [Google Scholar]
  • 84.Smits EL, Cools N, Lion E, et al. The Toll-like receptor 7/8 agonist resiquimod greatly increases the immunostimulatory capacity of human acute myeloid leukemia cells. Cancer Immunol Immunother. 2010;59:35–46. doi: 10.1007/s00262-009-0721-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Olsnes AM, Hatfield KJ, Bruserud O. The chemokine system and its contribution to leukemogenesis and treatment responsiveness in patients with acute myeloid leukemia. J BUON. 2009;14:S131–S140. [PubMed] [Google Scholar]
  • 86.Olsnes AM, Ryningen A, Ersvaer E, et al. In vitro induction of a dendritic cell phenotype in primary human acute myelogenous leukemia (AML) blasts alters the chemokine release profile and increases the levels of T cell chemotactic CCL17 and CCL22. J Interferon Cytokine Res. 2008;28:297–310. doi: 10.1089/jir.2007.0052. [DOI] [PubMed] [Google Scholar]
  • 87.Le Dieu R, Taussig DC, Ramsay AG, et al. Peripheral blood T cells in acute myeloid leukemia (AML) patients at diagnosis have abnormal phenotype and genotype and form defective immune synapses with AML blasts. Blood. 2009;114(18):3909–3916. doi: 10.1182/blood-2009-02-206946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Szczepanski MJ, Szajnik M, Czystowska M, et al. Increased frequency and suppression by regulatory T cells in patients with acute myelogenous leukemia. Clin Cancer Res. 2009;15(10):3325–3332. doi: 10.1158/1078-0432.CCR-08-3010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Cools N, Ponsaerts P, Van Tendeloo VF, et al. Balancing between immunity and tolerance: an interplay between dendritic cells, regulatory T cells, and effector T cells. J Leukoc Biol. 2007;82:1365–1374. doi: 10.1189/jlb.0307166. [DOI] [PubMed] [Google Scholar]
  • 90.Robson NC, Hoves S, Maraskovsky E, et al. Presentation of tumour antigens by dendritic cells and challenges faced. Curr Opin Immunol. 2010;22:137–144. doi: 10.1016/j.coi.2010.01.002. [DOI] [PubMed] [Google Scholar]
  • 91.Smits EL, Anguille S, Cools N, et al. Dendritic cell-based cancer gene therapy. Hum Gene Ther. 2009;20:1106–1118. doi: 10.1089/hum.2009.145. [DOI] [PubMed] [Google Scholar]
  • 92.Romani N, Gruner S, Brang D, et al. Proliferating dendritic cell progenitors in human blood. J Exp Med. 1994;180:83–93. doi: 10.1084/jem.180.1.83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Steinman RM, Banchereau J. Taking dendritic cells into medicine. Nature. 2007;449:419–426. doi: 10.1038/nature06175. [DOI] [PubMed] [Google Scholar]
  • 94.Anguille S, Smits EL, Cools N, et al. Short-term cultured, interleukin-15 differentiated dendritic cells have potent immunostimulatory properties. J Transl Med. 2009;7:109. doi: 10.1186/1479-5876-7-109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Brouwer RE, van der Hoorn M, Kluin-Nelemans HC, et al. The generation of dendritic-like cells with increased allostimulatory function from acute myeloid leukemia cells of various FAB subclasses. Hum Immunol. 2000;61:565–574. doi: 10.1016/S0198-8859(00)00111-7. [DOI] [PubMed] [Google Scholar]
  • 96.Li L, Reinhardt P, Schmitt A, et al. Dendritic cells generated from acute myeloid leukemia (AML) blasts maintain the expression of immunogenic leukemia associated antigens. Cancer Immunol Immunother. 2005;54:685–693. doi: 10.1007/s00262-004-0631-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Hicks C, Cheung C, Lindeman R. Restimulation of tumour-specific immunity in a patient with AML following injection with B7-1 positive autologous blasts. Leuk Res. 2003;27:1051–1061. doi: 10.1016/S0145-2126(03)00058-4. [DOI] [PubMed] [Google Scholar]
