Skip to main content
Clinical and Experimental Immunology logoLink to Clinical and Experimental Immunology
. 2007 May;148(2):189–198. doi: 10.1111/j.1365-2249.2007.03383.x

Translational Mini-Review Series on Vaccines: Peptide vaccines for myeloid leukaemias

A J Barrett 1, K Rezvani 1
PMCID: PMC1868869  PMID: 17437417

Abstract

ARTICLES PUBLISHED IN THIS MINI-REVIEW SERIES ON VACCINES Monitoring of human papillomavirus vaccination. Clin Exp Immunol 2007; 148: doi:10.1111/j.1365-2249.2007.03384.x The Edward Jenner Museum and the history of vaccination. Clin Exp Immunol 2007; 147: doi:10.1111/j.1365-2249.2007.03304.x Dendritic cell-based vaccines in renal cancer. Clin Exp Immunol 2007; 147: doi:10.1111/j.1365-2249.2007.03305.x Development and evaluation of improved vaccines against tuberculosis. Clin Exp Immunol 2007; 147: doi: 10.1111/j.1365-2249.2007.03306.x

The graft-versus-leukaemia (GVL) effect following allogeneic stem cell transplantation is clear evidence that T lymphocytes can control and eliminate myeloid leukaemias. The successful identification of a range of leukaemia specific antigens (LSA) in recent years has stimulated efforts to induce leukaemia specific T cell responses to these antigens with peptide vaccines. An ideal LSA should be restricted in its expression to leukaemia including progenitor cells, intrinsically connected with the leukaemic phenotype, and capable of inducing strong cytotoxic T cell responses to the leukaemia. Peptides from three well-characterized LSA, the breakpoint cluster region–abelson (BCR–ABL) fusion protein of chronic myelogenous leukaemia, proteinase-3 and Wilms tumour 1 protein, serve as the basis for several clinical trials using peptide and adjuvants to treat patients with a variety of myeloid malignancies. Preliminary results from these studies indicate that these peptides induce immune responses which can translate into clinical responses which include complete remissions from leukaemia. These promising early results point the way to optimizing the administration of peptide vaccines and suggest ways of combining vaccination with allogeneic stem cell transplantation to boost GVL effects.

Keywords: clinical trials, myeloid leukaemia, peptide, vaccines

Introduction

The idea of using vaccines to treat myeloid leukaemia is not new. In the 1970s several investigators treated acute myeloid leukaemia (AML) patients entering remission with vaccines combining bacille Calmette–Guérin (BCG) with irradiated leukaemia cells. Enthusiasm for the approach waned as it became clear that vaccine recipients had no survival advantage [1]. Clinical interest in immunotherapy for myeloid leukaemias turned towards allogeneic stem cell transplantation (SCT), where it became increasingly clear that the transplant conferred a powerful T cell-mediated graft-versus-leukaemia (GVL) effect. The potential of allogeneic donor lymphocyte infusions to induce remissions in patients relapsing after SCT was strong evidence that T cells could destroy leukaemia [25]. This realization led to efforts using in vitro-generated, leukaemia-specific T cells transferred adoptively to SCT recipients to treat relapsing leukaemia. Despite a successful demonstration of proof of principle, however, the approach has not been applied widely because of its impracticability [6]. Research into cellular vaccines was re-enthused with techniques to generate dendritic cells from AML and chronic myeloid leukaemia (CML) cells. Such dendritic cell vaccines have the potential to stimulate optimally T cell responses to the leukaemia [713]. Meanwhile, other groups have begun to re-explore cellular vaccines in the form of leukaemia cell lysates or heat-shock protein fractions from leukaemia cells [1417]. While these approaches may ultimately prove effective, they represent costly, patient-tailored techniques with variable reproducibility. For these reasons, this review focuses on efforts to discover and use defined leukaemia protein antigens for immunotherapy, because of their potential to establish universal and practical vaccines for myeloid leukaemias. In the last decade a variety of proteins giving rise to leukaemia-specific antigenic peptides have been characterized and several clinical peptide-based vaccine trials have been initiated in AML and CML and myelodysplastic syndrome (MDS).

Leukaemia-specific antigens

Antigen discovery

The identification of antigenic proteins capable of inducing cytotoxic T cell (CTL) responses against myeloid leukaemias was facilitated greatly by advances in basic immunology, which defined the molecular nature of the interaction of the T cell receptor with peptide antigens presented by major histocompatibility complex locus (MHC) molecules on target cells. At the same time the mapping of intracellular antigen-processing pathways gave us, for the first time, a complete picture of the way in which proteins within the cell are digested by the proteasome into short peptide fragments which enter the endoplasmic reticulum to become assembled into MHC class I/peptide complexes that are then transported to the cell surface for the scrutiny of T cells [1820]. This revelation led almost immediately to speculation that ‘neoantigen’ peptide fragments spanning the breakpoints of the leukaemia-specific breakpoint cluster region–abelson (BCR–ABL) fusion protein specific for CML could be incorporated into MHC molecules, presented on the cell surface and be recognized as foreign by cytotoxic T cells [2124]. The same peptides, it was argued, could be used as a vaccine to treat CML. In the last 15 years or so numerous leukaemia antigens inducing T cell responses have been described. They can be categorized broadly according to whether they are overexpressed or aberrantly expressed self proteins, or neoantigens derived from leukaemia-specific chromosomal recombinations, or alleleic polymorphisms (minor histocompatibility antigens: mHag) which are novel to a stem cell donor lacking that allele (Table 1). Two distinct approaches have been used to identify leukaemia-specific and tumour-specific antigens (LSA and TSA). The ‘classical’ approach, developed by Boon and colleagues, has been used to characterize a long list of TSA − of interest to haematologists because many TSA are also overexpressed by leukaemias [25,26]. The approach requires the purification and expansion of tumour-specific T cells, typically from tumour-infiltrating lymphocytes. A DNA library from the tumour transfected into antigen-presenting cells (APC) bearing a single human leucocyte antigen (HLA) restriction element is used to generate a series of cell pools containing parts of the library. The pools are screened for cytokine production [interferon (IFN)-γ or tumour necrosis factor (TNF)] by the T cell clone. Only APC-presenting peptides derived from the tumour antigen elicit T cell responses. In a stepwise procedure, the DNA fragments eliciting T cell responses can be narrowed to the precise fragment giving rise to the protein antigen, which can then be identified by database searching. With appropriate DNA fragmentation of the protein sequence, the antigenic peptides derived from the protein can then be defined. Another variation (used successfully to identify the mHag HA-2) is to generate a T cell clone by stimulating post-transplant (donor) T cells with cells from the patient. The clone is used to screen peptides eluted from MHC molecules of the target cell and separated by mass spectrometry into peptide fractions [27,28]. The alternative approach (termed ‘reverse immunology’) does not require the generation of a tumour-specific T cell clone. Instead, a search is made of known proteins with promising antigenic characteristics. Peptides derived from these proteins and predicted to bind to common HLA types are used to generate cytotoxic T cells which are then tested against leukaemia and normal targets to define HLA restriction, leukaemia cytotoxicity and specificity [2931].

Table 1.

Candidate proteins for peptide vaccination in myeloid leukaemias.

