Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Jan 11.
Published in final edited form as: Cytotherapy. 2010 Nov 10;13(1):8–18. doi: 10.3109/14653249.2010.530649

Tumor vaccines and beyond

JAN JOSEPH MELENHORST 1, AUSTIN JOHN BARRETT 1
PMCID: PMC8751546  NIHMSID: NIHMS1730530  PMID: 21067312

Abstract

For the last two decades the immunotherapy of patients with solid and hematopoietic tumors has met with variable success. We have reviewed the field of tumor vaccines to examine what has worked and what has not, why this has been the case, how the anti-tumor responses were examined, and how we can make tumor immunity successful for the majority of individuals rather than for the exceptional patients who currently show successful immune responses against their tumors.

Keywords: cancer vaccines, gene therapy, genes, stem cell transplantation, T-cell receptor

Introduction

To combat invading pathogens the adaptive immune system mobilizes antigen-presenting cells (APC), CD4+ and CD8+ T cells to attack the intruder, and B cells to make antibodies. Tumors developing within the body pose a different challenge because they are usually slowly developing (1) and originate from ‘self’. Tumors arise as a consequence of acquired and inherited genetic aberrations in oncogenes and tumor suppressor genes, resulting in uncontrolled growth of what is often considered a clonal initiator population (24) that accumulates a concatenation of further genetic lesions during the progression from benign to malignant (5). Loss of genetic and genomic control appears to be responsible for at least some of the process of tumorigenesis (6,7). There is ample evidence that the immune system plays a prominent role in preventing tumor development: patients with immune deficiencies such as human immunodeficiency virus (HIV)/acquired immunodeficiency syndrome (AIDS) have tumor types such as Kaposi sarcoma (8,9) and other malignancies (10,11) that rarely occur in healthy individuals. Tumor phenotypes are sculpted by the immune system, leading to escape from immune surveillance (12). In allogeneic stem cell transplants where leukemia patients receive T cells from human leukocyte antigen (HLA) haplotype-mismatched donors, the recipient’s leukemia may down-regulate the mismatched major histocompatibility antigen (MHC) molecules (13). A similar phenomenon has been described in a melanoma patient who developed melanoma subclones resistant to lysis by in vivo emergent autologous anti-melanoma CD8+ T-cell clones (12). Sometimes patients may possess pre-existing anti-tumor immunity that is ineffective but can be amplified by T-cell growth factors (12,14). Clearly immune control of malignancies is not perfect, and various strategies are being explored to boost immunity. Vaccination of the tumor-bearing host with tumor-associated antigen (TAA)-derived peptides is one such strategy. Another approach is to target polymorphic minor histocompatibility (mH) antigens that are differentially expressed by the tumor (see below). Such antigens, originally described in the allogeneic, HLA-identical transplant setting, can exhibit both broad and restricted tissue expression, and are usually targeted by T cells specific for the polymorphic epitope. As mH-specific T cells are likely to have a high avidity to the antigen, they may represent a more powerful alternative to monomorphic tumor antigens. Excellent reviews list the large repertoire of TAA and mH antigens now identified (1521).

We have focused on the progress made in vaccine therapies, strategies to monitor T-cell responses against the vaccine and the tumor, and the prospects for exploiting the knowledge gained from detailed studies on anti-tumor immunity to improve the efficiency of tumor vaccine therapy in the management of solid and hematopoietic malignancies.

Tumor and mH antigens

Researchers have identified a large array of tumor antigens targeted by autologous CD8+ T cells derived from the peripheral blood or tumor-infiltrating lymphocytes (TIL). These epitopes can be categorized broadly into antigens that are: (a) selectively or uniquely expressed in the tumor; (b) derived from tumor-specific mutations; (c) differentiation antigens; and (d) antigens overexpressed in the tumor (20,22). We and others have shown that proteins overexpressed in myeloid malignancies, such as proteinase 3 (PRTN3), neutrophil elastase (ELA2) and Wilms tumor-1 (WT1), are targeted by T cells. The primary granule proteins human ELA2 and PRTN3 are serine proteases expressed in normal myeloid cells but overexpressed in myeloid malignancies (23,24). Both contain a nonamer (VLQELNVTV) that is presented in the context of HLA-A*0201 (23,25). Leukemic cells presenting this epitope are effectively lysed by CD8+ T cells, while healthy myeloid cells are not (2326). Under certain circumstances, such as interferon (IFN) therapy (27) and allogeneic hematopoietic stem cell transplantation (HSCT) (27,28), increased frequencies of PRTN3/ELA2-derived peptide antigen (PR1)-specific CD8+ T cells have been detected in these patients. We recently demonstrated that such T cells reside predominantly in the bone marrow, and that they recognize their target antigen with high avidity (28), suggesting a contribution of PR1-specific CD8+ T cells to the eradication of myeloid leukemia cells following allogeneic HSCT. Using a platform of HLA tetramers that allowed differential detection of high- and low-avidity PR1-specific CD8+ T cells, we demonstrated persistence of low-avidity and loss of high-avidity TAA-specific CD8+ T cells in the bone marrow prior to transplant (29), consistent with in vitro findings by Molldrem et al. (30) that suggested that leukemic cells evade immune surveillance by specifically depleting T cells with high avidity for the leukemia antigen.

WT1 is a zinc finger transcription factor overexpressed in various malignancies, including leukemias (3133). WT1 fulfills an important criterion in immunotherapy: appearing to be essential for pathogenesis (3436). Nevertheless the picture is not entirely clear, as WT1 mutations occur in approximately 10% of leukemias (3739) and are associated with poor clinical prognosis (37,3941), suggesting that wild-type WT1 might in some circumstances regulate tumor progression. Patients with WT1 mutations have significantly higher WT1 gene expression levels (42), suggesting that these patients might be especially suitable for immunotherapy using WT1-specific T cells.

Minor histocompatibility antigens were first discovered in patients who developed severe graft-versus-host-disease (GvHD) after HSCT despite a full molecular match at the MHC class I and II loci, and thus the term mH antigen was used. An mH antigen arises from a genetic difference between donor and recipient that results in the presentation of an antigenic peptide (43). Many such HLA class I- and II-restricted mH antigens have been discovered over the past 15 years, and some display tissue-restricted expression, such as HA-1(44) and −2 (45), HB-1 (46), LRH-1 (47), CD19 (48) and the more recently characterized mH antigen HEATR (49). Obviously the strengths of these antigens lie in the high avidity with which the T cells target such antigens. Until recently, mH antigens were identified in the allogeneic HSCT setting in patients who mounted an anti-leukemia response in the absence of GvHD. However, the Riddell laboratory induced and selected T-cell responses against mH antigens with hematopoiesis-restricted expression for adoptive transfer prior to the transplant (49). Although this strategy is likely to target unidentified, useful mH antigens, some characteristics of these antigens have discouraged researchers from exploiting their use in immunotherapy. First, mH antigen responses can only be elicited if the donor and patient are discordant for such polymorphisms. Thus, even if the donor and patient express a highly prevalent HLA allele, for example HLA-B*0801, and the population frequency of the mH antigen is c. 50%, this relatively favorable mH antigen can elicit a response in only in 5.5% of donor–patient pairs expressing this HLA allele. Second, it is unclear whether such a response is uni- or bidirectional, i.e. whether the alternative allele can also yield a peptide that is presented in the same HLA allele and may be seen by T cells in the other direction. From studies on HA-1 is has become clear that only the product from one locus is presented in HLA (44), possibly because of unfavorable binding characteristics of the non-immunogenic variant (50). Third, none of the thus far characterized mH antigens are presented in the context of HLA class I as well as class II (21), which may negatively affect the long-term persistence of CD8 T-cell responses in the absence of a CD4 component (see below). Thus, despite the high likelihood that mH antigen responses will elicit a higher quality T-cell response than the monomorphic TAA, many more mH antigens will have to be characterized to allow their targeting in a vaccine setting.

A third type of tumor antigen was recently reported by Childs et al. (51), where they identified an endogenous retroviral envelope-derived epitope as the target of CD8 T cells in clear cell renal cell carcinoma (RCC). About 8% of the human genome consists of retroviral elements, most of which are no longer transcriptionally or translationally active (52). In RCC, however, the human endogenous retrovirus (HERV) E is reactivated and thereby yields HLA class I-presented epitopes that are new to the immune system and targeted with high efficiency (51). These data suggest that HERV can be reactivated in some tumors and targeted by the immune system. What is unclear at this moment is whether such HERV-specific T-cell responses can be elicited in the autologous T-cell pool of the patient via vaccination, and whether epitopes are presented in HLA class II.

