Allogeneic hematopoietic stem cell transplantation (allogeneic HSCT) remains a curative treatment for hematological malignancies resistant to other treatment approaches through the unique GVL effect. However, relapse remains a major cause of treatment failure after allogeneic HSCT for patients with high-risk hematological malignancies. Further improvements in exploiting the GVL effect to prevent relapse in high-risk leukemias while minimizing toxicity have focused on the use of targeted antileukemic immunotherapy. These strategies include methods to boost the GVL effect with leukemia vaccines or the adoptive transfer of leukemia-specific lymphocytes. Vaccines can be classified as those against defined antigens such as minor histocompatibility antigens (mHags) or leukemia-associated antigens (PR1, WT1, and BCR-ABL) and those that have broad “antileukemic” activity such as engineered irradiated leukemia cells or leukemia-derived dendritic cells (DCs). The unique posttransplantation milieu, which is characterized by lymphopenia, regulatory T-cell depletion, and the release of growth factors, provides a unique opportunity for effective antitumor immunotherapy and augmenting specific GVL responses. This review focuses on approaches to enhancimg the GVL response by combining allogeneic HSCT with vaccination.

Although important advances have been made in the treatment of hematological malignancies using chemotherapy, and more recently with targeted therapies such as tyrosine kinase inhibitors, curative treatments often require allogeneic hematopoietic stem cell transplantation (allogeneic HSCT). Unfortunately, even this intensive treatment fails to prevent relapse in 10%-60% of cases, depending on whether the disease was treated early or was refractory at the time of transplantation. The effectiveness of allogeneic HSCT for hematological malignancies is due primarily to immunologic recognition and elimination of recipient leukemia cells by donor T cells, the so-called GVL effect.1 

Some of the antigens that drive this GVL response have been characterized and can be categorized broadly into 3 classes: (1) ubiquitously expressed alloantigens, also known as minor histocompatibility antigens (mHags), widely expressed by normal tissues in the recipient as well as by leukemia cells, and capable of initiating both GVHD and GVL responses2 ; (2) alloantigens expressed uniquely by cells of the hematopoietic system (tissue-restricted mHags) such as HA-1 and HA-22 ; and (3) leukemia antigens including leukemia-specific antigens such as BCR-ABL in Philadelphia-chromosome–positive leukemia,3  and over- or aberrantly expressed leukemia-associated antigens (LAAs) such as proteinase 3 (PR3),3  Wilms tumor 1 (WT1),4,5  the preferentially expressed antigen of melanoma (PRAME),6,7  and BMI-1.8  Several studies have shown a temporal inverse relationship between circulating T cells directed against mHags or LAAs and minimal residual disease in patients with acute and chronic leukemia after allogeneic HSCT.9–14 

The increasing array of both alloantigens and leukemia antigens that can elicit antileukemia T-cell responses provides the basis for development of vaccines for leukemia both inside and outside of the context of allogeneic HSCT.

Vaccines can be classified as those against defined antigens and those with broad antileukemic activity that are not directed against a specific tumor antigen. Examples of vaccines against defined antigens include peptides15  and DNA or RNA encoding the entire sequence or major antigenic elements of the tumor protein.16  Vaccines with broad antileukemic activity include leukemic dendritic cells (DCs), DC fusions with leukemia cells,17  cell lysates,18  and tumor cells engineered to secrete GM-CSF.19  The advantage of the latter approach is that a prior knowledge of tumor antigens is not required and multiple antigens can be targeted simultaneously.