Antigenic protein Peptides under development HLA restriction Clinical trials Ref.
Leukaemia-specific antigens
 BCR-ABL b3a2 fusion region 9–25 mers HLA-A3,-11, -B-8, DR-1, -4, -11 CML [5355]
 Primary granule proteins PR1 A-2 MDS, AML, CML [72]
PR7 A-2 No [102]
 PML-RARA BCR 1–25 DR-11 No [103]
Tumour-specific antigens
 Wilms tumour-1 WT1-126 A-2 MDS, AML [36,38]
WT1-235 A-2, -24 MDS, AML [38,104]
WT1-187 A-2 No [38]
WT1-37 A-2 No [105]
 H-TERT A-2, -3, -24 No [106108]
 PRAME A-2 No [109]
 BCL-2 A-2 No [108]
 G-250 A-2 No [108,110]
mHags
 HA-1 protein A-2 No [111]
 HA-2 protein A-2 [111]
 CD33 allele A-2 No [111,112]
 CD45 allele A-2 No [113]

BCR–ABL: breakpoint cluster region–abelson; WT1: Wilms tumour 1; MDS: myelodysplastic syndrome; AML: acute myeloid leukaemia ; CML: chronic myeloid leukaemia; HLA: human leucocyte antigen.

Validation of LSA

Candidate peptides are validated for their applicability as vaccines using a cell panel to test HLA-restriction, leukaemia specificity and ability to induce cytotoxic T cells. Tissue libraries are screened for mRNA expression of the LSA to confirm leukaemia specificity. The anti-leukaemic potential of peptide-specific T cell clones is defined in vitro by their cytotoxicity against leukaemic progenitors, with colony and proliferation inhibition assays, or in vivo in non-obese diabetic (NOD)–SCID mice inoculated with human leukaemia. Alternatively, specific cytotoxicity against human leukaemic progenitors can be tested by direct cytotoxicity or by inhibition of proliferation [32,33].

Characteristics of an ideal LSA

With an abundance of already defined antigens as well as a growing list of candidate proteins, it is useful to define some criteria for selecting proteins most likely to serve as good LSA (summarized in Table 1).

Leukaemia restriction

Concern that vaccines for LSA also expressed by other tissues might cause autoimmune disease requires a comprehensive description of LSA tissue expression. Fusion proteins arising from specific chromosomal recombinations and linked uniquely to the leukaemic process are safe because they are restricted entirely to leukaemia cells. However, the requirement for exclusive expression by the leukaemia are not absolute: the cancer-testis antigen WT1, expressed on stem cells and gonadal cells, behaves in a leukaemia-restricted manner because only the malignant cells which overexpress WT1 are susceptible targets of T cell attack [3438]. In the context of SCT only broad graft-versus-marrow specificity is required for safety, as the eradicated recipient haematopoiesis is replaced by that of the donor lacking the mHag. There has been concern (so far unjustified) that immune reactions against proteins such as PR3 and NE may target not only the leukaemia cells which overexpress the protein, but alsonormal tissues that express lesser amounts of the protein such as normal early myeloid precursors. In addition, PR3 is a target of autoimmune attack in the disease Wegener's granulomatosis, a vasculitis associated with the production of IgG1 anti-neutrophil cytoplasmic antibodies (ANCA) with specificity for PR3. This has led to excluding patients with ANCA or a history of vasculitis from vaccination with PR1.

Stem cell expression

There is an assumption that effective immunotherapy requires the target antigen to be expressed by the earliest leukaemic progenitor. While this seems to be a reasonable criterion for antigen selection, it may not be a prerequisite in every case: individual leukaemias have diverse hierarchical patterns of progenitor and progeny. CML progression, for example, appears to derive from the committed granulocytic progenitor [39].

Functional importance for the leukaemic phenotype

Malignant cells have a predilection to escape immune control by down-regulating antigen expression. Selecting antigenic targets that are intrinsic to the maintenance of the leukaemic phenotype (e.g. BCR–ABL, WT1, PR3) may render it impossible for the leukaemia cell to escape and still retain leukaemic function [4043]. It should be remembered, however, that malignant cells can also escape T cell control by down-regulating MHC molecules allowing them to conserve essential leukaemic features.

Immune response

To be effective as vaccines LSA must elicit clonal expansions of cytotoxic T cells with high avidity for the antigen. Furthermore, such antigen-specific T cells should be validated for their anti-leukaemic effects as described above.

Global applicability

The broad expression of antigens such as WT1 by many leukaemias and solid tumours makes it an attractive protein for peptide manufacture. Similarly, proteins which encode for multiple antigenic peptides binding to multiple class I and II HLA molecules are attractive because this removes the constraints of HLA-restricted vaccine approaches.

Peptide vaccines under clinical development (Table 2)

Table 2.

Peptide vaccines under clinical evaluation in myeloid malignancies.

Characteristic BCR–ABL PR1 WT1
Expression Unique to CML Myeloid malignancies Many leukaemias and solid tumours
Essential for phenotype Yes Probably Yes
Peptide(s)/HLA restriction SSKALQRPV/A-2, KQSSKALQR/A-3 ATGFKQSSK/A-11 HSATGFKQSSK/A-3,A-11 GFKQSSKAL B-8 IVHSATGFKQSSKALQRP VASDFEP /DR-1, -4, -11 VLQENTVLV/A-2 RMFPNAPYL/A-2 CMTWNQMNL /A-24 CYTWNQMNL (heteroclitic)/A-24
Size (mer) 9,11,15 9 9
Adjuvant QS-21 (Quillaja saponaria) Montanide Montanide
GM-CSF GM-CSF GM-CSF
In vitro-generated CTL kill leukaemia cells Variable + + + + + +
Affinity of T cells to peptide Low High and low High and low
Natural T cell immunity
 In controls +/– < 1/100 000 < 1/100 000
 In leukaemia +/– < 1/10 000 1/10 000
 After SCT May be lytic to CML Up to 13% CD8 Up to 5% CD8
Antibodies identified Yes (after vaccination) Yes (ANCA) Yes (in leukaemia and MDS patients)
T cell response to vaccine + + +
Clinical responses to vaccine + + +

BCR–ABL: breakpoint cluster region–abelson; WT1: Wilms tumour 1; MDS: myelodysplastic syndrome; AML: acute myeloid leukaemia ; CML: chronic myeloid leukaemia; HLA: human leucocyte antigen; PR1: proteinase 1; CTL: cytotoxic T cell; GM-CSF: granulocyte–macrophage colony-stimulating factor; ANCA: anti-neutrophil cytoplasmic antibodies.

BCR–ABL vaccine

Four fusion variants of the BCR–ABL protein are known. Of these, the p210 b3a2 variant appears most promising in terms of HLA restriction with many HLA class I and II binding peptides [21,23,24,4447]. Recently, Melief and colleagues identified 17 novel, potentially antigenic, BCR–ABL fusion peptides [48]. Ten HLA class I binding fusion peptides fulfilled the essential requirement for antigen presentation of being excised after their C-terminus by the proteasome. It is clear from numerous descriptions that b3a2 peptides can induce peptide-specific T cells and that CML cells do present BCR–ABL peptides, but BCR–ABL peptides have not been found reproducibly to induce CTL lytic for CML. The most convincing results come from studies by Clark et al., who were able to generate MHC-restricted CTL responses to CML cells using an HLA A-3 binding peptide [49]. In vivo peptide-specific CTL have been identified after stem cell transplant in patients with controlled CML and in HLA-A3 positive healthy individuals [4952]. These cells had low affinity for the peptide with variable lytic activity against fresh CML targets.