The significance of CD4+ T cells

WT1 (and other TAA) has been targeted by various investigators using a short synthetic HLA class I binding peptide vaccine to treat patients with leukemias (5355). However, it is likely that such responses are not maintained in the absence of a CD4+ T-cell response, and that the stimulation of CD8+ T cells in the absence of helper cell engagement results in a functionally defective population often referred to as ‘helpless T cells’ (5661). CD4+ T cells, on the other hand, can do more than just provide help: numerous studies have demonstrated their cytotoxic potential (6270). Additionally, it has been shown that CD4+ T cells can be directly cytotoxic but also amplify a CD8+ T-cell response in the patient (71). Thus, aside from the fact that CD4+ T cells can regulate their own proliferation and that of CD8+ T cells by autocrine stimulation via interleukin (IL)-2 production, these cells can also be very effective at eradicating target cells. Clinical evidence for a role for allogeneic CD4+ T cells in tumor eradication comes from the HSCT setting, where CD4+ T-cell infusions have led to achievement of full remission in a number of clinical trials (72,73).

Immunotherapy of cancer

The concept of immune therapy of cancer is well established (reviewed in 74,75). Various immunotherapy strategies have been developed: monotherapies with cytokines, antibodies, autologous and allogeneic tumor vaccines; peptides, proteins, DNA, RNA and antigen-loaded APC; adoptively transferred lymphokine-activated killer cells, T cells and natural killer (NK) cells; allogeneic HSCT with or without delayed add-back of donor lymphocytes; any combination of the above. The molecular identification of antigens recognized by autologous tumor-specific CD8+ T cells since the early 1990s (19,20) has sparked an interest in using such peptides to more specifically direct the immune response against such antigens and to boost such responses in vivo or in vitro. In some studies dendritic cells (DC) have been pulsed with the antigenic peptide prior to administration to the patient, whereas in other studies a viral vector has been used to deliver the epitope to APC in the patient. A low percentage of melanoma patients vaccinated with these CD8 epitopes demonstrated tumor regression, and this regression appeared to correlate with frequencies of anti-TAA CD8+ T-cell responses (76). In the course of these studies it became clear that an effective CD8+ T-cell response against the tumor using peptide antigens would require help from CD4+ T cells or, as an alternative, the administration of IL-2. However, the lack of CD4 help cannot be used as the sole argument for why tumor immunotherapy fails in patients, because there are a multitude of explanations. Seven are given below.

  1. Tumors are hierarchically organized and originate from a dormant pool of tumor stem cells (7779) that may give rise to rapidly cycling progeny (reviewed in 80). It has been demonstrated that such quiescent tumor cells are resistant to T-cell attack. Lymphocyte infusion from an HLA-identical donor into leukemia patients post-HSCT has been used as immunotherapy of leukemia for two decades (81,82). In one study, however, it was found that while donor T cells isolated from the bone marrow of patients post-donor lymphocyte infusion recognized mature myeloid cells well, immature hematopoietic progenitor cells were resistant to lysis in the assays employed (83), suggesting that quiescent hematopoietic progenitor cells are not recognized by these T-cell clones. This suggests that additional strategies may be needed to revive the quiescent tumor, for example DNA demethylating agents or type I interferon (84). Alternatively, the establishment of a memory anti-tumor T-cell pool would allow the pruning of cycling tumor progeny by such cells, effectively containing the tumor within its small stem cell pool and preventing relapse.

  2. Vaccine induction of suppressor T cells (8590): Francois et al. (87) vaccinated melanoma patients with MAGE-A3 HLA-DP4-binding peptides and found that a significant proportion of clonally expanded antigen-specific CD4 T-cell clones suppressed the proliferation of MAGE-A3-specific CD4+ T-cell clones. Recently, Jandus et al. (88) identified FOXP3-expressing CD4+ T cells following vaccination of patients with a Melan-A vaccine using HLA-DQ tetramers, suggesting that the vaccine induced both effector and regulatory T cells. Increases in TAA-specific regulatory T cells have also been described in other settings (89). Bonertz et al. (90) analyzed the antigen specificity of some tumor-infiltrating regulatory T cells in patients with colorectal cancer. By priming regulatory T cells with synthetic peptides and incubating these cells with polyclonally stimulated control cells, they demonstrated that regulatory T cells primed with some tumor antigens inhibited the proliferation of non-specifically activated regulatory T-cell depleted peripheral blood mononuclear cells (PBMC). The authors showed furthermore that the regulatory T cells recognized fewer antigens than the effector T cells. Given that regulatory T cells suppress a T-cell response non-specifically, the data suggest that, even though the tumor microenvironment contains regulatory T cells with fewer antigen specificities, they can still suppress effector T cells via a bystander mechanism.

  3. Rather than eradicating the tumor, TAA-specific T cells may instead support tumor growth via the production of cytokines (65,91). The T-cell clones recognize the tumor but instead of destroying tumor cells they provide help to the malignancy (91).

  4. It has been shown that T cells infiltrating the tumor microenvironment are functionally inert or anergic, a state that can be reversed by the administration of IL-2 (9294).

  5. The tumor may express indoleamine 2,3-dioxygenase (IDO), which catabolizes tryptophan along the kynurenine pathway. IDO is expressed constitutively by certain tumors or induced by IFN-γ (95,96) or reverse signaling through cytotoxic T-lymphocyte antigen-4 (CTLA-4) and CD40 ligands (97,98). T-cell proliferation is inhibited by IDO-expressing cells (99) via either depletion of local tryptophan levels (100) or the apoptosis-inducing effect of its metabolites (101). Clinical evidence for the relevance of IDO-expressing DC comes from melanoma, where the presence of IDO-expressing DC in tumor draining lymph nodes is associated with a poor clinical outcome (102), thereby effectively depleting an essential amino acid from the milieu and suppressing a T-cell response by amino acid starvation. IDO has been demonstrated to be critical for maintenance of maternal tolerance to the fetus (103) and inhibiting anti-tumor responses (104). Furthermore, IDO gene and protein expression as well as functional activity occurs in acute myeloid leukemia (105).

  6. A variety of myeloid cells has been identified in the tumor environment and thought to play a role in tumorigenesis (106). One prominent yet heterogeneous group of myeloid cells is the myeloid-derived suppressor cells (MDSC), which have been detected in patients with various malignancies but not found in healthy donors (107). MDSC are the progeny of bone marrow-resident progenitor cells and express CD11b, CD33, CD34 and CD15 (106,108), have low or no expression of HLA-DR (109) and in some instances have been reported to express CD14 (110,111) and intracellular arginase I (112114). MDSC have been found in patients with tumors such as RCC (109,112114), prostate cancer (115), hepatocellular carcinoma (110), colon carcinoma (116), non-small cell lung cancer (117) and melanoma (107,116,118), all with slightly distinct phenotypes but similar functions, i.e. the suppression of T cells (118) and NK cells (111). How these cells suppress immune responses has not been fully elucidated, but may involve the secretion of transforming growth factor (TGF)-β (107) and arginase (110) by MDSC, which suppresses proliferation (113,116,118), cytokine production (113,118), CD3ζ chain expression level (117) and direct effector functions of lymphocytes (111,118).

  7. T cells responding to antigen stimulation will do so via the ligation of their antigen receptor with peptide-major histocompatibility complex (pMHC) complexes, and CD28 with CD80/CD86 on the target cells. However, during the response negative regulatory mechanisms are activated that significantly dampen the response, which include the alternative CD80/CD86 ligand CTLA-4 (119) and the PD-L1/PD-L2 ligand PD-1 (120). Some tumors (121125) are known to overexpress these ligands, which hampers the success of tumor immunity. It therefore makes good sense to target the various inhibitory pathways exploited by malignancies using pharmacologic inhibitors (126), or blocking antibodies (127) in combination with immunotherapy (114,128130).

Intriguingly, despite the many failed attempts to control the tumor via vaccine therapies, some patients do respond to this treatment by mounting either a T-cell response against the vaccine or against the antigens expressed by the tumor. Careful observational studies such as those reported by Coulie,Van der Bruggen (20) and others at the Ludwig Institute for Cancer Research (LICR; Brussels, Belgium) may help to extend successful anti-tumor immunity to patients who are currently unresponsive. Furthermore, the implementation of large-scale mH antigen discovery technologies (131) will greatly expand the repertoire of such antigens for vaccination and other forms of immunotherapies. Central to improving the efficacy of immunotherapy is the implementation of immune and tumor monitoring as described below.