The majority of vaccine trials in patients with leukemia have used peptide vaccines due to their ease of manufacture and relatively low cost (summarized in Table 1). Phase ½ trials on both sides of the Atlantic have investigated the use of BCR-ABL peptide combinations in patients with chronic myeloid leukemia (CML).20–22  In 16 chronic-phase CML patients with various degrees of residual disease on imatinib or IFN-α, Bocchia et al confirmed the immunogenicity of this approach and reported stable and complete cytogenetic remission in a minority of patients.21  In a safety study in 8 patients with myeloid malignancies, a single vaccination with PR1 and WT1 peptides, together with montanide and GM-CSF adjuvants, induced CD8+ T-cell responses to PR1 or WT1 or both vaccines in 8 of 8 patients, with a brief decrease in molecular markers of disease,23  although repeated vaccination failed to induce sustained immune responses against the leukemia antigens.24  A large pilot study of repeated vaccination with PR1, a 9–amino acid human leukocyte antigen A0201 (HLA-A0201)–restricted peptide derived from proteinase 3, in 66 HLA-A0201 patients with acute myeloid leukemia (AML), CML, or myelodysplastic syndrome (MDS) found immune responses in 25 (47%) of patients. Clinical responses ranging from improvements in blood counts to complete cytogenetic remission were observed in 9 of 25 immune responders (36%) compared with 3 of 28 nonresponders (10%). Interestingly, immune response to PR1 was associated with improved event-free survival with a trend toward longer overall survival.25  The group in Osaka were the first to show the safety and immunogenicity of WT1 peptide vaccination in patients with hematological and solid cancer.26  Several investigators have since confirmed immune responses to vaccination with WT1 peptide or WT1 mRNA–electroporated DCs in patients with MDS and AML.27–29  In these studies, clinical responses were observed in 30%-80% of evaluable patients (including stable disease and reduced expression of tumor markers), although correlation with the detection of immunological responses in peripheral blood was variable. A recent pilot study investigated the immunogenicity of a polyvalent WT1 vaccine in patients with AML in complete remission (CR). The vaccine was composed of a mixture of heteroclitic HLA class I peptides designed to induce stronger WT1-specific CD8+ T-cell responses and synthetic longer peptides to induce CD4+ T-cell responses across several HLA types and to provide help for long-lasting immunity. CD8+ and CD4+ T cells directed against WT1 were induced in the majority of patients, with some suggestion of improved survival.28  Another potential leukemia-associated antigen, the receptor for hyaluronic acid mediated motility (RHAMM or CD168), has also been used as a target for vaccine, with reports of clinical and immunologic responses after the administration of the HLA-A0201–restricted RHAMM R3 peptide emulsified with montanide adjuvant and GM-CSF in patients with AML, MDS, and multiple myeloma overexpressing RHAMM/CD168.30  Finally several studies have evaluated the safety and efficacy of other vaccine strategies using heat-shock protein, leukemic DCs (DC), or fusions of DCs with myeloma or lymphoma cells, tumor-derived idiotype protein, and tumor cells engineered to secrete GM-CSF in patients with hematological malignancies, and have shown promising results.17–19,31  A recent phase 3 clinical trial conducted in patients with follicular lymphoma who achieved CR after chemotherapy showed significant improvement in progression-free survival in patients vaccinated with autologous tumor–derived idiotype vaccine conjugated to keyhole limpet hemocyanin plus GM-CSF compared with controls.31 

These studies indicate that a variety of leukemia and lymphoma antigen–specific vaccination strategies can induce functional cytotoxic T-cell (CTL) responses that are associated with clinical improvement in some cases, especially in the setting of minimal residual disease, and support the use of such relatively simple approaches to boosting GVL responses posttransplantation.

The finding of increased frequencies of BCR-ABL-, PR1-, and WT1-specific CTL and T cells directed against recipient mHags after allogeneic HSCT suggests that GVL could be further enhanced by posttransplantation vaccination.9–14,32,33  The profoundly lymphopenic environment immediately after transplantation provides a favorable milieu for rapid and extensive lymphocyte expansion and facilitates immune responses to weak self-antigens (reviewed in Rezvani and Barrett34 ). TCR Vβ spectratyping reveals that in the first few months after transplantation, the T-cell repertoire is oligoclonal, with skewing of the T cells toward host, leukemia, and viral antigens35  (with the potential to cause GVHD and exert GVL and antiviral activity), despite global immune deficiency. Indeed, massive clonal T-cell expansions have been reported in a patient with severe GVHD in whom the T-cell compartment was nearly completely (> 95%) occupied by one GVHD clone.36 