  • 98.Houtenbos I, Westers TM, Ossenkoppele GJ, et al. Feasibility of clinical dendritic cell vaccination in acute myeloid leukemia. Immunobiology. 2006;211:677–685. doi: 10.1016/j.imbio.2006.05.013. [DOI] [PubMed] [Google Scholar]
  • 99.Li L, Giannopoulos K, Reinhardt P, et al. Immunotherapy for patients with acute myeloid leukemia using autologous dendritic cells generated from leukemic blasts. Int J Oncol. 2006;28:855–861. [PubMed] [Google Scholar]
  • 100.Roddie H, Klammer M, Thomas C, et al. Phase I/II study of vaccination with dendritic-like leukaemia cells for the immunotherapy of acute myeloid leukaemia. Br J Haematol. 2006;133:152–157. doi: 10.1111/j.1365-2141.2006.05997.x. [DOI] [PubMed] [Google Scholar]
  • 101.Houtenbos I, Westers TM, Ossenkoppele GJ, et al. Leukaemic dendritic cell vaccination for patients with acute myeloid leukaemia. Br J Haematol. 2006;134:445–446. doi: 10.1111/j.1365-2141.2006.06196.x. [DOI] [PubMed] [Google Scholar]
  • 102.Kremser A, Dressig J, Grabrucker C, et al. Dendritic cells (DCs) can be successfully generated from leukemic blasts in individual patients with AML or MDS: an evaluation of different methods. J Immunother. 2010;33:185–199. doi: 10.1097/CJI.0b013e3181b8f4ce. [DOI] [PubMed] [Google Scholar]
  • 103.Grabrucker C, Liepert A, Dreyig J, et al. The quality and quantity of leukemia-derived dendritic cells from patients with acute myeloid leukemia and myelodysplastic syndrome are a predictive factor for the lytic potential of dendritic cells-primed leukemia-specific T cells. J Immunother. 2010;33:523–537. doi: 10.1097/CJI.0b013e3181d87ffd. [DOI] [PubMed] [Google Scholar]
  • 104.Liepert A, Grabrucker C, Kremser A. Quality of T-cells after stimulation with leukemia-derived dendritic cells (DC) from patients with acute myeloid leukemia (AML) or myeloid dysplastic syndrome (MDS) is predictive for their leukemia cytotoxic potential. Cell Immunol. 2010;265:23–30. doi: 10.1016/j.cellimm.2010.06.009. [DOI] [PubMed] [Google Scholar]
  • 105.Osman Y, Takahashi M, Zheng Z, et al. Dendritic cells stimulate the expansion of PML-RAR alpha specific cytotoxic T-lymphocytes: its applicability for antileukemia immunotherapy. J Exp Clin Cancer Res. 1999;18:485–492. [PubMed] [Google Scholar]
  • 106.Zeis M, Siegel S, Wagner A, et al. Generation of cytotoxic responses in mice and human individuals against hematological malignancies using survivin-RNA-transfected dendritic cells. J Immunol. 2003;170:5391–5397. doi: 10.4049/jimmunol.170.11.5391. [DOI] [PubMed] [Google Scholar]
  • 107.Van Tendeloo VF, Van de Velde A, Van Driessche A, et al. Induction of complete and molecular remissions in acute myeloid leukemia by Wilms’ tumor 1 antigen-targeted dendritic cell vaccination. Proc Natl Acad Sci USA. 2010;107:13824–13829. doi: 10.1073/pnas.1008051107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Fujii S, Fujimoto K, Shimizu K, et al. Presentation of tumor antigens by phagocytic dendritic cell clusters generated from human CD34+ hematopoietic progenitor cells: induction of autologous cytotoxic T lymphocytes against leukemic cells in acute myelogenous leukemia patients. Cancer Res. 1999;59:2150–2158. [PubMed] [Google Scholar]