Monitoring anti-tumor responses: how and when

Depending on the frequencies of responder cells, availability of reagents and antigen used to stimulate the responder T cells, prior in vitro amplification of the anti-tumor response may be necessary to allow the characterization of such responses. This has been the strategy used most often by laboratories such as the LICR. The LICR has mainly generated T-cell clones from peripheral blood or from metastases and tested them for tumor reactivity in classic cytotoxicT lymphocyte (CTL) assays and by cytokine production (132,133), or by culture of PBMC with peptide under limiting dilution conditions, followed by tetramer-guided sorting of clones (134136). This approach gives a frequency of anti-TAA responses based on very limited CD8+ or CD4+ T-cell numbers capable of sustained in vitro proliferation and may not be truly representative of the in vivo situation. However, the advantage of this methodology is that it yields T-cell clones that can be examined in greater detail than otherwise possible, for example the analysis of antigen recognition, functional avidity and composition of the T-cell receptor (TCR); recognition of tumor cells from the same or different types; design of clonotype-specific polymerase chain reaction (PCR) primers for the ex vivo analysis of localization and frequency prior to and following immunotherapy; correlations with effective versus failing immune control; behavior in tumor metastases, etc.

The peripheral blood is most often sampled to examine the anti-tumor response; however, tumor-reactive T cells may accumulate at or near to the tumor site. Our studies have demonstrated that in patients following allogeneic HSCT from their related HLA-matched donors, anti-TAA CD8+ T-cell responses can be identified almost exclusively in the bone marrow and the site of the malignancy (28), suggesting that the monitoring of an anti-tumor response may miss the anti-tumor response if the wrong compartment is sampled. Similarly higher frequencies of anti-tumor T cells were found within the melanoma by Mazzocchi et al. (137) and Carrasco et al. (133). Carrasco et al. (133) identified anti- melanoma antigen (MAGE) CD4+ and CD8+ T cells in the peripheral blood and slightly enriched in slowly progressing metastases. The most interesting finding was that a T-cell response against a non-vaccine antigen, MAGE C2, was detected following vaccination and enriched by over three logs in metastases relative to blood, suggesting that effective anti-tumor responses preferentially localized to the tumor. Mazzocchi et al. (137) similarly demonstrated a higher preponderance of anti-tumor T cells at the site of the tumor than in the peripheral blood. This phenomenon of determinant spreading, possibly mediated via the generation of an inflammatory environment by some tumor-reactive T cells, which induces apoptosis of tumor cells followed by cross-presentation of tumor-derived antigens, has also been observed in other vaccine settings (138140) and may be an important mechanism of vaccine-associated anti-tumor immunity. Thus the question why the same vaccine works in some patients and not in others could be related to whether or not the patient can mount a T-cell response to antigens presented by the same tumor (12). Therefore, in the monitoring of vaccine therapies, ideally the response of a patient’s T cells against the vaccine but also the tumor itself should be analyzed.

In situations where the frequency of tumor-reactive T cells is higher (above 0.1%), other methodologies for direct ex vivo analysis that are less time consuming and more informative can be employed. The days where we would enumerate antigen-specific helper and cytotoxic precursor frequencies using limiting dilution assays have largely been replaced by polychromatic flow cytometry assays that quantify the frequencies, phenotypes and an array of functions of antigen-responsive T cells (28,141). With the introduction of soluble HLA class I (142) and class II (143) multimers, antigen-specific T cells can be examined directly and isolated for further analyzes, for example molecular analysis of the T-cell repertoire (144146). Obviously, the flow cytometry-based functional and phenotypic characterization of anti-tumor responses is preferable above other methodologies for the ex vivo monitoring of pre-existing anti-tumor responses and the effect vaccination and HSCT have on the quality and quantity of anti-TAA and virus responses (28,147), but the molecular characterization of TAA-specific CD4+ and CD8+ T cells could bring us one step closer to a better understanding of T-cell clones that may have contributed to TAA recognition (88,148150) and tumor eradication. An example where clonotype analysis has provided insight into features of protective immunity has recently been uncovered in a non-human primate model of human HIV infection, where the expansion of CD8+ T cells with a particular TCR-β sequence appeared to correlate with protection; these findings where then confirmed in a vaccine setting in the same model (151).

How can we generalize the success of tumor vaccine therapy?

Currently in any immunotherapy study only a few patients show a response. However, detailed study of responders may open the way to generalizing treatment strategies to make them effective for the majority of patients. Coulie et al. (12) described three patients vaccinated with autologous melanoma clones who had unusually favorable clinical courses that may have been the result of either the antigenicity of their tumors, their susceptibility to lysis by their autologous T-cell clones, or specifics of the responder population that made their efficacy so high. It seems unlikely that the antigenicity could have explained the favorable clinical progression, as the antigens identified in these three patients were expressed by a large number of melanomas. Rather, the immune response seems to have displayed qualities that allowed the eradication of initial and subsequent melanoma clones: all three patients had CTL targeting at least five distinct antigens. Thus the molecular characterization of the TCR of these cells could hint at a strategy that might provide a novel treatment modality in other patients harboring melanomas with similar HLA constitution but less efficient induction of a T-cell response against their autologous tumors. Rather than having to rely on their immune efficacy, one could simply target their malignancies with a multitude of distinct TCR.

To exploit fully the success of immunotherapy, mH antigen and TAA-specific T-cell clones will have to be generated to identify the TCR α- and β-chains and, via gene transfer studies, define their role in tumor eradication (see below). Antigen-specific T cells can be isolated from peripheral blood, bone marrow, tumors and other compartments by electronic sorting of T cells with defined specificities using HLA class I or II tetramers or, if the antigen is unknown, via the isolation of T cells that up-regulate CD40 ligand (152,153), CD107a (154) and CD137 (155) upon antigen recognition. Such cells can be sorted as lines and expanded by one stimulation with the selecting antigen source. The T-cell lines could then be examined for clonotype composition, and cloned to allow detailed functional analyzes and characterization of the TCRαβ pairs per clone. Clonotype analysis may identify T-cell clones with potential clinical relevance. With the rapid improvements in the ectopic expression of TCR (156161), this antigen specificity can be transferred to non-expressing cells, thereby conferring the therapeutic potential of the original T-cell clone onto new cells (162,163). Pre-clinical studies in animal models could help identify TCRαβ that could control and eradicate human tumor tissue. Such studies could help select TCR with the greatest potential, which could subsequently be used in phase I trials.

Conclusion

Studies on the interaction between tumors and the immune system have led to the identification of many TAA in the autologous setting and mH antigens in the HLA-matched sibling transplant settings. These studies have not only proved that various classes of tumor antigens are targeted by the immune system, but also that tumors escape by changing their phenotype (clonal evolution) and modifying the tumor environment. The molecular characterization of TAA brought the promise that antigens could be used in vaccine trials to boost an existing immune response to the malignancy. Tumor immunotherapy still has far to go to be universally successful, but we are making progress. Vaccines have great appeal in their relative simplicity; however, this approach depends mainly on how well a patient can mount an affective immune response to the vaccine or other tumor antigens. Through detailed immunologic studies we can define the criteria for a successful T-cell response. TCR gene transfer now provides the opportunity to exploit these molecularly characterized successful anti-tumor T-cell clones in the treatment of other patients.

Acknowledgments

This work was supported by the Intramural Research Program of the National Institutes of Health, National Heart, Lung, and Blood Institute.

Footnotes

Disclosure of interests: the authors declare no conflicts of interest.