BCR–ABL trials

Pinilla-lbarz and colleagues, at the Memorial Sloane Kettering Institute, were the first to develop a BCR–ABL vaccine composed of a pool of six peptide fragments (Table 2). A safety study showed that the vaccine and adjuvant were well tolerated and could elicit T cell immune responses to BCR–ABL [53]. They then performed a phase II study where 14 CML patients in chronic phase received five doses of the vaccine and adjuvant [54]. All patients developed delayed-type hypersensitivity (DTH) or CD4+ proliferative responses to the peptides. IFN-γ-producing CD4+ and CD8+ T cells developed in 11 and four patients, respectively. Reduction in marrow Ph chromosomes occurred in four patients, but three of these continued other treatments with IFN-α or imatinib. Three patients in molecular relapse after SCT had transient molecular remissions, but two had also been treated with donor lymphocyte infusions. These results, while promising, do not demonstrate convincingly the efficacy of the vaccine. In a subsequent, more stringent, trial using the a similar peptide combination Bocchia et al. in Italy studied 16 b3a2 variant CML patients with residual disease stable for at least 6 months, after a minimum of 12 months' treatment with imatinib or 24 months with IFN-α [55]. Patients received six vaccinations and were then assessed for immunological and disease response. Of 10 patients on imatinib, nine had stable cytogenetic disease and one was in cytogenetic remission for a median of 10 months. Five achieved a complete cytogenetic remission with a negative polymerase chain reaction (PCR) for BCR–ABL in three. Of six patients stable for a median of 17 months on IFN-α treatment with 13% median Ph+ chromosomes in the marrow, five showed reductions in the percentage of Ph+ chromosomes, with two reaching complete cytogenetic remission. Five of five patients showed specific responses to the peptide with IFN-γ production, 13 of 14 had proliferative responses and delayed-type hypersensitivity to the peptides was seen in 11 patients. These results suggest that clinical responses to BCR–ABL peptides can be induced in patients with CML with low levels of stable disease. These rather modest results raise concerns that the method of vaccine administration or the immune status of the treated patients may be suboptimal, or that BCR–ABL does not induce sufficiently powerful cytolytic T cell responses to CML.

Primary granule proteins

Primary granule proteins (PGPs) are a group of serine proteases, or closely related molecules, found in granulocytes and their precursors. They occur in high concentrations in maturing granulocytes and neutrophils − approximately 1 pg enzyme per cell [5658]. Two PGPs, PR3 and NE, have been studied in detail. Both are overexpressed in myeloid leukaemia blasts and CD34+ leukaemic progenitors [29,56]. They may be important in maintaining a leukaemia phenotype: PR3 anti-sense oligonucleotides halt cell division and induce maturation of the HL-60 promyelocytic leukaemia cell line [43,56] and NE-deficient mice are protected from developing acute promyelocytic leukaemia [59]. NE production by CML cells inhibits normal granulopoiesis in culture, suggesting that NE gives CML clonal dominance over normal granulopoiesis [6062]. In 1995 we chose to study PR3 as a potential source of LSA and using a reverse immunology approach identified PR1 a nine-amino acid HLA-A*0201-restricted peptide which induced myeloid leukaemia-specific CTL responses [30,31]. PR1 is naturally processed and presented on MHC class I molecules from CD34+ CML cells [63], and the degree of cytotoxicity exerted by PR1-specific T cells correlated with the degree of cytoplasmic PR3 expression by the target [30,31,52,64,65]. PR1-specific CD8+ T cells are found at low frequencies (approximately 1/100 000 CD8+ T cells) in healthy individuals, but in leukaemia patients and after SCT these frequencies may rise one or two logs, respectively [52]. High frequencies of PR1-specific CTL are also found in patients with CML responding to IFN-α, and in patients entering molecular remission after SCT as well as patients with AML [52,64,65]. PGP as a group are immunogenic: 40–60% of healthy individuals have CD4+ and CD8+ T cells recognizing NE, PR3 and cathepsin G. These protein-specific T cells can be expanded in vitro and are cytotoxic to CML cells [29]. The occurrence of autoreactive T cells to PGP is not well understood. It appears that T cells recognizing PGP escape the normal process of eliminating autoreactive T cells by the thymus. This may explain why PGP are a target of autoimmune attack in Wegener's granuloma and related vasculitides [6671].

PR-1 vaccine trials

A pilot study nearing completion evaluated PR1 vaccine in patients with refractory or progressing myeloid leukaemias, including many who relapsed after SCT. Patients received a course of 3-weekly PR1 vaccine with montanide adjuvant and granulocyte–macrophage colony-stimulating factor (GM-CSF) [72]. Increased frequencies of PR1-specific CTLs occurred in 22 of 37 patients. Sixteen responders showed clinical improvement including complete remissions in four patients, and a longer event-free survival for patients who had an immune response compared with those who did not. No patients developed cANCA antibodies or vasculitis. These encouraging results have led to the initiation of several new studies with PR1 in less advanced patients.

WT-1

Mice immunized with WT1 peptide or WT1 cDNA reject WT1-expressing tumours, suggesting that WT1 vaccines might be effective as immunotherapy for WT1 positive leukaemia. In man WT1 protein is overexpressed in a wide range of malignancies, including myeloid leukaemias and MDS [34,7375]. Furthermore, expression increases as disease progresses, making WT1 an attractive vaccine candidate in otherwise untreatable advanced leukaemia [76,77]. A number of HLA-A2- and HLA-24-restricted WT1 epitopes are known and CTLs specific for WT1 peptides and cytotoxic to myeloid leukaemias are generated readily from normal individuals [36,38,78]. Several investigators have reported the occurrence of WT1-specific CTL in patients with cancers, myeloid and lymphoid leukaemias and healthy volunteers [52,65,7981]. T cells from patients with leukaemia are polyclonal, often recognizing all four HLA-A*0201 immunodominant peptides, whereas healthy controls had lower frequency responses to fewer epitopes, suggesting that myeloid leukaemias naturally elicit T cell responses to WT1 [81]. WT1 IgM and IgG antibodies can also be detected in patients with haematopoietic malignancies [35,82,83]. These observations suggest that WT1 is highly immunogenic in man and an interesting vaccine candidate for haematological malignancies in general.

WT1 vaccine trials

Oka et al. reported the outcome of a phase I clinical study of WT1 peptide-based immunotherapy in patients with breast or lung cancer, MDS or AML [84,85]. Patients received an HLA-A24 9-mer WT1 peptide in Montanide adjuvant at 2-week intervals in a dose escalation study. The vaccine was well tolerated and 18 of 26 patients received three or more vaccinations. The only notable side effect was profound leukopenia in two MDS patients reversed by steroid treatment, which concommittently abrogated the WT1 T cell response. Twelve of the 20 evaluable patients had clinical responses, including reductions in blood or marrow leukaemic blasts or tumour size or tumour markers. Increases in WT1-specific CTL frequency correlated with a clinical response.

Scheibenbogen and colleagues [65] used a WT1-126 peptide vaccine in 16 patients with AML and 1 patient with MDS. Patients received a median of eight vaccinations (range 3–18) [86]. WT1-specific T cell responses were detected in nine of 13 patients by tetramer analysis and eight of 13 by intracellular cytokine staining. Clinical responses were seen in six of 12 patients, with one patient achieving complete remission for 12 months. The patient with MDS had an improvement in neutrophil and platelet counts, two patients had minor responses with transient reductions in leukaemic blasts and two patients achieved temporary disease stabilization. WT1 transcripts as molecular disease markers decreased in five of these six patients. These very promising results indicate that WT1 vaccination can induce functional CTL responses associated with clinical improvement.

Vaccine developments

Improving the applicability and efficacy of peptide vaccines

Peptide vaccines are attractive to use in clinical trials because they are relatively cheap, easily manufactured to clinical grade and easy to administer. However, they have significant limitations. The first is HLA restriction: vaccine use is restricted to patients who have the appropriate HLA type (usually HLA-A2 or A24). At least 60% of the population lack these HLA types and are ineligible for vaccination. Studies with adoptive transfer of immunity to cytomegalovirus have shown the importance of combining CD4+ with CD8+ antigen-specific T cells to achieve sustained immunity [87]. Inclusion of MHC class II binding peptides in a vaccine to elicit CD4+ cells in combination with CD8+ cells would therefore be a worthwhile goal.

Peptide library screening

Peptide library screening offers a way to find new peptide epitopes. Lymphocytes from individuals selected for HLA haplotypes of interest are screened against a 15 mer overlapping peptide library covering the entire protein sequence. Guided by computer-generated peptide–MHC binding predictions, candidate peptides are synthesized and tested for their ability to induce IFN-γ production and promising peptides are further validated as described above. A cocktail of peptides with broad specificity for most common HLA class I and II types can be produced in this way [88,89].

Heteroclitic peptides

Heteroclitic peptides represent a method to enhance peptide vaccine potency. Heteroclitic peptides are synthetic variations of the natural peptide sequence which retain the same HLA binding specificity and avidity but which have enhanced affinity to the T cell receptor (TCR). Several groups are currently exploring heteroclitic WT1 peptides as vaccines [9093].

Alternatives to peptide vaccines

The use of whole protein rather than defined peptides offers the opportunity to generate leukaemia-specific CTLs against multiple peptide epitopes, binding to a wide range of HLA class I and II molecules, thus eliminating the constraint of HLA restriction and allowing the generation of CD4+ T cells, which could provide help for CD8+ T cell expansion. The identification of PGP-specific CD4+ and CD8+ T cells in healthy donors [29] supports the development of PGP protein vaccines. Protein vaccines, nevertheless, have shortcomings. They are difficult to manufacture and are inefficient inducers of CD8+ CTL responses as they are processed mainly by APCs for MHC class II presentation to CD4+ T cells [9496]. As an alternative to proteins, DNA vaccines may ultimately represent the most practical approach to inducing T cell responses to a broad range of MHC class I and II epitopes (reviewed in [97]).

Clinical applications of LSA vaccines − the future

While vaccines could conceivably be used to prevent myeloid malignancies, it is unlikely that vaccines alone could eliminate established disease unless combined with other treatments.

Combining vaccines with allogeneic SCT

The finding of increased frequencies of BCR–ABL-, PR1- and WT1-specific CTL after SCT suggests that GVL could be enhanced by post-transplant vaccination. The transplantation of a healthy donor immune system in a leukaemic recipient offers a unique opportunity to boost GVL by also vaccinating the donor. Immediately after SCT, conditions may be favourable for antigen-specific T cell expansion because the preparative regimen creates a lymphopenic environment causing a surge of interleukin (IL)-12 and IL-15 which stimulates lymphoproliferation strongly [98100]. T cells recently activated by antigen can be boosted favourably by vaccine during this period. However, before vaccination can be applied effectively in SCT recipients it will be necessary to improve methods to selectively prevent acute graft-versus-host disease (GVHD) eliminating the need for post-transplant immunosuppression. The combination of the potent GVL effect of the allograft with the vaccine boost for leukaemia-specific T cells could prove to be a highly effective strategy to control refractory leukaemias.

Combining vaccines with non-transplant treatments

Low disease burdens in CML patients stabilized to low levels of molecular disease by imatinib, patients with AML inremission or patients with early MDS offer ideal opportunities for vaccination. Chemotherapy, like SCT, can induce a favourable lymphopenic milieu conducive to rapid expansion of vaccine-boosted T cells but may also blunt T cell responsiveness. To optimize vaccine efficiency, it may therefore be necessary to collect antigen-stimulated lymphocytes by apheresis before chemotherapy, re-infusing them with further vaccination following lymphoreductive chemotherapy. Indeed, work published recently by Rapoport and colleagues supports the feasibility of this approach [101]. They performed a randomized Phase I–II trial in 54 patients with advanced myeloma to determine whether combination immunotherapy consisting of vaccination with the pneumococcal conjugate vaccine (PCV) and adoptive T cell transfer could correct the immunodeficiency and lymphopenia induced by high-dose chemotherapy. They demonstrated that individuals who received a single early post-transplant infusion of in vivo vaccine-primed and ex vivo co-stimulated autologous T cells followed by post-transplant booster immunizations had accelerated immune reconstitution and enhanced antigen-specific CD4+ and CD8+ T cell function in vivo.

We are therefore exploring the strategy of inducing lymphopenia (with or without allogeneic transplantation) followed by PR1 and WT1 peptide vaccination to selectively expand leukaemia-specific CTLs during the phase of lymphocyte recovery.

References

  • 1.Powles RL, Russell J, Lister TA, et al. Immunotherapy for acute myelogenous leukaemia: a controlled clinical study 2½ years after entry of the last patient. Br J Cancer. 1977;35:265–72. doi: 10.1038/bjc.1977.38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Antin JH. Graft-versus-leukemia: no longer an epiphenomenon. Blood. 1993;82:2273–7. [PubMed] [Google Scholar]
  • 3.Drobyski WR, Keever CA, Roth MS, et al. Salvage immunotherapy using donor leukocyte infusions as treatment for relapsed chronic myelogenous leukemia after allogeneic bone marrow transplantation: efficacy and toxicity of a defined T-cell dose. Blood. 1993;82:2310–18. [PubMed] [Google Scholar]
  • 4.Kolb HJ, Mittermuller J, Clemm C, et al. Donor leukocyte transfusions for treatment of recurrent chronic myelogenous leukemia in marrow transplant patients. Blood. 1990;76:2462–5. [PubMed] [Google Scholar]
  • 5.Kolb HJ, Schattenberg A, Goldman JM, et al. Graft-versus-leukemia effect of donor lymphocyte transfusions in marrow grafted patients. Blood. 1995;86:2041–50. European Group for Blood and Marrow Transplantation Working Party Chronic Leukemia. [PubMed] [Google Scholar]
  • 6.Falkenburg JH, Wafelman AR, Joosten P, et al. Complete remission of accelerated phase chronic myeloid leukemia by treatment with leukemia-reactive cytotoxic T lymphocytes. Blood. 1999;94:1201–8. [PubMed] [Google Scholar]
  • 7.Choudhury A, Gajewski JL, Liang JC, et al. Use of leukemic dendritic cells for the generation of antileukemic cellular cytotoxicity against Philadelphia chromosome-positive chronic myelogenous leukemia. Blood. 1997;89:1133–42. [PubMed] [Google Scholar]
  • 8.Harrison BD, Adams JA, Briggs M, Brereton ML, Yin JA. Stimulation of autologous proliferative and cytotoxic T-cell responses by ‘leukemic dendritic cells’ derived from blast cells in acute myeloid leukemia. Blood. 2001;97:2764–71. doi: 10.1182/blood.v97.9.2764. [DOI] [PubMed] [Google Scholar]
  • 9.Klammer M, Waterfall M, Samuel K, Turner ML, Roddie PH. Fusion hybrids of dendritic cells and autologous myeloid blasts as a potential cellular vaccine for acute myeloid leukaemia. Br J Haematol. 2005;129:340–9. doi: 10.1111/j.1365-2141.2005.05477.x. [DOI] [PubMed] [Google Scholar]
  • 10.Deeb D, Gao X, Jiang H, Divine G, Dulchavsky SA, Gautam SC. Vaccination with leukemia-loaded dendritic cells eradicates residual disease and prevent relapse. J Exp Ther Oncol. 2006;5:183–93. [PubMed] [Google Scholar]
  • 11.Rosenblatt J, Avigan D. Can leukemia-derived dendritic cells generate antileukemia immunity? Exp Rev Vaccines. 2006;5:467–72. doi: 10.1586/14760584.5.4.467. [DOI] [PubMed] [Google Scholar]
  • 12.Lindner I, Kharfan-Dabaja MA, Ayala E, et al. Induced dendritic cell differentiation of chronic myeloid leukemia blasts is associated with down-regulation of BCR–ABL. J Immunol. 2003;171:1780–91. doi: 10.4049/jimmunol.171.4.1780. [DOI] [PubMed] [Google Scholar]
  • 13.Takahashi T, Tanaka Y, Nieda M, et al. Dendritic cell vaccination for patients with chronic myelogenous leukemia. Leuk Res. 2003;27:795–802. doi: 10.1016/s0145-2126(03)00011-0. [DOI] [PubMed] [Google Scholar]
  • 14.Galea-Lauri J, Darling D, Mufti G, Harrison P, Farzaneh F. 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]
  • 15.Hoos A, Levey DL. Vaccination with heat shock protein-peptide complexes: from basic science to clinical applications. Exp Rev Vaccines. 2003;2:369–79. doi: 10.1586/14760584.2.3.369. [DOI] [PubMed] [Google Scholar]
  • 16.Li Z, Qiao Y, Liu B, et al. Combination of imatinib mesylate with autologous leukocyte-derived heat shock protein and chronic myelogenous leukemia. Clin Cancer Res. 2005;11:4460–8. doi: 10.1158/1078-0432.CCR-05-0250. [DOI] [PubMed] [Google Scholar]
  • 17.Zeng Y, Feng H, Graner MW, Katsanis E. Tumor-derived, chaperone-rich cell lysate activates dendritic cells and elicits potent antitumor immunity. Blood. 2003;101:4485–91. doi: 10.1182/blood-2002-10-3108. [DOI] [PubMed] [Google Scholar]
  • 18.Trowsdale J, Ragoussis J, Campbell RD. Map of the human MHC. Immunol Today. 1991;12:443–6. doi: 10.1016/0167-5699(91)90017-n. [DOI] [PubMed] [Google Scholar]
  • 19.van Endert PM. Genes regulating MHC class I processing of antigen. Curr Opin Immunol. 1999;11:82–8. doi: 10.1016/s0952-7915(99)80015-6. [DOI] [PubMed] [Google Scholar]
  • 20.Yewdell JW, Bennink JR. Immunodominance in major histocompatibility complex class I-restricted T lymphocyte responses. Annu Rev Immunol. 1999;17:51–88. doi: 10.1146/annurev.immunol.17.1.51. [DOI] [PubMed] [Google Scholar]
  • 21.Greco G, Fruci D, Accapezzato D, et al. Two brc–abl junction peptides bind HLA-A3 molecules and allow specific induction of human cytotoxic T lymphocytes. Leukemia. 1996;10:693–9. [PubMed] [Google Scholar]
  • 22.Nieda M, Nicol A, Kikuchi A, et al. Dendritic cells stimulate the expansion of bcr–abl specific CD8+ T cells with cytotoxic activity against leukemic cells from patients with chronic myeloid leukemia. Blood. 1998;91:977–83. [PubMed] [Google Scholar]
  • 23.ten Bosch GJ, Kessler JH, Joosten AM, et al. A BCR–ABL oncoprotein p210b2a2 fusion region sequence is recognized by HLA-DR2a restricted cytotoxic T lymphocytes and presented by HLA-DR matched cells transfected with an Ii (b2a2) construct. Blood. 1999;94:1038–45. [PubMed] [Google Scholar]
  • 24.Yotnda P, Firat H, Garcia-Pons F, et al. Cytotoxic T cell response against the chimeric p210 BCR–ABL protein in patients with chronic myelogenous leukemia. J Clin Invest. 1998;101:2290–6. doi: 10.1172/JCI488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Boon T, van der Bruggen P. Human tumor antigens recognized by T lymphocytes. J Exp Med. 1996;183:725–9. doi: 10.1084/jem.183.3.725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Boon T, Coulie PG, Van den Eynde B. Tumor antigens recognized by T cells. Immunol Today. 1997;18:267–8. doi: 10.1016/s0167-5699(97)80020-5. [DOI] [PubMed] [Google Scholar]
  • 27.Marijt WA, Veenhof WF, Brand A, et al. Minor histocompatibility antigen-specific cytotoxic T cell lines, capable of lysing human hematopoietic progenitor cells, can be generated in vitro by stimulation with HLA-identical bone marrow cells. J Exp Med. 1991;173:101–9. doi: 10.1084/jem.173.1.101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.van Els CA, D'Amaro J, Pool J, et al. Immunogenetics of human minor histocompatibility antigens: their polymorphism and immunodominance. Immunogenetics. 1992;35:161–5. doi: 10.1007/BF00185109. [DOI] [PubMed] [Google Scholar]
  • 29.Fujiwara H, El OF, Grube M, et al. Identification and in vitro expansion of CD4+ and CD8+ T cells specific for human neutrophil elastase. Blood. 2004;103:3076–83. doi: 10.1182/blood-2003-07-2424. [DOI] [PubMed] [Google Scholar]
  • 30.Molldrem J, Dermime S, Parker K, et al. Targeted T-cell therapy for human leukemia: cytotoxic T lymphocytes specific for a peptide derived from proteinase 3 preferentially lyse human myeloid leukemia cells. Blood. 1996;88:2450–7. [PubMed] [Google Scholar]
  • 31.Molldrem JJ, Clave E, Jiang YZ, et al. Cytotoxic T lymphocytes specific for a nonpolymorphic proteinase 3 peptide preferentially inhibit chronic myeloid leukemia colony-forming units. Blood. 1997;90:2529–34. [PubMed] [Google Scholar]
  • 32.Bleakley M, Riddell SR. Molecules and mechanisms of the graft-versus-leukaemia effect. Nat Rev Cancer. 2004;4:371–80. doi: 10.1038/nrc1365. [DOI] [PubMed] [Google Scholar]
  • 33.Lu S, Wieder E, Komanduri K, Ma Q, Molldrem JJ. Vaccines in leukemia. Adv Pharmacol. 2004;51:255–70. doi: 10.1016/S1054-3589(04)51011-6. [DOI] [PubMed] [Google Scholar]
  • 34.Bergmann L, Miething C, Maurer U, et al. High levels of Wilms' tumor gene (wt1) mRNA in acute myeloid leukemias are associated with a worse long-term outcome. Blood. 1997;90:1217–25. [PubMed] [Google Scholar]
  • 35.Gaiger A, Reese V, Disis ML, Cheever MA. Immunity to WT1 in the animal model and in patients with acute myeloid leukemia. Blood. 2000;96:1480–9. [PubMed] [Google Scholar]
  • 36.Gao L, Bellantuono I, Elsasser A, et al. Selective elimination of leukemic CD34(+) progenitor cells by cytotoxic T lymphocytes specific for WT1. Blood. 2000;95:2198–203. [PubMed] [Google Scholar]
  • 37.Gao L, Xue SA, Hasserjian R, et al. Human cytotoxic T lymphocytes specific for Wilms' tumor antigen-1 inhibit engraftment of leukemia-initiating stem cells in non-obese diabetic-severe combined immunodeficient recipients. Transplantation. 2003;75:1429–36. doi: 10.1097/01.TP.0000061516.57346.E8. [DOI] [PubMed] [Google Scholar]
  • 38.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]
  • 39.Jamieson CH, Ailles LE, Dylla SJ, et al. Granulocyte-macrophage progenitors as candidate leukemic stem cells in blast-crisis CML. N Engl J Med. 2004;351:657–67. doi: 10.1056/NEJMoa040258. [DOI] [PubMed] [Google Scholar]
  • 40.Gordon MY, Dowding CR, Riley GP, Goldman JM, Greaves MF. Altered adhesive interactions with marrow stroma of haematopoietic progenitor cells in chronic myeloid leukaemia. Nature. 1987;328:342–4. doi: 10.1038/328342a0. [DOI] [PubMed] [Google Scholar]
  • 41.Puil L, Liu J, Gish G, et al. Bcr–Abl oncoproteins bind directly to activators of the Ras signalling pathway. EMBO J. 1994;13:764–73. doi: 10.1002/j.1460-2075.1994.tb06319.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Yamagami T, Sugiyama H, Inoue K, et al. Growth inhibition of human leukemic cells by WT1 (Wilms tumor gene) antisense oligodeoxynucleotides: implications for the involvement of WT1 in leukemogenesis. Blood. 1996;87:2878–84. [PubMed] [Google Scholar]
  • 43.Bories D, Raynal MC, Solomon DH, Darzynkiewicz Z, Cayre YE. Down-regulation of a serine protease, myeloblastin, causes growth arrest and differentiation of promyelocytic leukemia cells. Cell. 1989;59:959–68. doi: 10.1016/0092-8674(89)90752-6. [DOI] [PubMed] [Google Scholar]
  • 44.Bocchia M, Korontsvit T, Xu Q, et al. Specific human cellular immunity to bcr–abl oncogene-derived peptides. Blood. 1996;87:3587–92. [PubMed] [Google Scholar]
  • 45.Mannering SI, McKenzie JL, Fearnley DB, Hart DN. HLA-DR1-restricted bcr–abl (b3a2)-specific CD4+ T lymphocytes respond to dendritic cells pulsed with b3a2 peptide and antigen-presenting cells exposed to b3a2 containing cell lysates. Blood. 1997;90:290–7. [PubMed] [Google Scholar]
  • 46.Pawelec G, Max H, Halder T, et al. BCR/ABL leukemia oncogene fusion peptides selectively bind to certain HLA-DR alleles and can be recognized by T cells found at low frequency in the repertoire of normal donors. Blood. 1996;88:2118–24. [PubMed] [Google Scholar]
  • 47.Clark RE, Christmas SE. BCR–ABL fusion peptides and cytotoxic T cells in chronic myeloid leukaemia. Leuk Lymph. 2001;42:871–80. doi: 10.3109/10428190109097706. [DOI] [PubMed] [Google Scholar]
  • 48.Kessler JH, Bres-Vloemans SA, van Veelen PA, et al. BCR–ABL fusion regions as a source of multiple leukemia-specific CD8+ T-cell epitopes. Leukemia. 2006;20:1738–50. doi: 10.1038/sj.leu.2404354. [DOI] [PubMed] [Google Scholar]
  • 49.Clark RE, Dodi IA, Hill SC, et al. Direct evidence that leukemic cells present HLA-associated immunogenic peptides derived from the BCR–ABL b3a2 fusion protein. Blood. 2001;98:2887–93. doi: 10.1182/blood.v98.10.2887. [DOI] [PubMed] [Google Scholar]
  • 50.Butt NM, Rojas JM, Wang L, Christmas SE, bu-Eisha HM, Clark RE. Circulating bcr–abl-specific CD8+ T cells in chronic myeloid leukemia patients and healthy subjects. Haematologica. 2005;90:1315–23. [PubMed] [Google Scholar]
  • 51.Gannage M, Abel M, Michallet AS, et al. Ex vivo characterization of multiepitopic tumor-specific CD8 T cells in patients with chronic myeloid leukemia: implications for vaccine development and adoptive cellular immunotherapy. J Immunol. 2005;174:8210–18. doi: 10.4049/jimmunol.174.12.8210. [DOI] [PubMed] [Google Scholar]
  • 52.Rezvani K, Grube M, Brenchley JM, et al. Functional leukemia-associated antigen-specific memory CD8+ T cells exist in healthy individuals and in patients with chronic myelogenous leukemia before and after stem cell transplantation. Blood. 2003;102:2892–900. doi: 10.1182/blood-2003-01-0150. [DOI] [PubMed] [Google Scholar]
  • 53.Pinilla-Ibarz J, Cathcart K, Korontsvit T, et al. Vaccination of patients with chronic myelogenous leukemia with bcr–abl oncogene breakpoint fusion peptides generates specific immune responses. Blood. 2000;95:1781–7. [PubMed] [Google Scholar]
  • 54.Cathcart K, Pinilla-Ibarz J, Korontsvit T, et al. A multivalent bcr–abl fusion peptide vaccination trial in patients with chronic myeloid leukemia. Blood. 2004;103:1037–42. doi: 10.1182/blood-2003-03-0954. [DOI] [PubMed] [Google Scholar]
  • 55.Bocchia M, Gentili S, Abruzzese E, et al. Effect of a p210 multipeptide vaccine associated with imatinib or interferon in patients with chronic myeloid leukaemia and persistent residual disease: a multicentre observational trial. Lancet. 2005;365:657–62. doi: 10.1016/S0140-6736(05)17945-8. [DOI] [PubMed] [Google Scholar]
  • 56.Dengler R, Munstermann U, al-Batran S, et al. Immunocytochemical and flow cytometric detection of proteinase 3 (myeloblastin) in normal and leukaemic myeloid cells. Br J Haematol. 1995;89:250–7. doi: 10.1111/j.1365-2141.1995.tb03297.x. [DOI] [PubMed] [Google Scholar]
  • 57.Wiedow O, Meyer-Hoffert U. Neutrophil serine proteases: potential key regulators of cell signalling during inflammation. J Intern Med. 2005;257:319–28. doi: 10.1111/j.1365-2796.2005.01476.x. [DOI] [PubMed] [Google Scholar]
  • 58.Witko-Sarsat V, Cramer EM, Hieblot C, et al. Presence of proteinase 3 in secretory vesicles: evidence of a novel, highly mobilizable intracellular pool distinct from azurophil granules. Blood. 1999;94:2487–96. [PubMed] [Google Scholar]
  • 59.Lane AA, Ley TJ. Neutrophil elastase cleaves PML-RARalpha and is important for the development of acute promyelocytic leukemia in mice. Cell. 2003;115:305–18. doi: 10.1016/s0092-8674(03)00852-3. [DOI] [PubMed] [Google Scholar]
  • 60.El-Ouriaghli F, Fujiwara H, Melenhorst JJ, Sconocchia G, Hensel N, Barrett AJ. Neutrophil elastase enzymatically antagonizes the in vitro action of G-CSF. implications for the regulation of granulopoiesis. Blood. 2003;101:1752–8. doi: 10.1182/blood-2002-06-1734. [DOI] [PubMed] [Google Scholar]
  • 61.El-Ouriaghli F, Sloand E, Mainwaring L, et al. Clonal dominance of chronic myelogenous leukemia is associated with diminished sensitivity to the antiproliferative effects of neutrophil elastase. Blood. 2003;102:3786–92. doi: 10.1182/blood-2003-03-0861. [DOI] [PubMed] [Google Scholar]
  • 62.Skold S, Zeberg L, Gullberg U, Olofsson T. Functional dissociation between proforms and mature forms of proteinase 3, azurocidin, and granzyme B in regulation of granulopoiesis. Exp Hematol. 2002;30:689–96. doi: 10.1016/s0301-472x(02)00816-0. [DOI] [PubMed] [Google Scholar]
  • 63.Papadopoulos KP, Suciu-Foca N, Hesdorffer CS, Tugulea S, Maffei A, Harris PE. Naturally processed tissue- and differentiation stage-specific autologous peptides bound by HLA class I and II molecules of chronic myeloid leukemia blasts. Blood. 1997;90:4938–46. [PubMed] [Google Scholar]
  • 64.Molldrem JJ, Lee PP, Wang C, et al. Evidence that specific T lymphocytes may participate in the elimination of chronic myelogenous leukemia. Nat Med. 2000;6:1018–23. doi: 10.1038/79526. [DOI] [PubMed] [Google Scholar]
  • 65.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;100:2132–7. doi: 10.1182/blood-2002-01-0163. [DOI] [PubMed] [Google Scholar]
  • 66.Apenberg S, Andrassy K, Worner I, et al. Antibodies to neutrophil elastase: a study in patients with vasculitis. Am J Kidney Dis. 1996;28:178–85. doi: 10.1016/s0272-6386(96)90299-1. [DOI] [PubMed] [Google Scholar]
  • 67.Ballieux BE, van der Burg SH, Hagen EC, van der Woude FJ, Melief CJ, Daha MR. Cell-mediated autoimmunity in patients with Wegener's granulomatosis (WG) Clin Exp Immunol. 1995;100:186–93. doi: 10.1111/j.1365-2249.1995.tb03651.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Brouwer E, Stegeman CA, Huitema MG, Limburg PC, Kallenberg CG. T cell reactivity to proteinase 3 and myeloperoxidase in patients with Wegener's granulomatosis (WG) Clin Exp Immunol. 1994;98:448–53. doi: 10.1111/j.1365-2249.1994.tb05511.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Gallicchio MC, Savige JA. Detection of anti-myeloperoxidase and anti-elastase antibodies in vasculitides and infections. Clin Exp Immunol. 1991;84:232–7. doi: 10.1111/j.1365-2249.1991.tb08154.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Griffith ME, Coulthart A, Pusey CD. T cell responses to myeloperoxidase (MPO) and proteinase 3 (PR3) in patients with systemic vasculitis. Clin Exp Immunol. 1996;103:253–8. doi: 10.1046/j.1365-2249.1996.d01-629.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Tervaert JW, Mulder L, Stegeman C, et al. Occurrence of autoantibodies to human leucocyte elastase in Wegener's granulomatosis and other inflammatory disorders. Ann Rheum Dis. 1993;52:115–20. doi: 10.1136/ard.52.2.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Qazilbash M, Wieder E, Rios R, Lu S, et al. Vaccination with the PR1 leukemia-associated antigen can induce complete remission in patients with myeloid leukemia. Blood. 2004;104(Suppl. 1):259a. [Google Scholar]
  • 73.Menssen HD, Renkl HJ, Rodeck U, et al. Presence of Wilms' tumor gene (wt1) transcripts and the WT1 nuclear protein in the majority of human acute leukemias. Leukemia. 1995;9:1060–7. [PubMed] [Google Scholar]
  • 74.Niegemann E, Wehner S, Kornhuber B, Schwabe D, Ebener U. WT1 gene expression in childhood leukemias. Acta Haematol. 1999;102:72–6. doi: 10.1159/000040973. [DOI] [PubMed] [Google Scholar]
  • 75.Sugiyama H. Wilms tumor gene WT1 as a tumor marker for leukemic blast cells and its role in leukemogenesis. Meth Mol Med. 2002;68:223–37. [PubMed] [Google Scholar]
  • 76.Boublikova L, Kalinova M, Ryan J, et al. Wilms' tumor gene 1 (WT1) expression in childhood acute lymphoblastic leukemia: a wide range of WT1 expression levels, its impact on prognosis and minimal residual disease monitoring. Leukemia. 2006;20:254–63. doi: 10.1038/sj.leu.2404047. [DOI] [PubMed] [Google Scholar]
  • 77.Cilloni D, Gottardi E, Messa F, et al. Significant correlation between the degree of WT1 expression and the International Prognostic Scoring System Score in patients with myelodysplastic syndromes. J Clin Oncol. 2003;21:1988–95. doi: 10.1200/JCO.2003.10.503. [DOI] [PubMed] [Google Scholar]
  • 78.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;95:286–93. [PubMed] [Google Scholar]
  • 79.Gillmore R, Xue SA, Holler A, et al. Detection of Wilms' tumor antigen-specific CTL in tumor-draining lymph nodes of patients with early breast cancer. Clin Cancer Res. 2006;12:34–42. doi: 10.1158/1078-0432.CCR-05-1483. [DOI] [PubMed] [Google Scholar]
  • 80.Rezvani K, Yong A, Mielke S. WT1-specific CD8+ T lymphocytes may participate in the elimination of acute lymphoblastic leukemia following allogeneic stem cell transplantation. Blood. 2006;108(Suppl. 1):3679a. [Google Scholar]
  • 81.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;11:8799–807. doi: 10.1158/1078-0432.CCR-05-1314. [DOI] [PubMed] [Google Scholar]
  • 82.Elisseeva OA, Oka Y, Tsuboi A, et al. Humoral immune responses against Wilms tumor gene WT1 product in patients with hematopoietic malignancies. Blood. 2002;99:3272–9. doi: 10.1182/blood.v99.9.3272. [DOI] [PubMed] [Google Scholar]
  • 83.Wu F, Oka Y, Tsuboi A, et al. Th1-biased humoral immune responses against Wilms tumor gene WT1 product in the patients with hematopoietic malignancies. Leukemia. 2005;19:268–74. doi: 10.1038/sj.leu.2403539. [DOI] [PubMed] [Google Scholar]
  • 84.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;101:13885–90. doi: 10.1073/pnas.0405884101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Morita S, Oka Y, Tsuboi A, et al. A phase I/II trial of a WT1 (Wilms' tumor gene) peptide vaccine in patients with solid malignancy: safety assessment based on the phase I data. Jpn J Clin Oncol. 2006;36:231–6. doi: 10.1093/jjco/hyl005. [DOI] [PubMed] [Google Scholar]
  • 86.Keilholz U, Letsch A, Busse A, et al. Clinical and immune responses of WT1-peptide vaccination in patients with acute myeloid leukemia. Blood. 2006;108(Suppl. 1):567a. [Google Scholar]
  • 87.Walter EA, Greenberg PD, Gilbert MJ, et al. Reconstitution of cellular immunity against cytomegalovirus in recipients of allogeneic bone marrow by transfer of T-cell clones from the donor. N Engl J Med. 1995;333:1038–44. doi: 10.1056/NEJM199510193331603. [DOI] [PubMed] [Google Scholar]
  • 88.Rezvani K, Mielke S, Kilical Y, Grube M, et al. Identification of novel MHC class i and class ii epitopes of WT1 using a peptide library screen. Blood. 2005;106(Suppl. 1):2764a. [Google Scholar]
  • 89.Betts MR, Ambrozak DR, Douek DC, et al. Analysis of total human immunodeficiency virus (HIV)-specific CD4(+) and CD8(+) T-cell responses: relationship to viral load in untreated HIV infection. J Virol. 2001;75:11983–91. doi: 10.1128/JVI.75.24.11983-11991.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Pinilla-Ibarz J, May RJ, Korontsvit T, et al. Improved human T-cell responses against synthetic HLA-0201 analog peptides derived from the WT1 oncoprotein. Leukemia. 2006;20:2025–33. doi: 10.1038/sj.leu.2404380. [DOI] [PubMed] [Google Scholar]
  • 91.Rubio V, Stuge TB, Singh N, et al. Ex vivo identification, isolation and analysis of tumor-cytolytic T cells. Nat Med. 2003;9:1377–82. doi: 10.1038/nm942. [DOI] [PubMed] [Google Scholar]
  • 92.Tangri S, Ishioka GY, Huang X, et al. Structural features of peptide analogs of human histocompatibility leukocyte antigen class I epitopes that are more potent and immunogenic than wild-type peptide. J Exp Med. 2001;194:833–46. doi: 10.1084/jem.194.6.833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Zirlik KM, Zahrieh D, Neuberg D, Gribben JG. Cytotoxic T-cells generated against heteroclitic peptides kill primary tumor cells independent of the binding affinity of the native tumor antigen peptide. Blood. 2006;108:3865–70. doi: 10.1182/blood-2006-04-014415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Monaco JJ. Pathways for the processing and presentation of antigens to T cells. J Leukoc Biol. 1995;57:543–7. doi: 10.1002/jlb.57.4.543. [DOI] [PubMed] [Google Scholar]
  • 95.Rammensee HG. Antigen presentation − recent developments. Int Arch Allergy Immunol. 1996;110:299–307. doi: 10.1159/000237320. [DOI] [PubMed] [Google Scholar]
  • 96.Machiels JP, van Baren N, Marchand M. Peptide-based cancer vaccines. Semin Oncol. 2002;29:494–502. doi: 10.1053/sonc.2002.35244. [DOI] [PubMed] [Google Scholar]
  • 97.Stan R, Wolchok JD, Cohen AD. DNA vaccines against cancer. Hematol Oncol Clin North Am. 2006;20:613–36. doi: 10.1016/j.hoc.2006.02.004. [DOI] [PubMed] [Google Scholar]
  • 98.Alpdogan O, Eng JM, Muriglan SJ, et al. Interleukin-15 enhances immune reconstitution after allogeneic bone marrow transplantation. Blood. 2005;105:865–73. doi: 10.1182/blood-2003-09-3344. [DOI] [PubMed] [Google Scholar]
  • 99.Klebanoff CA, Finkelstein SE, Surman DR, et al. IL-15 enhances the in vivo antitumor activity of tumor-reactive CD8+ T cells. Proc Natl Acad Sci USA. 2004;101:1969–74. doi: 10.1073/pnas.0307298101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Reddy V, Winer AG, Eksioglu E, Meier-Kriesche HU, Schold JD, Wingard JR. Interleukin 12 is associated with reduced relapse without increased incidence of graft-versus-host disease after allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2005;11:1014–21. doi: 10.1016/j.bbmt.2005.08.032. [DOI] [PubMed] [Google Scholar]
  • 101.Rapoport AP, Stadtmauer EA, Aqui N, et al. Restoration of immunity in lymphopenic individuals with cancer by vaccination and adoptive T-cell transfer. Nat Med. 2005;11:1230–7. doi: 10.1038/nm1310. [DOI] [PubMed] [Google Scholar]
  • 102.Clave E, Molldrem J, Hensel N, Raptis A, Barrett AJ. Donor-recipient polymorphism of the proteinase 3 gene: a potential target for T-cell alloresponses to myeloid leukemia. J Immunother. 1999;22:1–6. doi: 10.1097/00002371-199901000-00001. [DOI] [PubMed] [Google Scholar]
  • 103.Gambacorti-Passerini C, Grignani F, Arienti F, Pandolfi PP, Pelicci PG, Parmiani G. Human CD4 lymphocytes specifically recognize a peptide representing the fusion region of the hybrid protein pml/RAR alpha present in acute promyelocytic leukemia cells. Blood. 1993;81:1369–75. [PubMed] [Google Scholar]
  • 104.Bellantuono I, Gao L, Parry S, et al. Two distinct HLA-A0201-presented epitopes of the Wilms tumor antigen 1 can function as targets for leukemia-reactive CTL. Blood. 2002;100:3835–7. doi: 10.1182/blood.V100.10.3835. [DOI] [PubMed] [Google Scholar]
  • 105.Rosenfeld C, Cheever MA, Gaiger A. WT1 in acute leukemia, chronic myelogenous leukemia and myelodysplastic syndrome: therapeutic potential of WT1 targeted therapies. Leukemia. 2003;17:1301–12. doi: 10.1038/sj.leu.2402988. [DOI] [PubMed] [Google Scholar]
  • 106.Vonderheide RH, Hahn WC, Schultze JL, Nadler LM. The telomerase catalytic subunit is a widely expressed tumor-associated antigen recognized by cytotoxic T lymphocytes. Immunity. 1999;10:673–9. doi: 10.1016/s1074-7613(00)80066-7. [DOI] [PubMed] [Google Scholar]
  • 107.Arai J, Yasukawa M, Ohminami H, Kakimoto M, Hasegawa A, Fujita S. Identification of human telomerase reverse transcriptase-derived peptides that induce HLA-A24-restricted antileukemia cytotoxic T lymphocytes. Blood. 2001;97:2903–7. doi: 10.1182/blood.v97.9.2903. [DOI] [PubMed] [Google Scholar]
  • 108.Vonderheide RH, Anderson KS, Hahn WC, Butler MO, Schultze JL, Nadler LM. Characterization of HLA-A3-restricted cytotoxic T lymphocytes reactive against the widely expressed tumor antigen telomerase. Clin Cancer Res. 2001;7:3343–8. [PubMed] [Google Scholar]
  • 109.Kessler JH, Beekman NJ, Bres-Vloemans SA, et al. Efficient identification of novel HLA-A(*)0201-presented cytotoxic T lymphocyte epitopes in the widely expressed tumor antigen PRAME by proteasome-mediated digestion analysis. J Exp Med. 2001;193:73–88. doi: 10.1084/jem.193.1.73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Greiner J, Schmitt M, Li L, et al. Expression of tumor-associated antigens in acute myeloid leukemia: implications for specific immunotherapeutic approaches. Blood. 2006;108:4109–17. doi: 10.1182/blood-2006-01-023127. [DOI] [PubMed] [Google Scholar]
  • 111.Goulmy E. Human minor histocompatibility antigens. Curr Opin Immunol. 1996;8:75–81. doi: 10.1016/s0952-7915(96)80108-7. [DOI] [PubMed] [Google Scholar]
  • 112.Bae J, Martinson JA, Klingemann HG. Heteroclitic CD33 peptide with enhanced anti-acute myeloid leukemic immunogenicity. Clin Cancer Res. 2004;10:7043–52. doi: 10.1158/1078-0432.CCR-04-0322. [DOI] [PubMed] [Google Scholar]
  • 113.Amrolia PJ, Reid SD, Gao L, et al. Allorestricted cytotoxic T cells specific for human CD45 show potent antileukemic activity. Blood. 2003;101:1007–14. doi: 10.1182/blood-2002-02-0525. [DOI] [PubMed] [Google Scholar]

Articles from Clinical and Experimental Immunology are provided here courtesy of British Society for Immunology

RESOURCES