The role of lymphopenia in antitumor immunity in murine models was first reported in the late 1970s.37  More recently, animal studies have shown that lymphoablation enhances the effectiveness of adoptively transferred, tumor-specific CD8+ T cells.38  Several preclinical murine studies have evaluated the role of lymphodepletion combined with vaccination strategies.39  The most direct evidence for the role of homeostatic T-cell proliferation in tumor eradication in humans comes from a clinical trial at the National Institutes of Health (NIH) involving 35 patients with advanced metastatic melanoma refractory to conventional treatments. Patients received in vitro expanded autologous tumor-infiltrating lymphocytes directed against overexpressed melanoma self-antigens in combination with IL-2 after conditioning with total body irradiation, fludarabine, and cyclophosphamide, resulting in huge expansions of the adoptively transferred clones with sustained regression of melanoma in 50% of cases.40  A drawback of this approach in the clinical setting is the technical difficulty of producing sufficient quantities of antigen-specific T cells for adoptive transfer. To overcome this limitation, in a subsequent study, the investigators genetically engineered peripheral blood lymphocytes carrying TCR chains specific for a melanoma antigen. The TCR-engineered T cells were transferred into 13 patients, with objective regression of metastatic melanoma lesions in 2 subjects.41 

Transient lymphopenia induced by sublethal total body irradiation or other chemotherapeutic regimens is thought to enhance the efficiency of adoptive immunotherapy by altering homeostatic mechanisms that promote the expansion and stimulation of tumor-reactive effector T cells and minimize tumor-induced immune suppression.42  In the allogeneic HSCT setting, the incoming donor T cells are not tolerized to the leukemia, and after transplantation, the lymphopenic environment allows strong expansion of respective antitumor T cells in the presence of cytokines responsible for thymic-independent homeostatic T-cell proliferation, such as IL-7, IL-15, and IL-21. In addition to eradicating cells that may suppress antitumor responses, such as regulatory T cells, lymphoid reconstitution of either donor or host origin may overcome inherent defects in T-cell signaling, processing, or presentation and may strengthen the costimulatory functions of APCs.42 

Because reconstitution of the T-cell compartment in lymphopenic hosts is regulated by peptides occupying MHC class I and II molecules,43  at the time of T-cell recovery, there may be an opportunity to skew the T-cell repertoire by engaging the available MHC class I and class II molecules with peptides of particular interest. Therefore, if tumor-associated peptides are presented to the proliferating lymphocytes during a lymphopenic episode, the host may be repopulated with tumor-reactive T cells that could lead to better tumor control. These observations imply that the first few months after transplantation offer a unique environment for delivering GVL directed against both leukemia-associated antigens and mHags expressed by the leukemia.

Combining vaccination with transplantation strategies for the prevention of relapse

There are several reasons for considering vaccines after allogeneic HSCT. First, with a global decrease in transplantation-related mortality, relapse remains the major unresolved obstacle to overcome in allogeneic HSCT for malignant diseases. Second, given the lower tumor burden, the early posttransplantation phase should be an ideal setting for antitumor immune responses to operate. Third, the unique immune milieu around the time of the transplantation is permissive to the generation of antileukemia immune responses. Ideally, an effective immunotherapeutic strategy should result in the in vivo generation of large numbers of high avidity, antileukemia lymphocytes without the induction of immune tolerance. As described in the previous section, the reconstituting immune system after allogeneic HSCT provides a unique approximation to this setting.

Several studies in patients with multiple myeloma and lymphoma incorporated vaccination in the posttransplantation setting.44,45  Whereas immunological responses could be detected, convincing clinical responses were absent. Notably, these studies administered the vaccines months after transplantation, after exponential T-cell proliferation had likely occurred.

Limitations to vaccine therapies after ASCT

One potential limitation of vaccinating early posttransplantation is that antigen-specific CD8+ T cells during this time may be at risk for rapid induction of senescence. Indeed, a recent study showed that while circulating LAA-specific CD8+ T cells are prominent in patients with myeloid leukemia after allogeneic HSCT, cells are functionally unresponsive and display features of replicative senescence.46  Another concern is the potential impact of immunosuppression on the vaccine-induced immune response. However, even in the face of immunosuppression to prevent GVHD, GVL responses can be observed. We showed that even in unvaccinated patients, levels of WT1- and PR1-specific T cells increase during the first 3 months after transplantation despite the use of low-dose cyclosporin,10,11  suggesting that the administration of vaccines in patients treated with low-dose calcineurin inhibitors may not interfere with the vaccine-induced T-cell response. In a recent phase 1 trial, Ho et al vaccinated patients with high-risk MDS and AML with lethally irradiated, autologous, GM-CSF–secreting leukemia cells early after allogeneic HSCT. They reported that despite the use of the calcineurin inhibitor tacrolimus as GVHD prophylaxis, immunization in this setting was safe, immunogenic, and associated with biological activity.19 

Combining vaccines with adoptive transfer of vaccine-primed lymphocytes after transplantation

The efficiency of vaccination can be increased further by combining adoptive T-cell transfer with vaccination. This involves vaccinating the patient, collecting vaccine-primed lymphocytes by apheresis before chemotherapy, and reinfusing them with further vaccination after lymphoreductive chemotherapy. Work published by June et al supports the feasibility and efficacy of this approach in stimulating specific immunity to influenza and pneumococcal antigens in the autologous setting.47,48  In randomized phase 1/2 trials, the investigators demonstrated that patients who received a single infusion of in vivo vaccine-primed, ex vivo–costimulated autologous T cells early posttransplantation, followed by booster immunizations posttransplantation, had accelerated immune reconstitution and enhanced antigen-specific CD4+ and CD8+ T-cell function in vivo. They subsequently extended the applicability of this approach to vaccination against tumor antigens in a phase 1/2 trial in 54 patients with myeloma. All patients received an autologous stem cell transplantation followed by vaccine-primed, ex vivo–costimulated autologous T cells at day 2 after transplantation. The patients were genetically randomized based on their HLA-A0201 genotype to receive an HLA-A0201–restricted multipeptide tumor antigen vaccine derived from the human telomerase reverse transcriptase and the antiapoptotic protein survivin and the pneumococcal conjugate vaccine before and after transplantation. HLA-A0201–negative patients received the pneumococcal conjugate vaccine only. The investigators reported augmented and accelerated cellular and humoral immune reconstitution, including antitumor immunity in 36% of HLA-A0201–positive patients, although no impact on event-free survival was noted.49 

This approach was also explored in a phase 2 study by investigators at Johns Hopkins University using autologous leukemia cells admixed with GM-CSF–secreting K562 cells (the GVAX platform). After a single pretransplantation dose of the vaccine, the primed lymphocytes were collected and reinfused with the stem cell graft as postremission therapy after autologous stem cell transplantation for AML.39  Fifty-four subjects were enrolled, 46 of whom achieved CR with chemotherapy and 28 of whom received a total of 9 vaccinations at 3-weekly intervals posttransplantation. For all patients who achieved CR, the 3-year relapse-free survival rate was 47.4% and the overall survival rate was 57.4% compared with 61.8% and 73.4%, respectively, in the 28 immunotherapy-treated patients. After posttransplantation immunotherapy, immune responses to the vaccine were detected in 100% of patients, including delayed-type hypersensitivity reactions in 7 of 18 (39%), T-cell responses (assessed by 7-day ELISpot assay) in 15 of 17, and antibody responses to GVAX in 17 of 17 (100%) of patients.

In the setting of allogeneic HSCT, the opportunity exists to vaccinate the donor before lymphocyte collection. This approach has the advantage that vaccine-primed lymphocytes are collected from a healthy donor with a healthy immune system, rather than from patients tolerant to their own tumor antigens with reduced immunity from prior chemotherapy. The first study to show that tumor-specific T cells can be safely induced in a healthy donor and transferred to the recipient after allogeneic HSCT was published by Kwak et al in 1995.50  These investigators immunized an HLA-matched donor with a patient-derived idiotype vaccine before stem cell collection, and demonstrated the successful transfer of idiotype-specific T cells posttransplantation, which was associated with a significant reduction in the serum paraprotein levels.

The next logical step will be to enhance the GVL effect of adoptively transferred vaccine-primed lymphocytes further by booster vaccinations early after allogeneic HSCT. Investigators at Stanford University recently demonstrated the feasibility of this approach, which they termed “immunotransplantation,” in a preclinical lymphoma model.51  They showed that the efficiency of intratumoral vaccination with an immunostimulatory CG-enriched oligodeoxynucleotide can be augmented significantly by the adoptive transfer of vaccine-primed, tumor-specific T cells into syngeneic HSCT recipients, followed by posttransplantation booster vaccinations.51  Clinical trials to assess the efficacy of this approach after allogeneic HSCT are under development.

Allogeneic HSCT continues to play a unique role in achieving cure of hematological malignancies that are otherwise resistant to treatment. However, as alternative treatment approaches continue to improve and increasing numbers of older patients present with leukemia, the challenge to cure more resistant malignancies with allogeneic HSCT will increase. Allogeneic HSCT is least effective against high-risk leukemias and current manipulations of conditioning regimens, posttransplantation immunosuppression, and donor lymphocyte infusions have probably reached their capacity to deliver GVL. To improve the natural GVL reactivity of allogeneic HSCT, it will be necessary to adopt new targeted treatments to boost GVL further; such approaches include the use of leukemia vaccines and the adoptive transfer of leukemia-specific T cells. Several improvements can be made to optimize the vaccine-transplantation strategy. Most vaccines currently used in clinical trials are small peptides presented to CD8+ T cells with no recruitment of CD4+ help. Whereas CD4+ T cells are dispensable for primary expansion of CD8+ T cells and their differentiation into cytotoxic effectors, secondary CTL expansion is wholly dependent on the presence of CD4+ T cells. Inclusion of MHC class II binding peptides in a vaccine or immunization using whole-tumor protein or lysate to elicit both CD4+ and CD8+ T cells may enhance the persistence of vaccine-induced responses. Ways to overcome tolerance are also being explored. Experimental murine models and human vaccine studies have shown that the preferential depletion of CD4+CD25+ regulatory T cells before vaccination enhances the vaccine-induced T-cell response.52,53 

In summary, vaccines targeting leukemia antigens make a logical adjuvant to the allogeneic GVL effect. In the next few years, we should be able to define the optimum way to use these 2 powerful immune modalities to reduce relapse after allogeneic HSCT for otherwise incurable malignancies. It is possible that a multifaceted approach to immunotherapy involving allogeneic HSCT, adoptive T-cell transfer, and vaccination could prove to be a highly effective strategy for the control of refractory leukemia.

Conflict-of-interest disclosure: The author declares no competing financial interests. Off-label drug use: Published results of phase 1/2 studies of vaccination against leukemia-associated antigens.

Katayoun Rezvani, Hammersmith Hospital, Imperial College, Du Cane Road, London W12 0HS United Kingdom; Phone: +44(0)2083832175; Fax: +44(0)2033138223; e-mail: k.rezvani@imperial.ac.uk.

1
Weiden
 
PL
Flournoy
 
N
Thomas
 
ED
, et al. 
Antileukemic effect of graft-versus-host disease in human recipients of allogeneic-marrow grafts
N Engl J Med
1979
, vol. 
300
 (pg. 
1068
-
1073
)
2
Goulmy
 
E
Human minor histocompatibility antigens
Curr Opin Immunol
1996
, vol. 
8
 (pg. 
75
-
81
)
3
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
, vol. 
88
 (pg. 
2450
-
2457
)
4
Gao
 
L
Bellantuono
 
I
Elsasser
 
A
, et al. 
Selective elimination of leukemic CD34(+) progenitor cells by cytotoxic T lymphocytes specific for WT1
Blood
2000
, vol. 
95
 (pg. 
2198
-
2203
)
5
Oka
 
Y
Udaka
 
K
Tsuboi
 
A
, et al. 
Cancer immunotherapy targeting Wilms' tumor gene WT1 product
J Immunol
2000
, vol. 
164
 (pg. 
1873
-
1880
)
6
Quintarelli
 
C
Dotti
 
G
De
 
AB
, et al. 
Cytotoxic T lymphocytes directed to the Preferentially Expressed Antigen of Melanoma (PRAME) target chronic myeloid leukemia
Blood
2008
, vol. 
112
 (pg. 
1876
-
1885
)
7
Rezvani
 
K
Yong
 
AS
Tawab
 
A
, et al. 
Ex vivo characterization of polyclonal memory CD8+ T-cell responses to PRAME-specific peptides in patients with acute lymphoblastic leukemia and acute and chronic myeloid leukemia
Blood
2009
, vol. 
113
 (pg. 
2245
-
2255
)
8
Yong
 
AS
Stephens
 
N
Weber
 
G
, et al. 
Improved outcome following allogeneic stem cell transplantation in chronic myeloid leukemia is associated with higher expression of BMI-1 and immune responses to BMI-1 protein
Leukemia
2011
, vol. 
25
 (pg. 
629
-
637
)
9
Marijt
 
WA
Heemskerk
 
MH
Kloosterboer
 
FM
, et al. 
Hematopoiesis-restricted minor histocompatibility antigens HA-1- or HA-2-specific T cells can induce complete remissions of relapsed leukemia
Proc Natl Acad Sci U S A
2003
, vol. 
100
 (pg. 
2742
-
2747
)
10
Rezvani
 
K
Yong
 
AS
Savani
 
BN
, et al. 
Graft-versus-leukemia effects associated with detectable Wilms tumor-1 specific T lymphocytes after allogeneic stem-cell transplantation for acute lymphoblastic leukemia
Blood
2007
, vol. 
110
 (pg. 
1924
-
1932
)
11
Rezvani
 
K
Price
 
D
Brenchley
 
J
, et al. 
Transfer of PR1-specific T-cell clones from donor to recipient by stem cell transplantation and association with GvL activity
Cytotherapy
2007
, vol. 
9
 (pg. 
245
-
251
)
12
Morita
 
Y
Heike
 
Y
Kawakami
 
M
, et al. 
Monitoring of WT1-specific cytotoxic T lymphocytes after allogeneic hematopoietic stem cell transplantation
Int J Cancer
2006
, vol. 
119
 (pg. 
1360
-
1367
)
13
Wang
 
ZD
Li
 
D
Huang
 
XJ
Graft-versus-leukemia effects of Wilms' tumor 1 protein-specific cytotoxic T lymphocytes in patients with chronic myeloid leukemia after allogeneic hematopoietic stem cell transplantation
Chin Med J (Engl)
2010
, vol. 
123
 (pg. 
912
-
916
)
14
Wei
 
L
Zuo
 
H
Sun
 
X
, et al. 
Comparison of Wilms' tumor antigen 1-specific T lymphocyte generation soon after nonmyeloablative allergenic stem-cell transplantation in acute and chronic leukemia patients
Int J Hematol
2010
, vol. 
91
 (pg. 
652
-
660
)
15
Rezvani
 
K
Peptide vaccine therapy for leukemia
Int J Hematol
2011
, vol. 
93
 (pg. 
274
-
280
)
16
Rice
 
J
Ottensmeier
 
CH
Stevenson
 
FK
DNA vaccines: precision tools for activating effective immunity against cancer
Nat Rev Cancer
2008
, vol. 
8
 (pg. 
108
-
120
)
17
Rosenblatt
 
J
Avigan
 
D
Can leukemia-derived dendritic cells generate antileukemia immunity?
Expert Rev Vaccines
2006
, vol. 
5
 (pg. 
467
-
472
)
18
Zeng
 
Y
Feng
 
H
Graner
 
MW
Katsanis
 
E
Tumor-derived, chaperone-rich cell lysate activates dendritic cells and elicits potent antitumor immunity
Blood
2003
, vol. 
101
 (pg. 
4485
-
4491
)
19
Ho
 
VT
Vanneman
 
M
Kim
 
H
, et al. 
Biologic activity of irradiated, autologous, GM-CSF-secreting leukemia cell vaccines early after allogeneic stem cell transplantation
Proc Natl Acad Sci U S A
2009
, vol. 
106
 (pg. 
15825
-
15830
)
20
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
, vol. 
95
 (pg. 
1781
-
1787
)
21
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
, vol. 
365
 (pg. 
657
-
662
)
22
Rojas
 
JM
Knight
 
K
Wang
 
L
Clark
 
RE
Clinical evaluation of BCR-ABL peptide immunisation in chronic myeloid leukaemia: results of the EPIC study
Leukemia
2007
, vol. 
21
 (pg. 
2287
-
2295
)
23
Rezvani
 
K
Yong
 
AS
Mielke
 
S
, et al. 
Leukemia-associated antigen-specific T-cell responses following combined PR1 and WT1 peptide vaccination in patients with myeloid malignancies
Blood
2008
, vol. 
111
 (pg. 
236
-
242
)
24
Rezvani
 
K
Yong
 
AS
Mielke
 
S
, et al. 
Repeated PR1 and WT1 peptide vaccination in Montanide-adjuvant fails to induce sustained high-avidity, epitope-specific CD8+ T cells in myeloid malignancies
Haematologica
2011
, vol. 
96
 (pg. 
432
-
440
)
25
Qazilbash
 
MH
Wieder
 
ED
Thall
 
PF
, et al. 
PR1 peptide vaccine-induced immune response is associated with better event-free survival in patients with myeloid leukemia [Abstract]
Blood
2007
, vol. 
110
 pg. 
283
 
26
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 U S A
2004
, vol. 
101
 (pg. 
13885
-
13890
)
27
Keilholz
 
U
Letsch
 
A
Busse
 
A
, et al. 
A clinical and immunologic phase 2 trial of Wilms tumor gene product 1 (WT1) peptide vaccination in patients with AML and MDS
Blood
2009
, vol. 
113
 (pg. 
6541
-
6548
)
28
Maslak
 
PG
Dao
 
T
Krug
 
LM
, et al. 
Vaccination with synthetic analog peptides derived from WT1 oncoprotein induces T-cell responses in patients with complete remission from acute myeloid leukemia
Blood
2010
, vol. 
116
 (pg. 
171
-
179
)
29
Van Tendeloo
 
VF
Van de Velde
 
V
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 U S A
2010
, vol. 
107
 (pg. 
13824
-
13829
)
30
Schmitt
 
M
Schmitt
 
A
Rojewski
 
MT
, et al. 
RHAMM-R3 peptide vaccination in patients with acute myeloid leukemia, myelodysplastic syndrome, and multiple myeloma elicits immunologic and clinical responses
Blood
2008
, vol. 
111
 (pg. 
1357
-
1365
)
31
Schuster
 
SJ
Neelapu
 
SS
Gause
 
BL
, et al. 
Vaccination with patient-specific tumor-derived antigen in first remission improves disease-free survival in follicular lymphoma
J Clin Oncol
2011
, vol. 
29
 (pg. 
2787
-
2794
)
32
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
, vol. 
6
 (pg. 
1018
-
1023
)
33
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
, vol. 
102
 (pg. 
2892
-
2900
)
34
Rezvani
 
K
Barrett
 
AJ
Characterizing and optimizing immune responses to leukaemia antigens after allogeneic stem cell transplantation
Best Pract Res Clin Haematol
2008
, vol. 
21
 (pg. 
437
-
453
)
35
Mackall
 
CL
Bare
 
CV
Granger
 
LA
, et al. 
Thymic-independent T cell regeneration occurs via antigen-driven expansion of peripheral T cells resulting in a repertoire that is limited in diversity and prone to skewing
J Immunol
1996
, vol. 
156
 (pg. 
4609
-
4616
)
36
Michálek
 
J
Collins
 
RH
Hill
 
BJ
Brenchley
 
JM
Douek
 
DC
Identification and monitoring of graft-versus-host specific T-cell clone in stem cell transplantation
Lancet
2003
, vol. 
361
 (pg. 
1183
-
1185
)
37
Hellström
 
KE
Hellstrom
 
I
Kant
 
JA
Tamerius
 
JD
Regression and inhibition of sarcoma growth by interference with a radiosensitive T-cell population
J Exp Med
1978
, vol. 
148
 (pg. 
799
-
804
)
38
Gattinoni
 
L
Finkelstein
 
SE
Klebanoff
 
CA
, et al. 
Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells
J Exp Med
2005
, vol. 
202
 (pg. 
907
-
912
)
39
Borrello
 
I
Sotomayor
 
EM
Rattis
 
FM
, et al. 
Sustaining the graft-versus-tumor effect through posttransplant immunization with granulocyte-macrophage colony-stimulating factor (GM-CSF)-producing tumor vaccines
Blood
2000
, vol. 
95
 (pg. 
3011
-
3019
)
40
Dudley
 
ME
Wunderlich
 
JR
Robbins
 
PF
, et al. 
Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes
Science
2002
, vol. 
298
 (pg. 
850
-
854
)
41
Morgan
 
RA
Dudley
 
ME
Wunderlich
 
JR
, et al. 
Cancer regression in patients after transfer of genetically engineered lymphocytes
Science
2006
, vol. 
314
 (pg. 
126
-
129
)
42
Surh
 
CD
Sprent
 
J
Regulation of mature T cell homeostasis
Semin Immunol
2005
, vol. 
17
 (pg. 
183
-
191
)
43
Goldrath
 
AW
Bevan
 
MJ
Low-affinity ligands for the TCR drive proliferation of mature CD8+ T cells in lymphopenic hosts
Immunity
1999
, vol. 
11
 (pg. 
183
-
190
)
44
Reichardt
 
VL
Okada
 
CY
Liso
 
A
, et al. 
Idiotype vaccination using dendritic cells after autologous peripheral blood stem cell transplantation for multiple myeloma–a feasibility study
Blood
1999
, vol. 
93
 (pg. 
2411
-
2419
)
45
Reece
 
DE
Foon
 
KA
Bhattacharya-Chatterjee
 
M
, et al. 
Use of the anti-idiotype antibody vaccine TriAb after autologous stem cell transplantation in patients with metastatic breast cancer
Bone Marrow Transplant
2000
, vol. 
26
 (pg. 
729
-
735
)
46
Beatty
 
GL
Smith
 
JS
Reshef
 
R
, et al. 
Functional unresponsiveness and replicative senescence of myeloid leukemia antigen-specific CD8+ T cells after allogeneic stem cell transplantation
Clin Cancer Res
2009
, vol. 
15
 (pg. 
4944
-
4953
)
47
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
, vol. 
11
 (pg. 
1230
-
1237
)
48
Stadtmauer
 
EA
Vogl
 
DT
Luning Prak
 
E
, et al. 
Transfer of influenza vaccine-primed costimulated autologous T cells after stem cell transplantation for multiple myeloma leads to reconstitution of influenza immunity: results of a randomized clinical trial
Blood
2011
, vol. 
117
 (pg. 
63
-
71
)
49
Rapoport
 
AP
Aqui
 
NA
Stadtmauer
 
EA
, et al. 
Combination immunotherapy using adoptive T-cell transfer and tumor antigen vaccination on the basis of hTERT and survivin after ASCT for myeloma
Blood
2011
, vol. 
117
 (pg. 
788
-
797
)
50
Kwak
 
LW
Taub
 
DD
Duffey
 
PL
, et al. 
Transfer of myeloma idiotype-specific immunity from an actively immunized marrow donor
Lancet
1995
, vol. 
345
 
8956
(pg. 
1016
-
1020
)
51
Brody
 
JD
Goldstein
 
MJ
Czerwinski
 
DK
Levy
 
R
Immunotransplantation preferentially expands T-effector cells over T-regulatory cells and cures large lymphoma tumors
Blood
2009
, vol. 
113
 (pg. 
85
-
94
)
52
Litzinger
 
MT
Fernando
 
R
Curiel
 
TJ
, et al. 
The IL-2 immunotoxin denileukin diftitox reduces regulatory T cells and enhances vaccine-mediated T-cell immunity
Blood
2007
, vol. 
110
 (pg. 
3192
-
3201
)
53
Casares
 
N
Arribillaga
 
L
Sarobe
 
P
, et al. 
CD4+/CD25+ regulatory cells inhibit activation of tumor-primed CD4+ T cells with IFN-gamma-dependent antiangiogenic activity, as well as long-lasting tumor immunity elicited by peptide vaccination
J Immunol
2003
, vol. 
171
 (pg. 
5931
-
5939
)