References

  • 1.Jones S, Chen WD, Parmigiani G, Diehl F, Beerenwinkel N, Antal T, et al. Comparative lesion sequencing provides insights into tumor evolution. Proc Natl Acad Sci USA 2008;105:4283–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Fialkow P, Singer J, Adamson J, Vaidya K, Dow L, Ochs J, et al. Acute nonlymphocytic leukemia: heterogeneity of stem cell origin. Blood 1981;57:1068–73. [PubMed] [Google Scholar]
  • 3.Fialkow PJ, Singer JW, Raskind WH, Adamson JW, Jacobson RJ, Bernstein ID, et al. Clonal development, stem-cell differentiation, and clinical remissions in acute nonlymphocytic leukemia. N Engl J Med 1987;317:468–73. [DOI] [PubMed] [Google Scholar]
  • 4.Fearon ER, Hamilton SR, Vogelstein B. Clonal analysis of human colorectal tumors. Science 1987;238:193–7. [DOI] [PubMed] [Google Scholar]
  • 5.Vogelstein B, Fearon ER, Hamilton SR, Kern SE, Preisinger AC, Leppert M, et al. Genetic alterations during colorectal-tumor development. N Engl J Med 1988;319:525–32. [DOI] [PubMed] [Google Scholar]
  • 6.Tsao JL, Yatabe Y, Salovaara R, Jarvinen HJ, Mecklin JP, Aaltonen LA, et al. Genetic reconstruction of individual colorectal tumor histories. Proc Natl Acad Sci USA 2000;97: 1236–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Cahill DP, Kinzler KW, Vogelstein B, Lengauer C. Genetic instability and Darwinian selection in tumours. Trends Cell Biol 1999;9:M57–60. [PubMed] [Google Scholar]
  • 8.Battegay M, Elzi L. Morbidity and mortality in HIV-infected individuals: a shift towards comorbidities. Swiss Med Wkly 2009;139:564–70. [DOI] [PubMed] [Google Scholar]
  • 9.Ramirez-Olivencia G, Valencia-Ortega ME, Martin-Carbonero L, Moreno-Celda V, Gonzalez-Lahoz J. [Malignancies in HIV infected patients: study of 139 cases]. Med Clin (Barc) 2009;133:729–35. [DOI] [PubMed] [Google Scholar]
  • 10.Marin-Muller C, Li M, Chen C,Yao Q. Current understanding and potential immunotherapy for HIV-associated squamous cell carcinoma of the anus (SCCA). World J Surg 2009;33:653–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Yarchoan R, Tosato G, Little RF. Therapy insight: AIDS-related malignancies: the influence of antiviral therapy on pathogenesis and management. Nat Clin Pract Oncol 2005;2:406–15, 23. [DOI] [PubMed] [Google Scholar]
  • 12.Coulie PG, Ikeda H, Baurain JF, Chiari R. Antitumor immunity at work in a melanoma patient. Adv Cancer Res 1999;76:213–42. [DOI] [PubMed] [Google Scholar]
  • 13.Vago L, Perna SK, Zanussi M, Mazzi B, Barlassina C, Stanghellini MT, et al. Loss of mismatched HLA in leukemia after stem-cell transplantation. N Engl J Med 2009;361:478–88. [DOI] [PubMed] [Google Scholar]
  • 14.Rosenberg SA, Packard BS, Aebersold PM, Solomon D, Topalian SL, Toy ST, et al. Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma. A preliminary report. N Engl J Med 1988;319:1676–80. [DOI] [PubMed] [Google Scholar]
  • 15.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;91:1653–61. [PubMed] [Google Scholar]
  • 16.Dao T, Scheinberg DA. Peptide vaccines for myeloid leukaemias. Best Pract Res Clin Haematol 2008;21:391–404. [DOI] [PubMed] [Google Scholar]
  • 17.Oka Y, Tsuboi A, Kawakami M, Elisseeva OA, Nakajima H, Udaka K, et al. Development of WT1 peptide cancer vaccine against hematopoietic malignancies and solid cancers. Curr Med Chem 2006;13:2345–52. [DOI] [PubMed] [Google Scholar]
  • 18.Rusakiewicz S, Molldrem JJ. Immunotherapeutic peptide vaccination with leukemia-associated antigens. Curr Opin Immunol 2006;18:599–604. [DOI] [PubMed] [Google Scholar]
  • 19.Boon T, Coulie PG, Van den Eynde BJ, van der Bruggen P. Human T cell responses against melanoma. Annu Rev Immunol 2006;24:175–208. [DOI] [PubMed] [Google Scholar]
  • 20.Van den Eynde B, Van der Bruggen P. Peptide database of T cell defined tumor antigens. Academy of Cancer Immunology http://www.cancerimmunity.org/peptidedatabase/Tcellepitopes.htm. 2010. [DOI] [PubMed]
  • 21.Akatsuka Y, Morishima Y, Kuzushima K, Kodera Y, Takahashi T. Minor histocompatibility antigens as targets for immunotherapy using allogeneic immune reactions. Cancer Sci 2007;98:1139–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Lucas S, Coulie PG. About human tumor antigens to be used in immunotherapy. Semin Immunol 2008;20:301–7. [DOI] [PubMed] [Google Scholar]
  • 23.Molldrem J, Dermime S, Parker K, Jiang YZ, Mavroudis D, Hensel N, 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]
  • 24.Molldrem JJ, Clave E, Jiang YZ, Mavroudis D, Raptis A, Hensel N, et al. Cytotoxic T lymphocytes specific for a non-polymorphic proteinase 3 peptide preferentially inhibit chronic myeloid leukemia colony-forming units. Blood 1997; 90:2529–34. [PubMed] [Google Scholar]
  • 25.Fujiwara H, El Ouriaghli F, Grube M, Price DA, Rezvani K, Gostick E, et al. Identification and in vitro expansion of CD4+ and CD8+ T cells specific for human neutrophil elastase. Blood 2004;103:3076–83. [DOI] [PubMed] [Google Scholar]
  • 26.Fujiwara H, Melenhorst JJ, El Ouriaghli F, Kajigaya S, Grube M, Sconocchia G, et al. In vitro induction of myeloid leukemia-specific CD4 and CD8 T cells by CD40 ligand-activated B cells gene modified to express primary granule proteins. Clin Cancer Res 2005;11:4495–503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Molldrem JJ, Lee PP, Wang C, Felio K, Kantarjian HM, Champlin RE, et al. Evidence that specific T lymphocytes may participate in the elimination of chronic myelogenous leukemia. Nat Med 2000;6:1018–23. [DOI] [PubMed] [Google Scholar]
  • 28.Melenhorst JJ, Scheinberg P, Chattopadhyay PK, Gostick E, Ladell K, Roederer M, et al. High avidity myeloid leukemia-associated antigen-specific CD8+ T cells preferentially reside in the bone marrow. Blood 2009;113:2238–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Melenhorst JJ, Scheinberg P, Chattopadhyay PK, Lissina A, Gostick E, Cole DK, et al. Detection of low avidity CD8(+) T cell populations with coreceptor-enhanced peptide-major histocompatibility complex class I tetramers. J Immunol Methods 2008;338:31–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Molldrem JJ, Lee PP, Kant S, Wieder E, Jiang W, Lu S, et al. Chronic myelogenous leukemia shapes host immunity by selective deletion of high-avidity leukemia-specific T cells. J Clin Invest 2003;111:639–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Inoue K, Ogawa H, Sonoda Y, Kimura T, Sakabe H, Oka Y, et al. Aberrant overexpression of the Wilms tumor gene (WT1) in human leukemia. Blood 1997;89:1405–12. [PubMed] [Google Scholar]
  • 32.Inoue K, Sugiyama H, Ogawa H, Nakagawa M, Yamagami T, Miwa H, et al. WT1 as a new prognostic factor and a new marker for the detection of minimal residual disease in acute leukemia. Blood 1994;84:3071–9. [PubMed] [Google Scholar]
  • 33.Keilholz U, Menssen HD, Gaiger A, Menke A, Oji Y, Oka Y, et al. Wilms’ tumour gene 1 (WT1) in human neoplasia. Leukemia 2005;19:1318–23. [DOI] [PubMed] [Google Scholar]
  • 34.Yamagami T, Sugiyama H, Inoue K, Ogawa H, Tatekawa T, Hirata M, 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]
  • 35.Hubinger G, Schmid M, Linortner S, Manegold A, Bergmann L, Maurer U. Ribozyme-mediated cleavage of wt1 transcripts suppresses growth of leukemia cells. Exp Hematol 2001;29:1226–35. [DOI] [PubMed] [Google Scholar]
  • 36.Tatsumi N, Oji Y, Tsuji N, Tsuda A, Higashio M, Aoyagi S, et al. Wilms’ tumor gene WT1-shRNA as a potent apoptosis-inducing agent for solid tumors. Int J Oncol 2008;32: 701–11. [PubMed] [Google Scholar]
  • 37.King-Underwood L, Renshaw J, Pritchard-Jones K. Mutations in the Wilms’ tumor gene WT1 in leukemias. Blood 1996;87:2171–9. [PubMed] [Google Scholar]
  • 38.Tosello V, Mansour MR, Barnes K, Paganin M, Sulis ML, Jenkinson S, et al. WT1 mutations in T-ALL. Blood 2009; 114:1038–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Hollink IH, van den Heuvel-Eibrink MM, Zimmermann M, Balgobind BV, Arentsen-Peters ST, Alders M, et al. Clinical relevance of Wilms tumor 1 gene mutations in childhood acute myeloid leukemia. Blood 2009;113:5951–60. [DOI] [PubMed] [Google Scholar]