  • 109.Galea-Lauri J, Darling D, Mufti G, et al. Eliciting cytotoxic T lymphocytes against acute myeloid leukemia-derived antigens: evaluation of dendritic cell-leukemia cell hybrids and other antigen-loading strategies for dendritic cell-based vaccination. Cancer Immunol Immunother. 2002;51:299–310. doi: 10.1007/s00262-002-0284-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Spisek R, Chevallier P, Morineau N, et al. Induction of leukemia-specific cytotoxic response by cross-presentation of late-apoptotic leukemic blasts by autologous dendritic cells of nonleukemic origin. Cancer Res. 2002;62:2861–2868. [PubMed] [Google Scholar]
  • 111.Banat GA, Usluoglu N, Hoeck M, et al. Dendritic cells fused with core binding factor-beta positive acute myeloid leukaemia blast cells induce activation of cytotoxic lymphocytes. Br J Haematol. 2004;126:593–601. doi: 10.1111/j.1365-2141.2004.05087.x. [DOI] [PubMed] [Google Scholar]
  • 112.Lee JJ, Kook H, Park MS, et al. Immunotherapy using autologous monocyte-derived dendritic cells pulsed with leukemic cell lysates for acute myeloid leukemia relapse after autologous peripheral blood stem cell transplantation. J Clin Apher. 2004;19:66–70. doi: 10.1002/jca.10080. [DOI] [PubMed] [Google Scholar]
  • 113.Klammer M, Waterfall M, Samuel K, et al. Fusion hybrids of dendritic cells and autologous myeloid blasts as a potential cellular vaccine for acute myeloid leukaemia. Br J Haematol. 2005;129:340–349. doi: 10.1111/j.1365-2141.2005.05477.x. [DOI] [PubMed] [Google Scholar]
  • 114.Lee JJ, Park MS, Park JS, et al. Induction of leukemic-cell-specific cytotoxic T lymphocytes by autologous monocyte-derived dendritic cells presenting leukemic cell antigens. J Clin Apher. 2006;21:188–194. doi: 10.1002/jca.20085. [DOI] [PubMed] [Google Scholar]
  • 115.Van Driessche A, Van de Velde AL, Nijs G, et al. Clinical-grade manufacturing of autologous mature mRNA-electroporated dendritic cells and safety testing in acute myeloid leukemia patients in a phase I dose-escalation clinical trial. Cytotherapy. 2009;11:653–668. doi: 10.1080/14653240902960411. [DOI] [PubMed] [Google Scholar]
  • 116.Cavdar AO, Babacan E, Gozdasoglu S, et al. T-lymphocyte sub-populations in orbito-ocular granulocytic sarcoma (OOGS) and acute myelocytic leukemia (AML): a preliminary study. Med Oncol Tumor Pharmacother. 1993;10:113–115. doi: 10.1007/BF02987777. [DOI] [PubMed] [Google Scholar]
  • 117.Bruserud O. Effects of imipenem and cilastatin on human T-lymphocytes derived from acute leukemia patients with chemotherapy-induced leucopenia: studies of T-lymphocyte responses in the presence of acute myelogenous leukemia (AML) blast accessory cells. Int J Immunopharmacol. 2000;22:69–81. doi: 10.1016/S0192-0561(99)00070-3. [DOI] [PubMed] [Google Scholar]
  • 118.Costello RT, Sivori S, Marcenaro E, et al. Defective expression and function of natural killer cell-triggering receptors in patients with acute myeloid leukemia. Blood. 2002;99:3661–3667. doi: 10.1182/blood.V99.10.3661. [DOI] [PubMed] [Google Scholar]
  • 119.Fauriat C, Moretta A, Olive D, et al. Defective killing of dendritic cells by autologous natural killer cells from acute myeloid leukemia patients. Blood. 2005;106:2186–2188. doi: 10.1182/blood-2005-03-1270. [DOI] [PubMed] [Google Scholar]
  • 120.Nowbakht P, Ionescu MC, Rohner A, et al. Ligands for natural killer cell-activating receptors are expressed upon the maturation of normal myelomonocytic cells but at low levels in acute myeloid leukemias. Blood. 2005;105:3615–3622. doi: 10.1182/blood-2004-07-2585. [DOI] [PubMed] [Google Scholar]
  • 121.Fauriat C, Just-Landi S, Mallet F, et al. Deficient expression of NCR in NK cells from acute myeloid leukemia: Evolution during leukemia treatment and impact of leukemia cells in NCRdull phenotype induction. Blood. 2007;109:323–330. doi: 10.1182/blood-2005-08-027979. [DOI] [PubMed] [Google Scholar]
  • 122.Molldrem JJ. Vaccination for leukemia. Biol Blood Marrow Transplant. 2006;12:13–18. doi: 10.1016/j.bbmt.2005.10.014. [DOI] [PubMed] [Google Scholar]
  • 123.Heslop HE, Stevenson FK, Molldrem JJ (2003) Immunotherapy of hematologic malignancy. Hematology Am Soc Hematol Educ Program 331–349 [DOI] [PubMed]
  • 124.Schmid C, Labopin M, Nagler A, et al. Donor lymphocyte infusion in the treatment of first hematological relapse after allogeneic stem-cell transplantation in adults with acute myeloid leukemia: a retrospective risk factors analysis and comparison with other strategies by the EBMT Acute Leukemia Working Party. J Clin Oncol. 2007;25:4938–4945. doi: 10.1200/JCO.2007.11.6053. [DOI] [PubMed] [Google Scholar]
  • 125.Yegin ZA, Ozkurt ZN, Aki SZ, et al. Donor lymphocyte infusion for leukemia relapse after hematopoietic stem cell transplantation. Transfus Apher Sci. 2010;42:239–245. doi: 10.1016/j.transci.2010.03.011. [DOI] [PubMed] [Google Scholar]
  • 126.Gesundheit B, Shapira MY, Resnick IB, et al. Successful cell-mediated cytokine-activated immunotherapy for relapsed acute myeloid leukemia after hematopoietic stem cell transplantation. Am J Hematol. 2009;84:188–190. doi: 10.1002/ajh.21346. [DOI] [PubMed] [Google Scholar]
  • 127.Gribben JG, Guinan EC, Boussiotis VA, et al. Complete blockade of B7 family-mediated costimulation is necessary to induce human alloantigen-specific anergy: a method to ameliorate graft-versus-host disease and extend the donor pool. Blood. 1996;87:4887–4893. [PubMed] [Google Scholar]
  • 128.Larsen CP, Elwood ET, Alexander DZ, et al. Long-term acceptance of skin and cardiac allografts after blocking CD40 and CD28 pathways. Nature. 1996;381:434–438. doi: 10.1038/381434a0. [DOI] [PubMed] [Google Scholar]
  • 129.Blazar BR, Taylor PA, Panoskaltsis-Mortari A, et al. Blockade of CD40 ligand-CD40 interaction impairs CD4+ T cell-mediated alloreactivity by inhibiting mature donor T cell expansion and function after bone marrow transplantation. J Immunol. 1997;158:29–39. [PubMed] [Google Scholar]
  • 130.Guinan EC, Boussiotis VA, Neuberg D, et al. Transplantation of anergic histoincompatible bone marrow allografts. N Engl J Med. 1999;340:1704–1714. doi: 10.1056/NEJM199906033402202. [DOI] [PubMed] [Google Scholar]
  • 131.Cavazzana-Calvo M, Fromont C, Le Deist F, et al. Specific elimination of alloreactive T cells by an anti-interleukin-2 receptor B chain-specific immunotoxin. Transplantation. 1990;50:1–7. doi: 10.1097/00007890-199007000-00001. [DOI] [PubMed] [Google Scholar]
  • 132.Valteau-Couanet D, Cavazzana-Calvo M, Le Deist F, et al. Functional study of residual T lymphocytes after specific elimination of alloreactive T cells by a specific anti-interleukin-2 receptor Bk chain immunotoxin. Transplantation. 1993;56:1574–1576. [PubMed] [Google Scholar]
  • 133.Harris DT, Sakiestewa D, Lyons C, et al. Prevention of graft-versus-host disease (GVHD) by elimination of recipient-reactive donor T cells with recombinant toxins that target the interleukin 2 (IL-2) receptor. Bone Marrow Transplant. 1999;23:137–144. doi: 10.1038/sj.bmt.1701535. [DOI] [PubMed] [Google Scholar]
  • 134.Montagna D, Yvon E, Calcaterra V, et al. Depletion of alloreactive T cells by a specific anti-interleukin-2 receptor p55 chain immunotoxin does not impair in vitro antileukemia and antiviral activity. Blood. 1999;93:3550–3557. [PubMed] [Google Scholar]
  • 135.van Dijk AM, Kessler FL, Stadhouders-Keet SA, et al. Selective depletion of major and minor histocompatibility antigen reactive T cells: towards prevention of acute graft-versus-host disease. Br J Haematol. 1999;107:169–175. doi: 10.1046/j.1365-2141.1999.01675.x. [DOI] [PubMed] [Google Scholar]
  • 136.Andre-Schmutz I, Le Deist F, Hacein-Bey-Abina S, et al. Immune reconstitution without graft-versus-host disease after haemopoietic stem-cell transplantation: a phase 1/2 study. Lancet. 2002;360:130–137. doi: 10.1016/S0140-6736(02)09413-8. [DOI] [PubMed] [Google Scholar]
  • 137.Koh MB, Prentice HG, Corbo M, et al. Alloantigen-specific T-cell depletion in a major histocompatibility complex fully mismatched murine model provides effective graft-versus-host disease prophylaxis in the presence of lymphoid engraftment. Br J Haematol. 2002;118:108–116. doi: 10.1046/j.1365-2141.2002.03682.x. [DOI] [PubMed] [Google Scholar]
  • 138.Solomon SR, Mielke S, Savani BN, et al. Selective depletion of alloreactive donor lymphocytes: a novel method to reduce the severity of graft-versus-host disease in older patients undergoing matched sibling donor stem cell transplantation. Blood. 2005;106:1123–1129. doi: 10.1182/blood-2005-01-0393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Wehler TC, Nonn M, Brandt B, et al. Targeting the activation-induced antigen CD137 can selectively deplete alloreactive T cells from antileukemic and antitumor donor T-cell lines. Blood. 2007;109:365–373. doi: 10.1182/blood-2006-04-014100. [DOI] [PubMed] [Google Scholar]
  • 140.Ge X, Brown J, Sykes M, et al. CD134-allodepletion allows selective elimination of alloreactive human T cells without loss of virus-specific and leukemia-specific effectors. Biol Blood Marrow Transplant. 2008;14:518–530. doi: 10.1016/j.bbmt.2008.02.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Hartwig UF, Nonn M, Khan S, et al. Depletion of alloreactive donor T lymphocytes by CD95-mediated activation-induced cell death retains antileukemic, antiviral, and immunoregulatory T cell immunity. Biol Blood Marrow Transplant. 2008;14:99–109. doi: 10.1016/j.bbmt.2007.10.002. [DOI] [PubMed] [Google Scholar]
  • 142.Nonn M, Herr W, Khan S, et al. Selective depletion of alloreactive T lymphocytes using patient-derived nonhematopoietic stimulator cells in allograft engineering. Transplantation. 2008;86:1427–1435. doi: 10.1097/TP.0b013e31818810d6. [DOI] [PubMed] [Google Scholar]
  • 143.Marijt E, Wafelman A, van der Hoorn M, et al. Phase I/II feasibility study evaluating the generation of leukemia-reactive cytotoxic T lymphocyte lines for treatment of patients with relapsed leukemia after allogeneic stem cell transplantation. Haematologica. 2007;92:72–80. doi: 10.3324/haematol.10433. [DOI] [PubMed] [Google Scholar]
  • 144.Colvin GA, Berz D, Ramanathan M, et al. Nonengraftment haploidentical cellular immunotherapy for refractory malignancies: tumor responses without chimerism. Biol Blood Marrow Transplant. 2009;15:421–431. doi: 10.1016/j.bbmt.2008.12.503. [DOI] [PubMed] [Google Scholar]
  • 145.Eljaafari A, Farre A, Duperrier K, et al. Generation of helper and cytotoxic CD4+T cell clones specific for the minor histocompatibility antigen H-Y, after in vitro priming of human T cells by HLA-identical monocyte-derived dendritic cells. Transplantation. 2001;71:1449–1455. doi: 10.1097/00007890-200105270-00016. [DOI] [PubMed] [Google Scholar]
  • 146.Azuma T, Makita M, Ninomiya K, et al. Identification of a novel WT1-derived peptide which induces human leucocyte antigen-A24-restricted anti-leukaemia cytotoxic T lymphocytes. Br J Haematol. 2002;116:601–603. doi: 10.1046/j.0007-1048.2001.03329.x. [DOI] [PubMed] [Google Scholar]
  • 147.Falkenburg JH, Marijt WA, Heemskerk MH, et al. Minor histocompatibility antigens as targets of graft-versus-leukemia reactions. Curr Opin Hematol. 2002;9:497–502. doi: 10.1097/00062752-200211000-00005. [DOI] [PubMed] [Google Scholar]
  • 148.Sadovnikova E, Parovichnikova EN, Savchenko VG, et al. The CD68 protein as a potential target for leukaemia-reactive CTL. Leukemia. 2002;16:2019–2026. doi: 10.1038/sj.leu.2402635. [DOI] [PubMed] [Google Scholar]
  • 149.Amrolia PJ, Reid SD, Gao L, et al. Allorestricted cytotoxic T cells specific for human CD45 show potent antileukemic activity. Blood. 2003;101:1007–1014. doi: 10.1182/blood-2002-02-0525. [DOI] [PubMed] [Google Scholar]
  • 150.Mutis T. Targeting alloreactive donor T-cells to hematopoietic system-restricted minor histocompatibility antigens to dissect graft-versus-leukemia effects from graft-versus-host disease after allogeneic stem cell transplantation. Int J Hematol. 2003;78:208–212. doi: 10.1007/BF02983796. [DOI] [PubMed] [Google Scholar]
  • 151.Fujiwara H, El Ouriaghli F, Grube M, et al. Identification and in vitro expansion of CD4+ and CD8+ T cells specific for human neutrophil elastase. Blood. 2004;103:3076–3083. doi: 10.1182/blood-2003-07-2424. [DOI] [PubMed] [Google Scholar]
  • 152.Guo Y, Niiya H, Azuma T, et al. Direct recognition and lysis of leukemia cells by WT1-specific CD4+ T lymphocytes in an HLA class II-restricted manner. Blood. 2005;106:1415–1418. doi: 10.1182/blood-2005-01-0413. [DOI] [PubMed] [Google Scholar]
  • 153.Tsuji T, Yasukawa M, Matsuzaki J, et al. 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. 2005;106:470–476. doi: 10.1182/blood-2004-09-3663. [DOI] [PubMed] [Google Scholar]
  • 154.Xue SA, Gao L, Hart D, et al. Elimination of human leukemia cells in NOD/SCID mice by WT1-TCR gene-transduced human T cells. Blood. 2005;106:3062–3067. doi: 10.1182/blood-2005-01-0146. [DOI] [PubMed] [Google Scholar]
  • 155.Ho WY, Nguyen HN, Wolfl M, et al. In vitro methods for generating CD8+ T-cell clones for immunotherapy from the naive repertoire. J Immunol Methods. 2006;310:40–52. doi: 10.1016/j.jim.2005.11.023. [DOI] [PubMed] [Google Scholar]
  • 156.Heemskerk MH. T-cell receptor gene transfer for the treatment of leukemia and other tumors. Haematologica. 2010;95:15–19. doi: 10.3324/haematol.2009.016022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Ochi T, Fujiwara H, Yasukawa M (2010) Application of adoptive T-cell therapy using tumor antigen-specific T-cell receptor gene transfer for the treatment of human leukemia. J Biomed Biotechnol 521248 [DOI] [PMC free article] [PubMed]
  • 158.Vera JF, Brenner MK, Dotti G. Immunotherapy of human cancers using gene modified T lymphocytes. Curr Gene Ther. 2009;9:396–408. doi: 10.2174/156652309789753338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Xue SA, Gao L, Thomas S. Development of a Wilms’ tumor antigen-specific T-cell receptor for clinical trials: engineered patient’s T cells can eliminate autologous leukemia blasts in NOD/SCID mice. Haematologica. 2010;95:126–134. doi: 10.3324/haematol.2009.006486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Peinert S, Prince HM, Guru PM, et al. Gene-modified T cells as immunotherapy for multiple myeloma and acute myeloid leukemia expressing the Lewis Y antigen. Gene Ther. 2010;5:678–686. doi: 10.1038/gt.2010.21. [DOI] [PubMed] [Google Scholar]
  • 161.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–e22. doi: 10.1200/JCO.2006.05.9964. [DOI] [PubMed] [Google Scholar]
  • 162.Brentjens R, Yeh R, Bernal Y, et al. 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;18:666–668. doi: 10.1038/mt.2010.31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Mohle R, Bautz F, Rafii S, et al. The chemokine receptor CXCR-4 is expressed on CD34+ hematopoietic progenitors and leukemic cells and mediates transendothelial migration induced by stromal cell-derived factor-1. Blood. 1998;91:4523–4530. [PubMed] [Google Scholar]
  • 164.Bleakley M, Riddell SR. Molecules and mechanisms of the graft-versus-leukaemia effect. Nat Rev Cancer. 2004;4:371–380. doi: 10.1038/nrc1365. [DOI] [PubMed] [Google Scholar]
  • 165.Ruggeri L, Aversa F, Martelli MF, et al. Allogeneic hematopoietic transplantation and natural killer cell recognition of missing self. Immunol Rev. 2006;214:202–218. doi: 10.1111/j.1600-065X.2006.00455.x. [DOI] [PubMed] [Google Scholar]
  • 166.Waldhauer I, Steinle A. NK cells and cancer immunosurveillance. Oncogene. 2008;27:5932–5943. doi: 10.1038/onc.2008.267. [DOI] [PubMed] [Google Scholar]
  • 167.Farag SS, Fehniger TA, Ruggeri L, et al. Natural killer cell receptors: new biology and insights into the graft-versus-leukemia effect. Blood. 2002;100:1935–1947. doi: 10.1182/blood-2002-02-0350. [DOI] [PubMed] [Google Scholar]
  • 168.Ruggeri L, Capanni M, Urbani E. Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants. Science. 2002;295:2097–2100. doi: 10.1126/science.1068440. [DOI] [PubMed] [Google Scholar]
  • 169.Leung W, Iyengar R, Turner V, et al. Determinants of antileukemia effects of allogeneic NK cells. J Immunol. 2004;172:644–650. doi: 10.4049/jimmunol.172.1.644. [DOI] [PubMed] [Google Scholar]
  • 170.Verheyden S, Demanet C. NK cell receptors and their ligands in leukemia. Leukemia. 2008;22:249–257. doi: 10.1038/sj.leu.2405040. [DOI] [PubMed] [Google Scholar]
  • 171.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;105:3051–3057. doi: 10.1182/blood-2004-07-2974. [DOI] [PubMed] [Google Scholar]
  • 172.Rubnitz JE, Inaba H, Ribeiro RC, et al. NKAML: a pilot study to determine the safety and feasibility of haploidentical natural killer cell transplantation in childhood acute myeloid leukemia. J Clin Oncol. 2010;28:955–959. doi: 10.1200/JCO.2009.24.4590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Cho D, Campana D. Expansion and activation of natural killer cells for cancer immunotherapy. Korean J Lab Med. 2009;29:89–96. doi: 10.3343/kjlm.2009.29.2.89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Siegler U, Meyer-Monard S, Jorger S, et al. Good manufacturing practice-compliant cell sorting and large-scale expansion of single KIR-positive alloreactive human natural killer cells for multiple infusions to leukemia patients. Cytotherapy. 2010;12:750–763. doi: 10.3109/14653241003786155. [DOI] [PubMed] [Google Scholar]
  • 175.Koehl U, Sorensen J, Esser R, et al. IL-2 activated NK cell immunotherapy of three children after haploidentical stem cell transplantation. Blood Cells Mol Dis. 2004;33:261–266. doi: 10.1016/j.bcmd.2004.08.013. [DOI] [PubMed] [Google Scholar]
  • 176.Robertson MJ. Role of chemokines in the biology of natural killer cells. J Leukoc Biol. 2002;71:173–183. [PubMed] [Google Scholar]
  • 177.Bruserud O, Kittang AO. The chemokine system in experimental and clinical hematology. Curr Top Microbiol Immunol. 2010;341:3–12. doi: 10.1007/82_2010_18. [DOI] [PubMed] [Google Scholar]
  • 178.Maghazachi AA. Role of chemokines in the biology of natural killer cells. Curr Top Microbiol Immunol. 2010;341:37–58. doi: 10.1007/82_2010_20. [DOI] [PubMed] [Google Scholar]
  • 179.Muller I, Kordowich S, Holzwarth C, et al. Application of multipotent mesenchymal stromal cells in pediatric patients following allogeneic stem cell transplantation. Blood Cells Mol Dis. 2008;40:25–32. doi: 10.1016/j.bcmd.2007.06.021. [DOI] [PubMed] [Google Scholar]
  • 180.Meuleman N, Tondreau T, Ahmad I, et al. Infusion of mesenchymal stromal cells can aid hematopoietic recovery following allogeneic hematopoietic stem cell myeloablative transplant: a pilot study. Stem Cells Dev. 2009;18:1247–1252. doi: 10.1089/scd.2009.0029. [DOI] [PubMed] [Google Scholar]
  • 181.Gotherstrom C, Lundqvist A, Duprez IR, et al. Fetal and adult multipotent mesenchymal stromal cells are killed by different pathways. Cytotherapy. 2011;13:269–278. doi: 10.3109/14653249.2010.523077. [DOI] [PubMed] [Google Scholar]
  • 182.Klyuchnikov E, Asenova S, Kern W, et al. Post-transplant immune reconstitution after unrelated allogeneic stem cell transplant in patients with acute myeloid leukemia. Leuk Lymphoma. 2010;51:1450–1463. doi: 10.3109/10428194.2010.496015. [DOI] [PubMed] [Google Scholar]
  • 183.Gross S, Walden P. Immunosuppressive mechanisms in human tumors: why we still cannot cure cancer. Immunol Lett. 2010;116:7–14. doi: 10.1016/j.imlet.2007.11.012. [DOI] [PubMed] [Google Scholar]
  • 184.Rosenberg SA, Restifo NP, Yang JC, et al. Adoptive cell transfer: a clinical path to effective cancer immunotherapy. Nat Rev Cancer. 2008;8:299–308. doi: 10.1038/nrc2355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Smits EL, Ponsaerts P, Berneman ZN, et al. The use of TLR7 and TLR8 ligands for the enhancement of cancer immunotherapy. Oncologist. 2008;13:859–875. doi: 10.1634/theoncologist.2008-0097. [DOI] [PubMed] [Google Scholar]
  • 186.Weber J. Review: anti-CTLA-4 antibody ipilimumab: case studies of clinical response and immune-related adverse events. Oncologist. 2007;12:864–872. doi: 10.1634/theoncologist.12-7-864. [DOI] [PubMed] [Google Scholar]

Articles from Cancer Immunology, Immunotherapy : CII are provided here courtesy of Springer

RESOURCES