  • 40.King-Underwood L, Pritchard-Jones K.Wilms’ tumor (WT1) gene mutations occur mainly in acute myeloid leukemia and may confer drug resistance. Blood 1998;91:2961–8. [PubMed] [Google Scholar]
  • 41.Renneville A, Boissel N, Zurawski V, Llopis L, Biggio V, Nibourel O, et al. Wilms tumor 1 gene mutations are associated with a higher risk of recurrence in young adults with acute myeloid leukemia: a study from the Acute Leukemia French Association. Cancer 2009;115:3719–27. [DOI] [PubMed] [Google Scholar]
  • 42.Willasch AM, Gruhn B, Coliva T, Kalinova M, Schneider G, Kreyenberg H, et al. Standardization of WT1 mRNA quantitation for minimal residual disease monitoring in childhood AML and implications of WT1 gene mutations: a European multicenter study. Leukemia 2009;23:1472–9. [DOI] [PubMed] [Google Scholar]
  • 43.Barrett AJ, Melenhorst JJ. Minor histocompatibility antigen discovery: turning up the HEATR. Blood 2010;115: 4630–1. [DOI] [PubMed] [Google Scholar]
  • 44.den Haan JM, Meadows LM, Wang W, Pool J, Blokland E, Bishop TL, et al. The minor histocompatibility antigen HA-1: a diallelic gene with a single amino acid polymorphism. Science 1998;279:1054–7. [DOI] [PubMed] [Google Scholar]
  • 45.Pierce RA, Field ED, Mutis T, Golovina TN, Von Kap-Herr C, Wilke M, et al. The HA-2 minor histocompatibility antigen is derived from a diallelic gene encoding a novel human class I myosin protein. J Immunol 2001;167:3223–30. [DOI] [PubMed] [Google Scholar]
  • 46.Dolstra H, Fredrix H, Maas F, Coulie PG, Brasseur F, Mensink E, et al. A human minor histocompatibility antigen specific for B cell acute lymphoblastic leukemia. J Exp Med 1999;189:301–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.de Rijke B, van Horssen-Zoetbrood A, Beekman JM, Otterud B, Maas F, Woestenenk R, et al. A frameshift polymorphism in P2X5 elicits an allogeneic cytotoxic T lymphocyte response associated with remission of chronic myeloid leukemia. J Clin Invest 2005;115:3506–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Spaapen RM, Lokhorst HM, van den Oudenalder K, Otterud BE, Dolstra H, Leppert MF, et al. Toward targeting B cell cancers with CD4+ CTLs: identification of a CD19-encoded minor histocompatibility antigen using a novel genome-wide analysis. J Exp Med 2008;205:2863–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Bleakley M, Otterud BE, Richardt JL, Mollerup AD, Hudecek M, Nishida T, et al. Leukemia-associated minor histocompatibility antigen discovery using T-cell clones isolated by in vitro stimulation of naive CD8+ T cells. Blood 2010;115:4923–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Spierings E, Gras S, Reiser JB, Mommaas B, Almekinders M, Kester MG, et al. Steric hindrance and fast dissociation explain the lack of immunogenicity of the minor histocompatibility HA-1Arg Null allele. J Immunol 2009;182:4809–16. [DOI] [PubMed] [Google Scholar]
  • 51.Takahashi Y, Harashima N, Kajigaya S, Yokoyama H, Cherkasova E, McCoy JP, et al. Regression of human kidney cancer following allogeneic stem cell transplantation is associated with recognition of an HERV-E antigen by T cells. J Clin Invest 2008;118:1099–109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Bannert N, Kurth R. The evolutionary dynamics of human endogenous retroviral families. Annu Rev Genomics Hum Genet 2006;7:149–73. [DOI] [PubMed] [Google Scholar]
  • 53.Oka Y, Tsuboi A, Taguchi T, Osaki T, Kyo T, Nakajima H, 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] [PMC free article] [PubMed] [Google Scholar]
  • 54.Rezvani K, Yong AS, Mielke S, Savani BN, Musse L, Superata J, et al. Leukemia-associated antigen-specific T-cell responses following combined PR1 and WT1 peptide vaccination in patients with myeloid malignancies. Blood 2008; 111:236–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Keilholz U, Letsch A, Busse A, Asemissen AM, Bauer S, Blau IW, 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;113:6541–8. [DOI] [PubMed] [Google Scholar]
  • 56.Janssen EM, Lemmens EE,Wolfe T, Christen U, von Herrath MG, Schoenberger SP. CD4+ T cells are required for secondary expansion and memory in CD8+ T lymphocytes. Nature 2003;421:852–6. [DOI] [PubMed] [Google Scholar]
  • 57.Janssen EM, Droin NM, Lemmens EE, Pinkoski MJ, Bensinger SJ, Ehst BD, et al. CD4+ T-cell help controls CD8+ T-cell memory via TRAIL-mediated activation-induced cell death. Nature 2005;434:88–93. [DOI] [PubMed] [Google Scholar]
  • 58.Shedlock DJ, Shen H. Requirement for CD4 T cell help in generating functional CD8 T cell memory. Science 2003; 300:337–9. [DOI] [PubMed] [Google Scholar]
  • 59.Ryu SJ, Jung KM, Yoo HS, Kim TW, Kim S, Chang J, et al. Cognate CD4 help is essential for the reactivation and expansion of CD8 memory T cells directed against the hematopoietic cell-specific dominant minor histocompatibility antigen, H60. Blood 2009;113:4273–80. [DOI] [PubMed] [Google Scholar]
  • 60.Riddell SR, Watanabe KS, Goodrich JM, Li CR, Agha ME, Greenberg PD. Restoration of viral immunity in immunodeficient humans by the adoptive transfer of T cell clones. Science 1992;257:238–41. [DOI] [PubMed] [Google Scholar]
  • 61.Walter EA, Greenberg PD, Gilbert MJ, Finch RJ, Watanabe KS, Thomas ED, 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] [PubMed] [Google Scholar]
  • 62.Jiang YZ, Mavroudis D, Dermime S, Hensel N, Couriel D, Molldrem J, et al. Alloreactive CD4+ T lymphocytes can exert cytotoxicity to chronic myeloid leukaemia cells processing and presenting exogenous antigen. Br J Haematol 1996;93:606–12. [DOI] [PubMed] [Google Scholar]
  • 63.Faber LM, van Luxemburg-Heijs SA, Veenhof WF, Willemze R, Falkenburg JH. Generation of CD4+ cytotoxic T-lymphocyte clones from a patient with severe graft-versus-host disease after allogeneic bone marrow transplantation: implications for graft-versus-leukemia reactivity. Blood 1995;86:2821–8. [PubMed] [Google Scholar]
  • 64.Dodi IA,Van Rhee F, Forde HC, Roura-Mir C, Jaraquemada D, Goldman JM, et al. CD4(+) bias in T cells cloned from a CML patient with active graft versus leukemia effect. Cytotherapy 2002;4:353–63. [DOI] [PubMed] [Google Scholar]
  • 65.Yasukawa M, Ohminami H, Kaneko S, Yakushijin Y, Nishimura Y, Inokuchi K, et al. CD4(+) cytotoxic T-cell clones specific for bcr-abl b3a2 fusion peptide augment colony formation by chronic myelogenous leukemia cells in a b3a2-specific and HLA-DR-restricted manner. Blood 1998;92:3355–61. [PubMed] [Google Scholar]
  • 66.Griffioen M, van der Meijden ED, Slager EH, Honders MW, Rutten CE, van Luxemburg-Heijs SA, et al. Identification of phosphatidylinositol 4-kinase type II beta as HLA class II-restricted target in graft versus leukemia reactivity. Proc Natl Acad Sci USA 2008;105:3837–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Nishimura M, Uchida S, Mitsunaga S, Yahagi Y, Nakajima K, Tadokoro K, et al. Characterization of T-cell clones derived from peripheral blood lymphocytes of a patient with transfusion-associated graft-versus-host disease: Fas-mediated killing by CD4+ and CD8+ cytotoxic T-cell clones and tumor necrosis factor beta production by CD4+ T-cell clones. Blood 1997;89:1440–5. [PubMed] [Google Scholar]
  • 68.Appay V The physiological role of cytotoxic CD4(+) T-cells: the holy grail? Clin Exp Immunol 2004;138:10–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Casazza JP, Betts MR, Price DA, Precopio ML, Ruff LE, Brenchley JM, et al. Acquisition of direct antiviral effector functions by CMV-specific CD4+ T lymphocytes with cellular maturation. J Exp Med 2006;203:2865–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Guo Y, Niiya H, Azuma T, Uchida N, Yakushijin Y, Sakai I, et al. Direct recognition and lysis of leukemia cells by WT1-specific CD4+ T lymphocytes in an HLA class II-restricted manner. Blood 2005;106:1415–8. [DOI] [PubMed] [Google Scholar]
  • 71.Zorn E, Wang KS, Hochberg EP, Canning C, Alyea EP, Soiffer RJ, et al. Infusion of CD4+ donor lymphocytes induces the expansion of CD8+ donor T cells with cytolytic activity directed against recipient hematopoietic cells. Clin Cancer Res 2002;8:2052–60. [PubMed] [Google Scholar]
  • 72.Giralt S, Hester J, Huh Y, Hirsch-Ginsberg C, Rondon G, Seong D, et al. CD8-depleted donor lymphocyte infusion as treatment for relapsed chronic myelogenous leukemia after allogeneic bone marrow transplantation. Blood 1995;86:4337–43. [PubMed] [Google Scholar]
  • 73.Alyea EP, Soiffer RJ, Canning C, Neuberg D, Schlossman R, Pickett C, et al. Toxicity and efficacy of defined doses of CD4(+) donor lymphocytes for treatment of relapse after allogeneic bone marrow transplant. Blood 1998;91: 3671–80. [PubMed] [Google Scholar]
  • 74.Mocellin S, Mandruzzato S, Bronte V, Lise M, Nitti D. I. Vaccines for solid tumours. Lancet Oncol 2004;5:681–9. [DOI] [PubMed] [Google Scholar]
  • 75.Mocellin S, Semenzato G, Mandruzzato S, Riccardo Rossi C. II. Vaccines for haematological malignant disorders. Lancet Oncol 2004;5:727–37. [DOI] [PubMed] [Google Scholar]
  • 76.Lonchay C, van der Bruggen P, Connerotte T, Hanagiri T, Coulie P, Colau D, et al. Correlation between tumor regression and T cell responses in melanoma patients vaccinated with a MAGE antigen. Proc Natl Acad Sci USA 2004; 101(Suppl 2):14631–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Guan Y, Gerhard B, Hogge DE. Detection, isolation, and stimulation of quiescent primitive leukemic progenitor cells from patients with acute myeloid leukemia (AML). Blood 2003;101:3142–9. [DOI] [PubMed] [Google Scholar]
  • 78.Hope KJ, Jin L, Dick JE. Acute myeloid leukemia originates from a hierarchy of leukemic stem cell classes that differ in self-renewal capacity. Nat Immunol 2004;5:738–43. [DOI] [PubMed] [Google Scholar]
  • 79.Jamieson CH, Ailles LE, Dylla SJ, Muijtjens M, Jones C, Zehnder JL, et al. Granulocyte–macrophage progenitors as candidate leukemic stem cells in blast-crisis CML. N Engl J Med 2004;351:657–67. [DOI] [PubMed] [Google Scholar]
  • 80.Dick JE. Stem cell concepts renew cancer research. Blood 2008;112:4793–807. [DOI] [PubMed] [Google Scholar]
  • 81.Kolb HJ, Mittermuller J, Clemm C, Holler E, Ledderose G, Brehm G, et al. Donor leukocyte transfusions for treatment of recurrent chronic myelogenous leukemia in marrow transplant patients. Blood 1990;76:2462–5. [PubMed] [Google Scholar]
  • 82.Slavin S, Naparstek E, Nagler A, Ackerstein A, Samuel S, Kapelushnik J, et al. Allogeneic cell therapy with donor peripheral blood cells and recombinant human interleukin-2 to treat leukemia relapse after allogeneic bone marrow transplantation. Blood 1996;87:2195–204. [PubMed] [Google Scholar]
  • 83.von dem Borne PA, van Luxemburg-Heijs SA, Heemskerk MH, Jedema I, Mulder A, Willemze R, et al. Molecular persistence of chronic myeloid leukemia caused by donor T cells specific for lineage-restricted maturation antigens not recognizing immature progenitor-cells. Leukemia 2006;20:1040–6. [DOI] [PubMed] [Google Scholar]
  • 84.Essers MA, Offner S, Blanco-Bose WE, Waibler Z, Kalinke U, Duchosal MA, et al. IFNalpha activates dormant haematopoietic stem cells in vivo. Nature 2009;458:904–8. [DOI] [PubMed] [Google Scholar]
  • 85.Vence L, Palucka AK, Fay JW, Ito T, Liu YJ, Banchereau J, et al. Circulating tumor antigen-specific regulatory T cells in patients with metastatic melanoma. Proc Natl Acad Sci USA 2007;104:20884–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Nicholaou T, Ebert LM, Davis ID, McArthur GA, Jackson H, Dimopoulos N, et al. Regulatory T-cell-mediated attenuation of T-cell responses to the NY-ESO-1 ISCOMATRIX vaccine in patients with advanced malignant melanoma. Clin Cancer Res 2009;15:2166–73. [DOI] [PubMed] [Google Scholar]
  • 87.Francois V, Ottaviani S, Renkvist N, Stockis J, Schuler G, Thielemans K, et al. The CD4(+) T-cell response of melanoma patients to a MAGE-A3 peptide vaccine involves potential regulatory T cells. Cancer Res 2009;69:4335–45. [DOI] [PubMed] [Google Scholar]
  • 88.Jandus C, Bioley G, Dojcinovic D, Derre L, Baitsch L, Wieckowski S, et al. Tumor antigen-specific FOXP3+ CD4 T cells identified in human metastatic melanoma: peptide vaccination results in selective expansion of Th1-like counterparts. Cancer Res 2009;69:8085–93. [DOI] [PubMed] [Google Scholar]
  • 89.Lehe C, Ghebeh H, Al-Sulaiman A, Al Qudaihi G, Al-Hussein K, Almohareb F, et al. The Wilms’ tumor antigen is a novel target for human CD4+ regulatory T cells: implications for immunotherapy. Cancer Res 2008;68:6350–9. [DOI] [PubMed] [Google Scholar]
  • 90.Bonertz A, Weitz J, Pietsch DH, Rahbari NN, Schlude C, Ge Y, et al. Antigen-specific Tregs control T cell responses against a limited repertoire of tumor antigens in patients with colorectal carcinoma. J Clin Invest 2009;119:3311–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Aspord C, Pedroza-Gonzalez A, Gallegos M, Tindle S, Burton EC, Su D, et al. Breast cancer instructs dendritic cells to prime interleukin 13-secreting CD4+ T cells that facilitate tumor development. J Exp Med 2007;204:1037–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Lee PP, Yee C, Savage PA, Fong L, Brockstedt D, Weber JS, et al. Characterization of circulating T cells specific for tumor-associated antigens in melanoma patients. Nat Med 1999;5:677–85. [DOI] [PubMed] [Google Scholar]
  • 93.Zippelius A, Batard P, Rubio-Godoy V, Bioley G, Lienard D, Lejeune F, et al. Effector function of human tumor-specific CD8 T cells in melanoma lesions: a state of local functional tolerance. Cancer Res 2004;64:2865–73. [DOI] [PubMed] [Google Scholar]
  • 94.Mortarini R, Piris A, Maurichi A, Molla A, Bersani I, Bono A, et al. Lack of terminally differentiated tumor-specific CD8+ T cells at tumor site in spite of antitumor immunity to self-antigens in human metastatic melanoma. Cancer Res 2003;63:2535–45. [PubMed] [Google Scholar]
  • 95.Yasui H,Takai K,Yoshida R, Hayaishi O. Interferon enhances tryptophan metabolism by inducing pulmonary indoleamine 2,3-dioxygenase: its possible occurrence in cancer patients. Proc Natl Acad Sci USA 1986;83:6622–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Munn DH, Sharma MD, Lee JR, Jhaver KG, Johnson TS, Keskin DB, et al. Potential regulatory function of human dendritic cells expressing indoleamine 2,3-dioxygenase. Science 2002;297:1867–70. [DOI] [PubMed] [Google Scholar]
  • 97.Grohmann U, Orabona C, Fallarino F, Vacca C, Calcinaro F, Falorni A, et al. CTLA-4-Ig regulates tryptophan catabolism in vivo. Nat Immunol 2002;3:1097–101. [DOI] [PubMed] [Google Scholar]
  • 98.Guillonneau C, Hill M, Hubert FX, Chiffoleau E, Herve C, Li XL, et al. CD40Ig treatment results in allograft acceptance mediated by CD8CD45RCT cells, IFN-gamma, and indoleamine 2,3-dioxygenase. J Clin Invest 2007;117:1096–106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Mellor AL, Keskin DB, Johnson T, Chandler P, Munn DH. Cells expressing indoleamine 2,3-dioxygenase inhibit T cell responses. J Immunol 2002;168:3771–6. [DOI] [PubMed] [Google Scholar]
  • 100.Munn DH, Shafizadeh E, Attwood JT, Bondarev I, Pashine A, Mellor AL. Inhibition of T cell proliferation by macrophage tryptophan catabolism. J Exp Med 1999;189:1363–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Frumento G, Rotondo R, Tonetti M, Damonte G, Benatti U, Ferrara GB.Tryptophan-derived catabolites are responsible for inhibition of T and natural killer cell proliferation induced by indoleamine 2,3-dioxygenase. J Exp Med. 2002;196:459–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Munn DH, Sharma MD, Hou D, Baban B, Lee JR, Antonia SJ, et al. Expression of indoleamine 2,3-dioxygenase by plasmacytoid dendritic cells in tumor-draining lymph nodes. J Clin Invest 2004;114:280–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Munn DH, Zhou M, Attwood JT, Bondarev I, Conway SJ, Marshall B, et al. Prevention of allogeneic fetal rejection by tryptophan catabolism. Science 1998;281:1191–3. [DOI] [PubMed] [Google Scholar]
  • 104.Uyttenhove C, Pilotte L, Theate I, Stroobant V, Colau D, Parmentier N, et al. Evidence for a tumoral immune resistance mechanism based on tryptophan degradation by indoleamine 2,3-dioxygenase. Nat Med 2003;9:1269–74. [DOI] [PubMed] [Google Scholar]
  • 105.Curti A, Aluigi M, Pandolfi S, Ferri E, Isidori A, Salvestrini V, et al. Acute myeloid leukemia cells constitutively express the immunoregulatory enzyme indoleamine 2,3-dioxygenase. Leukemia 2007;21:353–5. [DOI] [PubMed] [Google Scholar]
  • 106.Murdoch C, Muthana M, Coffelt SB, Lewis CE. The role of myeloid cells in the promotion of tumour angiogenesis. Nat Rev Cancer 2008;8:618–31. [DOI] [PubMed] [Google Scholar]
  • 107.Filipazzi P, Valenti R, Huber V, Pilla L, Canese P, Iero M, et al. Identification of a new subset of myeloid suppressor cells in peripheral blood of melanoma patients with modulation by a granulocyte–macrophage colony-stimulation factor-based antitumor vaccine. J Clin Oncol 2007;25:2546–53. [DOI] [PubMed] [Google Scholar]
  • 108.Bronte V Myeloid-derived suppressor cells in inflammation: uncovering cell subsets with enhanced immunosuppressive functions. Eur J Immunol 2009;39:2670–2. [DOI] [PubMed] [Google Scholar]
  • 109.Kusmartsev S, Su Z, Heiser A, Dannull J, Eruslanov E, Kubler H, et al. Reversal of myeloid cell-mediated immunosuppression in patients with metastatic renal cell carcinoma. Clin Cancer Res 2008;14:8270–8. [DOI] [PubMed] [Google Scholar]
  • 110.Hoechst B, Ormandy LA, Ballmaier M, Lehner F, Kruger C, Manns MP, et al. A new population of myeloid-derived suppressor cells in hepatocellular carcinoma patients induces CD4(+)CD25(+)Foxp3(+) T cells. Gastroenterology 2008; 135:234–43. [DOI] [PubMed] [Google Scholar]
  • 111.Hoechst B, Voigtlaender T, Ormandy L, Gamrekelashvili J, Zhao F, Wedemeyer H, et al. Myeloid derived suppressor cells inhibit natural killer cells in patients with hepatocellular carcinoma via the NKp30 receptor. Hepatology 2009;50:799–807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Zea AH, Rodriguez PC, Atkins MB, Hernandez C, Signoretti S, Zabaleta J, et al. Arginase-producing myeloid suppressor cells in renal cell carcinoma patients: a mechanism of tumor evasion. Cancer Res 2005;65:3044–8. [DOI] [PubMed] [Google Scholar]
  • 113.Rodriguez PC, Ernstoff MS, Hernandez C, Atkins M, Zabaleta J, Sierra R, et al. Arginase I-producing myeloid-derived suppressor cells in renal cell carcinoma are a subpopulation of activated granulocytes. Cancer Res 2009;69:1553–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Ochoa AC, Zea AH, Hernandez C, Rodriguez PC. Arginase, prostaglandins, and myeloid-derived suppressor cells in renal cell carcinoma. Clin Cancer Res 2007;13:721–6s. [DOI] [PubMed] [Google Scholar]
  • 115.Vuk-Pavlovic S, Bulur PA, Lin Y, Qin R, Szumlanski CL, Zhao X, et al. Immunosuppressive CD14+HLA-DRlow/– monocytes in prostate cancer. Prostate 2010;70:443–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Mandruzzato S, Solito S, Falisi E, Francescato S, Chiarion-Sileni V, Mocellin S, et al. IL4Ralpha+ myeloid-derived suppressor cell expansion in cancer patients. J Immunol 2009;182:6562–8. [DOI] [PubMed] [Google Scholar]
  • 117.Liu CY, Wang YM, Wang CL, Feng PH, Ko HW, Liu YH, et al. Population alterations of L: –arginase– and inducible nitric oxide synthase-expressed CD11b(+)/CD14 (–)/CD15 (+)/CD33 (+) myeloid-derived suppressor cells and CD8 (+) T lymphocytes in patients with advanced-stage non-small cell lung cancer. J Cancer Res Clin Oncol 2009. Jul 2. [Epub ahead of print]. [DOI] [PubMed]
  • 118.Poschke I, Mougiakakos D, Hansson J, Masucci GV, Kiessling R. Immature immunosuppressive CD14+HLA-DR–/low cells in melanoma patients are Stat3hi and overexpress CD80, CD83, and DC-sign. Cancer Res 2010;70:4335–45. [DOI] [PubMed] [Google Scholar]
  • 119.Rudd CE, Taylor A, Schneider H. CD28 and CTLA-4 coreceptor expression and signal transduction. Immunol Rev 2009;229:12–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Riley JL. PD-1 signaling in primary T cells. Immunol Rev 2009;229:114–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Ahmadzadeh M, Johnson LA, Heemskerk B, Wunderlich JR, Dudley ME, White DE, et al. Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood 2009;114:1537–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Zhang Y, Huang S, Gong D, Qin Y, Shen Q. Programmed death-1 upregulation is correlated with dysfunction of tumor-infiltrating CD8(+) T lymphocytes in human non-small cell lung cancer. Cell Mol Immunol 2010;7:389–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Matsuzaki J, Gnjatic S, Mhawech-Fauceglia P, Beck A, Miller A, Tsuji T, et al. Tumor-infiltrating NY-ESO-1-specific CD8+ T cells are negatively regulated by LAG-3 and PD-1 in human ovarian cancer. Proc Natl Acad Sci USA 2010;107:7875–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Hino R, Kabashima K, Kato Y,Yagi H, Nakamura M, Honjo T, et al. Tumor cell expression of programmed cell death-1 ligand 1 is a prognostic factor for malignant melanoma. Cancer 2010;116:1757–66. [DOI] [PubMed] [Google Scholar]
  • 125.Lee H, Kim JH, Yang SY, Kong J, Oh M, Jeong DH, et al. Peripheral blood gene expression of B7 and CD28 family members associated with tumor progression and microscopic lymphovascular invasion in colon cancer patients. J Cancer Res Clin Oncol 2010;136:1445–52. [DOI] [PubMed] [Google Scholar]
  • 126.Kao J, Ko EC, Eisenstein S, Sikora AG, Fu S, Chen SH. Targeting immune suppressing myeloid-derived suppressor cells in oncology. Crit Rev Oncol/Hematol (2010),doi: 10.1016/j.critrevonc.2010.02.004. [DOI] [PMC free article] [PubMed]
  • 127.Brahmer JR, Drake CG, Wollner I, Powderly JD, Picus J, Sharfman WH, et al. Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol 2010;28:3167–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Albini A, Sporn MB. The tumour microenvironment as a target for chemoprevention. Nat Rev Cancer 2007;7:139–47. [DOI] [PubMed] [Google Scholar]
  • 129.Lob S, Konigsrainer A, Rammensee HG, Opelz G, Terness P. Inhibitors of indoleamine-2,3-dioxygenase for cancer therapy: can we see the wood for the trees? Nat Rev Cancer 2009;9:445–52. [DOI] [PubMed] [Google Scholar]
  • 130.Mangsbo SM, Sandin LC, Anger K, Korman AJ, Loskog A, Totterman TH. Enhanced tumor eradication by combining CTLA-4 or PD-1 blockade with CpG therapy. J Immunother 2010;33:225–35. [DOI] [PubMed] [Google Scholar]
  • 131.Kamei M, Nannya Y, Torikai H, Kawase T, Taura K, Inamoto Y, et al. HapMap scanning of novel human minor histocompatibility antigens. Blood 2009;113:5041–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Coulie PG, Somville M, Lehmann F, Hainaut P, Brasseur F, Devos R, et al. Precursor frequency analysis of human cytolytic T lymphocytes directed against autologous melanoma cells. Int J Cancer 1992;50:289–97. [DOI] [PubMed] [Google Scholar]
  • 133.Carrasco J, Van Pel A, Neyns B, Lethe B, Brasseur F, Renkvist N, et al. Vaccination of a melanoma patient with mature dendritic cells pulsed with MAGE-3 peptides triggers the activity of nonvaccine anti-tumor cells. J Immunol 2008;180:3585–93. [DOI] [PubMed] [Google Scholar]
  • 134.Coulie PG, Karanikas V, Colau D, Lurquin C, Landry C, Marchand M, et al. A monoclonal cytolytic T-lymphocyte response observed in a melanoma patient vaccinated with a tumor-specific antigenic peptide encoded by gene MAGE-3. Proc Natl Acad Sci USA 2001;98:10290–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Karanikas V, Lurquin C, Colau D, van Baren N, De Smet C, Lethe B, et al. Monoclonal anti-MAGE-3 CTL responses in melanoma patients displaying tumor regression after vaccination with a recombinant canarypox virus. J Immunol 2003; 171:4898–904. [DOI] [PubMed] [Google Scholar]
  • 136.van Baren N, Bonnet MC, Dreno B, Khammari A, Dorval T, Piperno-Neumann S, et al. Tumoral and immunologic response after vaccination of melanoma patients with an ALVAC virus encoding MAGE antigens recognized by T cells. J Clin Oncol 2005;23:9008–21. [DOI] [PubMed] [Google Scholar]
  • 137.Mazzocchi A, Belli F, Mascheroni L, Vegetti C, Parmiani G, Anichini A. Frequency of cytotoxic T lymphocyte precursors (CTLp) interacting with autologous tumor via the T-cell receptor: limiting dilution analysis of specific CTLp in peripheral blood and tumor-invaded lymph nodes of melanoma patients. Int J Cancer 1994;58:330–9. [DOI] [PubMed] [Google Scholar]
  • 138.Butterfield LH, Ribas A, Dissette VB, Amarnani SN, Vu HT, Oseguera D, et al. Determinant spreading associated with clinical response in dendritic cell-based immunotherapy for malignant melanoma. Clin Cancer Res 2003;9:998–1008. [PubMed] [Google Scholar]
  • 139.Disis ML, Gooley TA, Rinn K, Davis D, Piepkorn M, Cheever MA, et al. Generation of T-cell immunity to the HER-2/neu protein after active immunization with HER-2/neu peptide-based vaccines. J Clin Oncol 2002;20:2624–32. [DOI] [PubMed] [Google Scholar]
  • 140.Lally KM, Mocellin S, Ohnmacht GA, Nielsen MB, Bettinotti M, Panelli MC, et al. Unmasking cryptic epitopes after loss of immunodominant tumor antigen expression through epitope spreading. Int J Cancer 2001;93:841–7. [DOI] [PubMed] [Google Scholar]
  • 141.Chattopadhyay PK, Melenhorst JJ, Ladell K, Gostick E, Scheinberg P, Barrett AJ, et al. Techniques to improve the direct ex vivo detection of low frequency antigen-specific CD8+ T cells with peptide-major histocompatibility complex class I tetramers. Cytometry A 2008;73:1001–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Altman JD, Moss PA, Goulder PJ, Barouch DH, McHeyzer-Williams MG, Bell JI, et al. Phenotypic analysis of antigen-specific T lymphocytes. Science 1996;274:94–6. [PubMed] [Google Scholar]
  • 143.Novak EJ, Liu AW, Nepom GT, Kwok WW. MHC class II tetramers identify peptide-specific human CD4(+) T cells proliferating in response to influenza A antigen. J Clin Invest 1999;104:R63–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Douek DC, Betts MR, Brenchley JM, Hill BJ, Ambrozak DR, Ngai KL, et al. A novel approach to the analysis of specificity, clonality, and frequency of HIV-specific T cell responses reveals a potential mechanism for control of viral escape. J Immunol 2002;168:3099–104. [DOI] [PubMed] [Google Scholar]
  • 145.Price DA, West SM, Betts MR, Ruff LE, Brenchley JM, Ambrozak DR, et al. T cell receptor recognition motifs govern immune escape patterns in acute SIV infection. Immunity 2004;21:793–803. [DOI] [PubMed] [Google Scholar]
  • 146.Scheinberg P, Melenhorst JJ, Hill BJ, Keyvanfar K, Barrett AJ, Price DA, et al. The clonal composition of human CD4+CD25+Foxp3+ cells determined by a comprehensive DNA-based multiplex PCR for TCRB gene rearrangements. J Immunol Methods 2007;321:107–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Precopio ML, Betts MR, Parrino J, Price DA, Gostick E, Ambrozak DR, et al. Immunization with vaccinia virus induces polyfunctional and phenotypically distinctive CD8(+) T cell responses. J Exp Med 2007;204:1405–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Bioley G, Dousset C, Yeh A, Dupont B, Bhardwaj N, Mears G, et al. Vaccination with recombinant NY-ESO-1 protein elicits immunodominant HLA-DR52b-restricted CD4+ T cell responses with a conserved T cell receptor repertoire. Clin Cancer Res 2009;15:4467–74. [DOI] [PubMed] [Google Scholar]
  • 149.Wieckowski S, Baumgaertner P, Corthesy P, Voelter V, Romero P, Speiser DE, et al. Fine structural variations of alphabetaTCRs selected by vaccination with natural versus altered self-antigen in melanoma patients. J Immunol 2009;183:5397–406. [DOI] [PubMed] [Google Scholar]
  • 150.Derre L, Bruyninx M, Baumgaertner P, Ferber M, Schmid D, Leimgruber A, et al. Distinct sets of alphabeta TCRs confer similar recognition of tumor antigen NY-ESO-1157–165 by interacting with its central Met/Trp residues. Proc Natl Acad Sci USA 2008;105:15010–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Price DA, Asher TE, Wilson NA, Nason MC, Brenchley JM, Metzler IS, et al. Public clonotype usage identifies protective Gag-specific CD8+ T cell responses in SIV infection. J Exp Med 2009;206:923–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Frentsch M, Arbach O, Kirchhoff D, Moewes B, Worm M, Rothe M, et al. Direct access to CD4+ T cells specific for defined antigens according to CD154 expression. Nat Med 2005;11:1118–24. [DOI] [PubMed] [Google Scholar]
  • 153.Chattopadhyay PK, Yu J, Roederer M. A live-cell assay to detect antigen-specific CD4+ T cells with diverse cytokine profiles. Nat Med 2005;11:1113–7. [DOI] [PubMed] [Google Scholar]
  • 154.Betts MR, Brenchley JM, Price DA, De Rosa SC, Douek DC, Roederer M, et al. Sensitive and viable identification of antigen-specific CD8+ T cells by a flow cytometric assay for degranulation. J Immunol Methods 2003;281: 65–78. [DOI] [PubMed] [Google Scholar]
  • 155.Wolfl M, Kuball J, Ho WY, Nguyen H, Manley TJ, Bleakley M, et al. Activation-induced expression of CD137 permits detection, isolation, and expansion of the full repertoire of CD8+ T cells responding to antigen without requiring knowledge of epitope specificities. Blood 2007;110: 201–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Heemskerk MH, de Paus RA, Lurvink EG, Koning F, Mulder A, Willemze R, et al. Dual HLA class I and class II restricted recognition of alloreactive T lymphocytes mediated by a single T cell receptor complex. Proc Natl Acad Sci USA 2001;98:6806–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Cohen CJ, Zhao Y, Zheng Z, Rosenberg SA, Morgan RA. Enhanced antitumor activity of murine-human hybrid T-cell receptor (TCR) in human lymphocytes is associated with improved pairing and TCR/CD3 stability. Cancer Res 2006;66:8878–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Cohen CJ, Li YF, El-Gamil M, Robbins PF, Rosenberg SA, Morgan RA. Enhanced antitumor activity of T cells engineered to express T-cell receptors with a second disulfide bond. Cancer Res 2007;67:3898–903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Okamoto S, Mineno J, Ikeda H, Fujiwara H, Yasukawa M, Shiku H, et al. Improved expression and reactivity of transduced tumor-specific TCRs in human lymphocytes by specific silencing of endogenous TCR. Cancer Res 2009;69: 9003–11. [DOI] [PubMed] [Google Scholar]
  • 160.Jones S, Peng PD, Yang S, Hsu C, Cohen CJ, Zhao Y, et al. Lentiviral vector design for optimal T cell receptor gene expression in the transduction of peripheral blood lymphocytes and tumor-infiltrating lymphocytes. Hum Gene Ther 2009;20:630–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Kuball J, Hauptrock B, Malina V, Antunes E, Voss RH, Wolfl M, et al. Increasing functional avidity of TCR-redirected T cells by removing defined N-glycosylation sites in the TCR constant domain. J Exp Med 2009;206:463–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Morgan RA, Dudley ME, Wunderlich JR, Hughes MS, Yang JC, Sherry RM, et al. Cancer regression in patients after transfer of genetically engineered lymphocytes. Science 2006;314:126–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Johnson LA, Morgan RA, Dudley ME, Cassard L, Yang JC, Hughes MS, et al. Gene therapy with human and mouse T-cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen. Blood 2009;114: 535–46. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES