Skip to main content
Clinical and Experimental Immunology logoLink to Clinical and Experimental Immunology
. 2003 Jan;131(1):1–7. doi: 10.1046/j.1365-2249.2003.02055.x

Prospects for immunotherapy of malignant disease

E C MORRIS 1, G M BENDLE 1, H J STAUSS 1
PMCID: PMC1808603  PMID: 12519379

Abstract

The majority of T cell-recognized tumour antigens in humans are encoded by genes that are also present in normal tissues. Low levels of gene expression in normal cells can lead to the inactivation of high-avidity T cells by immunological tolerance mechanisms. As a consequence, low-avidity T cell responses in patients are often inadequate in providing tumour protection. Recently, several technologies have been developed to overcome tolerance, allowing the isolation of high-affinity, HLA-restricted receptors specific for tumour-associated peptide epitopes. Furthermore, transfer of HLA-restricted antigen receptors provides an opportunity to empower patient T cells with new tumour-reactive specificities that cannot be retrieved from the autologous T cell repertoire.

Keywords: immunotherapy, vaccination, adoptive transfer, allo-restricted CTL, TCR gene transfer

INTRODUCTION

The goal of therapy for malignant disease remains the long-term eradication of tumour cells without adverse effects on normal tissue. Conventional approaches utilizing chemotherapy and radiotherapy are limited by both their lack of specificity and toxicity. Since the first description of a human tumour-associated antigen recognized by cytotoxic T cells (CTL) 10 years ago [1], advances in understanding the nature of tumour-specific immune responses and mechanisms of tolerance induction have encouraged researchers and clinicians alike to develop a more refined approach to immune-mediated therapies. Current immunotherapeutic approaches are many and varied with respect to target specificities, effector mechanisms, mode of administration and prospects for translation into clinical practice.

In this review we will discuss recently developed vaccination strategies that have led to a large number of clinical trials. We will compare the limited clinical success of vaccination with the well-documented curative potential of adoptive tumour therapy with allogeneic T lymphocytes. Finally, we will highlight novel approaches exploiting the allogeneic T cell repertoire and the repertoire of HLA transgenic mice, as well as in vitro selected receptors against defined MHC/peptide combinations, as a basis for antigen-specific gene therapy of cancer.

VACCINATION STRATEGIES

Active vaccines

T cell-recognized tumour antigens can be divided into two main categories. The first are known as tumour specific antigens (TSA), and the genes encoding TSA are present only in tumour cells and not in normal tissues. The second group, called tumour-associated antigens (TAA), are expressed at elevated levels in tumour cells but are also present in normal cells.

Theoretically, TSA represent the most desirable target antigens for anticancer vaccination or adoptive therapy. Their tumour cell-specific expression means there will be no pre-existing immunological self-tolerance, and immune responses directed against TSA will be unlikely to damage normal tissues. Examples of TSA include the products of mutated genes, such as the oncogene RAS and the bcr/abl fusion protein. The major limitation of targeting TSA for immunotherapy is that they are frequently invisible to CTL, due to restrictions dictated by rules governing MHC class I presentation. Presentation of peptides on the cell surface requires proteasome-aided peptide release from larger precursors, peptide transport by TAP molecules and high-affinity peptide binding to MHC class I molecules. Frequently, peptides with specific mutations will not successfully compete against the large number of peptides produced from normal cellular proteins, and hence be poorly presented on the cell surface. This may help to explain the observation that the vast majority of peptides that have been identified as CTL targets in cancer patients are products of normal cellular proteins expressed at elevated levels in tumours, i.e. TAA.

Active vaccination strategies for cancer are usually directed against defined TAA [2], or against an unidentified number of antigens in cases when the vaccine consists of whole tumour cells [3], cell lysates [4,5] or heat shock proteins [6] and RNA [7,8] isolated from tumour cells. Although vaccines based on multiple unidentified antigens have shown promise in the experimental setting [9], the monitoring of vaccine-induced immune responses is difficult without knowledge of relevant target antigens. The progressive, rational improvement of vaccines is more probable with defined antigens, as it provides an opportunity to link the nature of antigen-specific immune responses with clinical outcome. Also, there is a growing consensus that antigen-specific cancer vaccines will prove to be more effective than vaccination strategies based on unidentified antigens [1012].

Vaccination in the murine model

Prophylactic active vaccination with a range of TAA has been shown to protect from tumour challenge and prevent tumour occurrence in some animal models [1315]. Murine models have demonstrated that vaccination with melanocyte differentiation antigens can provide protection against melanoma. Bronte et al. [16] have found that vaccination with murine TRP-2 protects mice from challenge with melanoma cells, while Mullins et al. [17] have shown in HLA-A*0201-transgenic mice that vaccination with tyrosinase or gp100 can control melanoma outgrowth. Soares et al. have recently demonstrated in a MUC1 transgenic mouse model that vaccination with dendritic cells pulsed with a 140 amino acid long peptide derived from the TAA MUC1 improved survival of tumour challenged mice. Surprisingly, the level of protection in MUC-1 transgenic mice was similar to that seen in wild-type mice, suggesting that T cell tolerance did not interfere with the development of protective immunity. It is possible that the 140 amino acid long peptide stimulated unconventional, MHC-unrestricted T cells responses [18], which may explain the failure to identify MHC-restricted CTL responses in vaccinated mice [13]. Romieu et al. [19] used the SV40 large T transgenic tumour model to demonstrate that vaccination with MHC class I presented large T epitopes together with heterologous helper peptides stimulated low-avidity CTL which did not prevent the development of large T induced spontaneous tumours, whereas adoptive transfer of higher avidity CTL specific for the same epitope substantially decreased the tumour burden.

Frequently, vaccination can generate antigen-specific CTL, but their avidity is low. However, it may be possible to exploit the low avidity self-specific T cell repertoire for tumour immunotherapy as suggested by experiments in transgenic models [20]. For example, in transgenic mice expressing a CTL-recognized influenza nucleoprotein (NP) epitope in most normal tissues, an oligoclonal population of NP-specific CTL can escape thymic and peripheral deletion and can be activated subsequently by immunization. Although primary CTL were of low avidity, repeated vaccination led to the generation of memory CTL with improved avidity, and these CTL were capable of delaying the growth of tumour cells expressing the NP epitope. Thus, multiple vaccinations with a tumour-associated self-antigen may provide partial tumour protection by improving both the quality (avidity) and quantity (frequency) of antigen-specific CTL [21,22].

In contrast to their success in the prophylactic setting, TAA-based vaccines were largely unsuccessful in the therapeutic setting, suggesting that the quality and quantity of the CTL response is not protective against existing tumours. One possible explanation is that pre-existing tolerance prevents rapid activation and expansion of high avidity CTL required for the inhibition of existing tumours. This explanation is compatible with the observation that vaccination against murine TSAs that are not present in normal cells can be effective in the therapeutic setting and inhibit growth of existing tumours [2325].

Studies in the context of non-self target antigens have been performed where high avidity-CTL have been shown to provide better protection than low avidity CTL against viral infection [26]. Derby et al. demonstrated that high avidity viral-specific CTL were capable of recognizing lower antigen densities and more rapidly lysing target cells at a given antigen density than the lower-avidity CTL. Similarly, high-avidity CTL for tumour associated self-antigens have been shown to have superior antitumour efficacy both in vitro and in vivo in animal models [27].

Vaccination in humans

In the past decade antigen-specific cancer vaccines have entered phase I and II clinical trials. The majority of trials were performed in melanoma patients using TAA-based vaccine preparations. Many trials have reported detectable vaccine induced T cell responses in the absence of clinical responses [2830]. For example, in one trial, end-stage melanoma patients were vaccinated with mature monocyte derived dendritic cells pulsed with the MAGE-3A2·1 peptide, which resulted in the generation of MAGE-3 specific T cells in patients without signs of tumour reduction [30]. As discussed above, this may be due to lack of activation of high avidity CTL caused by immunological tolerance mechanisms. Other important factors determining clinical responses are level of antigen expression in the target tumour cells. Expression of a given target antigen is likely to vary between patients and at different time points in the malignant process. Some tumours have been shown to escape immunological surveillance via down-regulation or loss of expression of HLA Class I molecules [31,32].

Nevertheless, it is clear that tolerance to melanoma antigens is incomplete. For example, in a few clinical trials there have been reports of clinical responses including stabilization of disease, partial and even complete tumour regression [3340]. A statistically significant positive correlation between the percentage of tetramer positive antigen-specific CD8+ T cells and the clinical response of patients was observed by Fong et al. [36], indicating that clinical responses correlated directly with detectable immune responses. In contrast, Marchand et al. [37] found clinical responses in the absence detectable T cell responses. Together, the studies in melanoma patients indicate that more trials with well-defined vaccination and monitoring protocols are needed to establish a link between immunological response profiles and clinical outcome.

Immunological tolerance

Negative selection (deletion) of T cells reacting with intrathymic self-antigens and self-MHC is the principle mechanism by which tolerance is achieved [41]. In addition, peripheral tolerance mechanisms exist to prevent effective triggering of self-reactive T cells that have escaped thymic deletion [42]. Peripheral tolerance mechanisms include ignorance [43], anergy [44], TCR down-regulation [45], deletion [4648], or suppression of autoreactive T cells by CD4+/CD25+ regulatory T cells [49].

Purging of high-avidity CTL as a consequence of extrathymic antigen expression in a small number of cells in the peripheral tissues has been documented in murine models [5052]. However, peripheral tolerance mechanisms are leaky, allowing escape of a small number of autoreactive T cells. Using a double transgenic model, Morgan et al. demonstrated that the number of adoptively transferred autoreactive T cells determined the level of tolerance [53]. Transfer of 100 high-avidity CTL specific for a self-antigen expressed in the pancreas resulted in tolerance within 5 days, whereas tolerance induction of 10 000 high-avidity CTL required 120–220 days. This indicates that small numbers of high-avidity CTL, matching naturally occurring numbers that escape thymic deletion, are tolerized efficiently in the periphery, while large numbers of high-avidity CTL, achievable in the adoptive therapy setting, can escape tolerance and may provide tumour protection.

As TSA are neoantigens not present in normal tissue, the level of antigen expression on target tumour cells may be less important in determining the functional avidity of the antigen-specific CTL, as high-avidity CTL are not susceptible to tolerance. However, when the target antigen is an overexpressed TAA, self-restricted CTL will be susceptible to the tolerance mechanisms as described above, resulting in deletion/anergy of high-avidity CTL. Therefore, tumour cells will be required to express higher levels of antigen to trigger low-avidity CTL, which escape tolerance and normally ignore low level expression of the self-antigen in normal tissues.

Adoptive therapy

Reduced immune competence and immunological tolerance are major limitations for vaccination strategies that need to be overcome by alternative approaches. One of the major benefits of adoptive therapy is the ability to transfer antigen-specific CTL of known avidity and specificity. The transfer of antigen-specific T cells has major applications in the therapy of viral infectious diseases, virus-associated malignancies and cancer. The successful translation of adoptive immunotherapy into clinical practice has been achieved first for the management of cytomegalovirus (CMV) and Epstein–Barr virus (EBV) infection complicating haematopoietic stem cell transplantation (SCT). The adoptive transfer of CMV-specific CTL clones has reduced the incidence of CMV disease [54]. Similarly, infusions of donor-derived EBV-specific T cell lines following T cell-depleted SCT have prevented EBV-triggered lymphoproliferative disorder [55,56] and reduced the viral load in patients with high titre EBV DNA post-transplant [57]. For patients undergoing solid organ transplantation the use of EBV-specific CTL has been explored with some success [58,59].

Adoptive transfer of allogeneic T cells is currently the only immunotherapy strategy in humans with proven curative potential against cancers of non-viral aetiology. The first description of the allogeneic graft-versus-leukaemia (GVL) effect was as early as 1956, when Barnes et al. observed the erradication of leukaemia in irradiated mice receiving allogeneic, but not syngeneic bone marrow transplants [60]. GVL is now regarded as a donor T cell-mediated antileukaemic immune response. The first evidence of a similar effect in humans followed the observation that relapse rates were lower following T cell-containing allografts and in the presence of acute and chronic GVHD [6163].

Donor lymphocyte infusions (DLI) were first used to treat relapse after HLA-identical sibling SCT in the late 1980s [64]. Subsequent work has confirmed the efficacy of DLI for molecular, cytogenetic and haematological relapse of CML following stem cell transplantation [6567]. The use of DLI is now commonplace in the management of haematological malignancies following allogeneic SCT [6870]. The major disadvantage of DLI is the development of acute or chronic GVHD, which significantly increases the transplant-related mortality, reduces quality of life and increases infectious complications if long-term immunosuppression is required [71,72]. The risk of GVHD following a given DLI dose increases with the degree of HLA disparity between donor and recipient and is greater if DLI is given early post-transplant.

The target antigens involved in both GVL and GVHD have not yet been elucidated fully, and it is likely that some are shared. For example, the minor histocompatibility antigen 1 (HA1) has been implicated in GVHD [73,74], and more recently in GVL [75]. CTL directed against minor histocompatibility antigens expressed exclusively in haematopoietic tissues are currently in clinical studies to test if they can mediate GVL without GVHD. It is anticipated that adoptive CTL transfer will be beneficial in patients after SCT, as normal haematopoietic cells will be antigen-negative because they are of donor origin, whereas residual leukaemia cells will be antigen-positive and thus function as CTL targets [76,77].

The role of CD4+ CD25+ suppressor T cells in tumour immunobiology is not yet well understood, although there is increasing evidence that they inhibit tumour protective immune responses [78]. It has been shown recently that lung tumours contain large numbers of CD4+ CD25+ cells expressing high levels of CD152 (CTLA-4). These regulatory T cells have been shown to inhibit the proliferation of autologous but not allogeneic T cells [79], although the mechanism leading to the selective suppression is unclear. The implications of this study are that resident CD4+ CD25+ cells suppress tumour-infiltrating autologous T cells, and that adoptively transferred CTL might escape this suppression.

Future of antigen-specific immunotherapy

A number of novel approaches have been developed recently to circumvent the tolerance mechanisms responsible for limiting efficacy of CTL responses to self-antigens (Fig. 1). Although this had led to the isolation of high avidity CTL with killing activity against malignant cells, the expression levels in normal cells were insufficient to trigger CTL killing.

Fig. 1.

Fig. 1

Novel approaches for the generation of high-avidity CTL specific for tumour-associated antigens. Overview of four recently developed technologies to isolate high-affinity receptors specific for HLA class I presented peptide epitopes (HLA-A2 is shown because it is one of the most frequent class I alleles in humans). (a) and (b) take advantage of the TCR repertoire of healthy individuals or transgenic mice that are not tolerant to A2-presented peptide epitopes of tumour-associated antigens (TAA); (c) and (d) take advantage of in vitro mutagenesis and selection of high affinity TCRs (c) or high-affinity single chain antibodies specific for the peptide/HLA combination. (e) Strategies to transfer high avidity HLA-restricted receptors into patient T cells or stem cells to produce tumour-reactive T cells.

One approach, the allorestricted strategy, has grown from a greater understanding of the alloresponse and the observation that T cell tolerance is self-MHC-restricted [41,80]. Using this approach in a murine model, CTL have been generated against the mdm2 protein that is over-expressed in a large number of tumours [81]. Subsequently, allorestricted tumour-specific T cells were generated from normal human donors [82]. PBMCs from HLA-A2 negative donors were stimulated with APCs presenting synthetic peptides derived from cyclin-D1 in the context of HLA-A2. Cloning of bulk T cell cultures was used to isolate peptide-specific CTL lines that killed HLA-A2 positive breast cancer cells over-expressing cyclin-D1.

Similarly, HLA-A2-restricted CTL specific for the Wilm's tumour antigen-1 (WT1), over-expressed in most leukaemias, have been shown to inhibit leukaemic stem cells, while ignoring normal stem cells [83,84]. The allorestricted approach has also been used to produce CTL against the minor histocompatibility antigen HA1, for possible therapy of patients after HLA-mismatched SCT [76].

Allorestricted approaches allow isolation of high avidity antigen-specific CTL when the target antigen is an overexpressed self-antigen. There are limitations, however, which may reduce its applicability outside the context of HLA-mismatched allogeneic SCT. Allorestricted CTL generated from an HLA-mismatched donor are likely to be rejected rapidly by the recipient unless they are profoundly immunosuppressed. Therefore, the transfer of an allorestricted TCR into patient T cells provides a technology to overcome these limitations.

The feasibility of TCR transfer has been demonstrated recently using CTL specific for an mdm2 peptide presented by HLA-A2 molecules. To bypass self-tolerance to this tumour antigen the CTL were isolated from HLA-A2 transgenic mice and from A2-negative donors via the allorestricted strategy [85]. Murine HLA A2-restricted CTL were generated by immunizing the mice with a human mdm2 peptide epitope differing by a single amino acid to the naturally presented murine peptide. A high-avidity TCR was cloned from murine CTL, partially humanized and retrovirally transferred into human T cells to produce CTL capable of high-avidity killing of mdm2-expressing cells. Both avidity and antigen-specificity were reconstituted in the human T cells after retroviral transduction of TCR genes. Such therapeutic TCR transfer provides a method of rescuing the self-MHC-restricted human T cell repertoire by producing high-affinity, broad-spectrum tumour reactive TCRs that are usually deleted by tolerance mechanisms.

Alternative approaches of generating high-affinity receptors for tumour-associated peptide epitopes have been described recently. For example, in vitro mutagenesis followed by tetramer selection can be used to isolate TCRs with improved antigen-specific binding affinity [86]. This technology can be used to convert low-affinity TCRs specific for tumour-associated self-peptides into high-affinity TCRs and thus improve tumour cell recognition [87]. Phage display of single chain antibody fragments provides another technology for the isolation of antibody molecules that bind specifically to a given MHC/peptide complex, thus mimicking the binding specificity of TCRs. Recently, phage display was used to isolate a single chain antibody with high-affinity binding to a melanoma-associated MAGE-3 peptide presented by HLA-A1 class I molecules. The single chain antibody fragment was fused to the FcɛR1 gamma-chain and introduced into human T cells via retroviral gene transfer. The transduced T cells showed specific killing activity against HLA-A1+, MAGE-3+ melanoma cells, illustrating that the chimeric receptor can elicit antigen-specific effector function in human T cells [88].

In addition to receptor transfer into mature T cells, it is also possible to target the haematopoietic stem cell. It has been demonstrated recently that TCR transfer into murine haematopoietic stem cells produced mature T cells that responded to antigen-specific challenge in vivo[89]. A significant advantage of this approach is the potential to continually generate antigen-specific T cells from a self-renewing stem cell pool.

Together, several new technologies are now available to isolate high-affinity TCRs and TCR-like molecules to overcome immunological tolerance. This provides a platform to overcome one of the major limitations of autologous immune responses against tumours, namely low-avidity T cell responses against tumour-associated antigens.

Acknowledgments

The authors thank the Leukaemia Research Fund, the Medical Research Council and Cancer Research UK for support.

References

  • 1.Van der Bruggen P, Traversari C, Chomez P, et al. A gene encoding an antigen recognised by cytolytic T lymphocytes on a human melanoma. Science. 1991;254:1643–7. doi: 10.1126/science.1840703. [DOI] [PubMed] [Google Scholar]
  • 2.Disis ML, Gooley TA, Rinn K, 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: 10.1200/JCO.2002.06.171. [DOI] [PubMed] [Google Scholar]
  • 3.Dredge K, Marriott JB, Todryk SM, et al. Protective antitumor immunity induced by a costimulatory thalidomide analog in conjunction with whole tumor cell vaccination is mediated by increased Th1-type immunity. J Immunol. 2002;168:4914–9. doi: 10.4049/jimmunol.168.10.4914. [DOI] [PubMed] [Google Scholar]
  • 4.Geiger JD, Hutchinson RJ, Hohenkirk LF, et al. Vaccination of pediatric solid tumor patients with tumor lysate-pulsed dendritic cells can expand specific T cells and mediate tumor regression. Cancer Res. 2001;61:8513–9. [PubMed] [Google Scholar]
  • 5.Schnurr M, Galambos P, Scholz C, et al. Tumor cell lysate-pulsed human dendritic cells induce a T-cell response against pancreatic carcinoma cells: an in vitro model for the assessment of tumor vaccines. Cancer Res. 2001;61:6445–50. [PubMed] [Google Scholar]
  • 6.Udono H, Levey DL, Srivastava PK. Cellular requirements for tumor-specific immunity elicited by heat shock proteins: tumor rejection antigen gp96 primes CD8+ T cells in vivo. Proc Natl Acad Sci USA. 1994;91:3077–81. doi: 10.1073/pnas.91.8.3077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Ponsaerts P, Van Den Bosch G, Cools N, et al. Messenger RNA electroporation of human monocytes, followed by rapid in vitro differentiation, leads to highly stimulatory antigen-loaded mature dendritic cells. J Immunol. 2002;169:1669–75. doi: 10.4049/jimmunol.169.4.1669. [DOI] [PubMed] [Google Scholar]
  • 8.Boczkowski D, Nair SK, Nam JH, Lyerly HK, Gilboa E. Induction of tumor immunity and cytotoxic T lymphocyte responses using dendritic cells transfected with messenger RNA amplified from tumor cells. Cancer Res. 2000;60:1028–34. [PubMed] [Google Scholar]
  • 9.Hurwitz AA, Yu TFY, Leach DR, Allison JP. CTLA-4 blockade synergizes with tumor-derived granulocyte-macrophage colony-stimulating factor for treatment of an experimental mammary carcinoma. Proc Natl Acad Sci USA. 1998;95:10067–71. doi: 10.1073/pnas.95.17.10067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Offringa R, Van der Burg SH, Ossendorp F, Toes RE, Melief CJ. Design and evaluation of antigen-specific vaccination strategies against cancer. Curr Opin Immunol. 2000;12:576–82. doi: 10.1016/s0952-7915(00)00145-x. [DOI] [PubMed] [Google Scholar]
  • 11.Rosenberg S. Progress in human tumour immunology and immunotherapy. Nature. 2001;411:380–4. doi: 10.1038/35077246. [DOI] [PubMed] [Google Scholar]
  • 12.Jager E, Jager D, Knuth A. Clinical cancer vaccine trials. Curr Opin Immunol. 2002;14:178–82. doi: 10.1016/s0952-7915(02)00318-7. [DOI] [PubMed] [Google Scholar]
  • 13.Soares MM, Mehta V, Finn OJ. Three different vaccines based on the 140-amino acid MUC1 peptide with seven tandemly repeated tumor-specific epitopes elicit distinct immune effector mechanisms in wild-type versus MUC1-transgenic mice with different potential for tumor rejection. J Immunol. 2001;166:6555–63. doi: 10.4049/jimmunol.166.11.6555. [DOI] [PubMed] [Google Scholar]
  • 14.Mathiassen S, Lauemoller SL, Ruhwald M, Claesson MH, Buus S. Tumor-associated antigens identified by mRNA expression profiling induce protective anti-tumor immunity. Eur J Immunol. 2001;31:1239–46. doi: 10.1002/1521-4141(200104)31:4<1239::aid-immu1239>3.0.co;2-c. [DOI] [PubMed] [Google Scholar]
  • 15.Xiang R, Primus FJ, Ruehlmann JM, et al. A dual-function DNA vaccine encoding carcinoembryonic antigen and CD40 ligand trimer induces T cell-mediated protective immunity against colon cancer in carcinoembryonic antigen-transgenic mice. J Immunol. 2001;167:4560–5. doi: 10.4049/jimmunol.167.8.4560. [DOI] [PubMed] [Google Scholar]
  • 16.Bronte V, Apolloni E, Ronca R, et al. Genetic vaccination with ‘self’ tryosinase-related protein 2 causes melanoma eradication but not vitiligo. Cancer Res. 2000;60:253–8. [PMC free article] [PubMed] [Google Scholar]
  • 17.Mullins DW, Bullock TNJ, Collela TA, Robila VV, Engelhard VH. Immune responses to the HLA-A*0201-restricted epitopes of tyrosinase and glycoprotein 100 enable control of melanoma outgrowth in HLA-A*0201-transgenic mice. J Immunol. 2001;167:4853–60. doi: 10.4049/jimmunol.167.9.4853. [DOI] [PubMed] [Google Scholar]
  • 18.Magarian-Blander J, Ciborowski P, Hsia S, Watkins SC, Finn OJ. Intercellular and intracellular events following the MHC-unrestricted TCR recognition of a tumor-specific peptide epitope on the epithelial antigen MUC1. J Immunol. 1998;160:311–20. [PubMed] [Google Scholar]
  • 19.Romieu R, Baratin M, Kayibanda M, et al. Passive but not active CD8+ T cell-based immunotherapy interferes with liver tumor progression in a transgenic mouse model. J Immunol. 1998;161:5133–7. [PubMed] [Google Scholar]
  • 20.Cordaro TA, de Visser KE, Tirion FH, Schumacher TNM, Kruisbeek AM. Can the low-avidity self-specific T cell repertoire be exploited for tumor rejection? J Immunol. 2002;168:651–60. doi: 10.4049/jimmunol.168.2.651. [DOI] [PubMed] [Google Scholar]
  • 21.Zimmermann C, Prevost-Blondel A, Blaser C, Pircher H. Kinetics of the response of naive and memory CD8 T cells to antigen: similarities and differences. Eur J Immunol. 1999;29:284–90. doi: 10.1002/(SICI)1521-4141(199901)29:01<284::AID-IMMU284>3.0.CO;2-C. [DOI] [PubMed] [Google Scholar]
  • 22.Veiga-Fernandes H, Walter U, Bourgeois C, McLean A, Rocha B. Response of naive and memory CD8+ T cells to antigen stimulation in vivo. Nat Immunol. 2000;1:47–53. doi: 10.1038/76907. [DOI] [PubMed] [Google Scholar]
  • 23.Klein C, Bueler H, Mulligan RC. Comparative analysis of genetically modified dendritic cells and tumor cells as therapeutic cancer vaccines. J Exp Med. 2000;191:1699–708. doi: 10.1084/jem.191.10.1699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Cheng WF, Hung CF, Chai CY, et al. Tumor-specific immunity and antiangiogenesis generated by a DNA vaccine encoding calreticulin linked to a tumor antigen. J Clin Invest. 2001;108:669–78. doi: 10.1172/JCI12346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Merad M, Sugie T, Engleman EG, Fong L. In vivo manipulation of dendritic cells to induce therapeutic immunity. Blood. 2002;99:1676–82. doi: 10.1182/blood.v99.5.1676. [DOI] [PubMed] [Google Scholar]
  • 26.Derby MA, Alexander-Miller MA, Tse R, Berzofsky JA. High-avidity CTL exploit two complementary mechanisms to provide better protection against viral infection than low-avidity CTL. J Immunol. 2001;166:1690–7. doi: 10.4049/jimmunol.166.3.1690. [DOI] [PubMed] [Google Scholar]
  • 27.Zeh HJ, III, Perry-Lalley D, Dudley ME, Rosenberg SA, Yang JC. High avidity CTL for two self-antigens demonstrate superior in vitro and in vivo antitumour efficacy. J Immunol. 1999;162:989–94. [PubMed] [Google Scholar]
  • 28.Jager E, Ringhoffer M, Altmannsberger M, et al. Immunoselection in vivo: independent loss of MHC class I and melanocyte differentiation antigen expression in metastatic melanoma. Int J Cancer. 1997;71:142–7. doi: 10.1002/(sici)1097-0215(19970410)71:2<142::aid-ijc3>3.0.co;2-0. [DOI] [PubMed] [Google Scholar]
  • 29.Knutson KL, Schiffman K, Disis ML. Immunization with a HER-2/neu helper peptide vaccine generates HER-2/neu CD8 T-cell immunity in cancer patients. J Clin Invest. 2001;107:477–84. doi: 10.1172/JCI11752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Schuler-Thurner B, Dieckmann D, Keikavoussi P, et al. Mage-3 and influenza-matrix peptide-specific cytotoxic T cells are inducible in terminal stage HLA-A2: 1+ melanoma patients mature monocyte-derived dendritic cells. J Immunol. 2000;165:3492–6. doi: 10.4049/jimmunol.165.6.3492. [DOI] [PubMed] [Google Scholar]
  • 31.Momburg F, Koch S. Selective loss of β2-microglobulin mRNA in human colon carcinoma. J Exp Med. 1989;169:309–14. doi: 10.1084/jem.169.1.309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Cromme FV, Airey J, Heemels MT, et al. Loss of transporter protein, encoded by the TAP-1 gene, is highly correlated with loss of HLA expression in cervical carcinomas. J Exp Med. 1994;179:335–40. doi: 10.1084/jem.179.1.335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Rosenberg SA, Yang JC, Schwartzentruber DJ, et al. Immunologic and therapeutic evaluation of a synthetic peptide vaccine for the treatment of patients with metastatic melanoma. Nat Med. 1998;4:321–7. doi: 10.1038/nm0398-321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Nestle FO, Alijagic S, Gilliet M, et al. Vaccination of melanoma patients with peptide- or tumor lysate-pulsed dendritic cells. Nat Med. 1998;4:328–32. doi: 10.1038/nm0398-328. [DOI] [PubMed] [Google Scholar]
  • 35.Rosenberg SA, Yang JC, Schwartzentruber DJ, et al. Impact of cytokine administration on the generation of antitumor reactivity in patients with metastatic melanoma receiving a peptide vaccine. J Immunol. 1999;163:1690–5. [PMC free article] [PubMed] [Google Scholar]
  • 36.Fong L, Hou Y, Rivas A, et al. Altered peptide ligand vaccination with Flt3 ligand expanded dendritic cells for tumor immunotherapy. Proc Natl Acad Sci USA. 2001;98:8809–14. doi: 10.1073/pnas.141226398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Marchand M, van Baren N, Weynants P, et al. Tumor regressions observed in patients with metastatic melanoma treated with an antigenic peptide encoded by gene MAGE-3 and presented by HLA-A1. Int J Cancer. 1999;80:219–30. doi: 10.1002/(sici)1097-0215(19990118)80:2<219::aid-ijc10>3.0.co;2-s. [DOI] [PubMed] [Google Scholar]
  • 38.Jager E, Maeurer M, Hohn H, et al. Clonal expansion of Melan A-specific cytotoxic T lymphocytes in a melanoma patient responding to continued immunization with melanoma-associated peptides. Int J Cancer. 2000;86:538–47. doi: 10.1002/(sici)1097-0215(20000515)86:4<538::aid-ijc16>3.0.co;2-g. [DOI] [PubMed] [Google Scholar]
  • 39.Coulie PG, Karanikas V, Colau D, 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: 10.1073/pnas.161260098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Thurner B, Haendle I, Roder C, et al. Vaccination with mage-3A1 peptide-pulsed mature, monocyte-derived dendritic cells expands specific cytotoxic T cells and induces regression of some metastases in advanced stage IV melanoma. J Exp Med. 1999;190:1669–78. doi: 10.1084/jem.190.11.1669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Matzinger P, Zamoyska R, Waldmann H. Self tolerance is H-2-restricted. Nature. 1984;308:738–41. doi: 10.1038/308738a0. [DOI] [PubMed] [Google Scholar]
  • 42.Lo D, Burkly LC, Widera G, et al. Diabetes and tolerance in transgenic mice expressing class II MHC molecules in pancreatic B cells. Cell. 1988;53:159–68. doi: 10.1016/0092-8674(88)90497-7. [DOI] [PubMed] [Google Scholar]
  • 43.Ohashi PS, Oehen S, Buerki K, et al. Ablation of ‘tolerance’ and induction of diabetes by virus infection in viral antigen transgenic mice. Cell. 1991;65:305–17. doi: 10.1016/0092-8674(91)90164-t. [DOI] [PubMed] [Google Scholar]
  • 44.Jenkins MK, Schwartz RH. Antigen presentation by chemically modified splenocytes induces antigen-specific T cell unresponsiveness in vitro and in vivo. J Exp Med. 1987;165:302–19. doi: 10.1084/jem.165.2.302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Schonrich G, Kalinke U, Momburg F, et al. Down-regulation of T cell receptors on self-reactive T cells as a novel mechanism for extrathymic tolerance induction. Cell. 1991;65:293–304. doi: 10.1016/0092-8674(91)90163-s. [DOI] [PubMed] [Google Scholar]
  • 46.Watanabe-Fukunaga R, Brannan CI, Copeland NG, Jenkins NA, Nagata S. Lymphoproliferation disorder in mice explained by defects in Fas antigen that mediates apoptosis. Nature. 1992;356:314–7. doi: 10.1038/356314a0. [DOI] [PubMed] [Google Scholar]
  • 47.Refaeli Y, Van Parijs L, London CA, Tschopp J, Abbas AK. Biochemical mechanisms of IL-2 regulated FAS-mediated T cell apoptosis. Immunity. 1998;8:615–23. doi: 10.1016/s1074-7613(00)80566-x. [DOI] [PubMed] [Google Scholar]
  • 48.Singer GG, Abbas AK. The fas antigen is involved in peripheral but not thymic deletion of T lymphocytes in T cell receptor transgenic mice. Immunity. 1994;1:365–71. doi: 10.1016/1074-7613(94)90067-1. [DOI] [PubMed] [Google Scholar]
  • 49.Walker L, Abbas AK. The enemy within: keeping self reactive T cells at bay in the periphery. Nature Rev Immunol. 2002;2:11–9. doi: 10.1038/nri701. [DOI] [PubMed] [Google Scholar]
  • 50.Collela TA, Bullock TNJ, Russell LB, et al. Self-tolerance to the murine homologue of a tyrosinase-derived melanoma antigen: implications for tumor immunotherapy. J Exp Med. 2000;191:1221–31. doi: 10.1084/jem.191.7.1221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Lo D, Freedman J, Hesse S, Palmiter RD, Brinster RL, Sherman LA. Peripheral tolerance to an islet cell-specific hemagglutinin transgene affects both CD4+ and CD8+ T cells. Eur J Immunol. 1992;22:1013–22. doi: 10.1002/eji.1830220421. [DOI] [PubMed] [Google Scholar]
  • 52.Morgan DJ, Kurts C, Kreuwel HT, Holst KL, Heath WR, Sherman LA. CD8+ T cell-mediated spontaneous diabetes in neonatal mice. Proc Natl Acad Sci USA. 1999;96:3854–8. doi: 10.1073/pnas.96.7.3854. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Morgan DJ, Kreuwel HT, Sherman LA. Antigen concentration and precursor frequency determine the rate of CD8+ T cell tolerance to peripherally expressed antigens. J Immunol. 1999;163:723–7. [PubMed] [Google Scholar]
  • 54.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 cells clones. Science. 1992;257:238–41. doi: 10.1126/science.1352912. [DOI] [PubMed] [Google Scholar]
  • 55.Rooney CM, Smith CA, Ng CY, et al. Use of gene-modified virus-specific T lymphocytes to control Epstein–Barr-virus-related lymphoproliferation. Lancet. 1995;345:9–13. doi: 10.1016/s0140-6736(95)91150-2. [DOI] [PubMed] [Google Scholar]
  • 56.Rooney CM, Smith CA, Ng CY, et al. Infusion of cytotoxic T cells for the prevention and treatment of Epstein–Barr virus infection by adoptive transfer of gene-modified virus-specific T lymphocytes. Blood. 1998;92:1549–55. [PubMed] [Google Scholar]
  • 57.Gustafsson A, Levitsky V, Zou JZ, et al. Epstein–Barr virus (EBV) laod in bone marrow transplant recipients at risk to develop posttransplant lymphoproliferative disease: prophylactic infusion of EBV-specific cytotoxic T cells. Blood. 2000;95:807–14. [PubMed] [Google Scholar]
  • 58.Khanna R, Bell S, Sherritt M, et al. Activation and adoptive transfer of Epstein–Barr virus-specific cytotoxic T cells in solid organ transplant patients with posttransplant lymphoproliferative disease. Proc Natl Acad Sci USA. 1999;96:10391–6. doi: 10.1073/pnas.96.18.10391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Haque T, Haque T, Amlot PL, et al. Reconstitution of EBV-specific T cell immunity in solid organ transplant recipients. J Immunol. 1998;160:6204–9. [PubMed] [Google Scholar]
  • 60.Barnes DWH, Corp MJ, Loutit JF, Neal FE. Treatment of murine leukaemia with x-rays and homologous bone marrow. Preliminary communication. Br Med J. 1956;2:626–7. doi: 10.1136/bmj.2.4993.626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Weiden PL, Fluornoy N, Thomas ED, et al. Antileukaemic effect of graft-versus-host disease in human recipients of allogeneic marrow grafts. N Engl J Med. 1979;300:1068–73. doi: 10.1056/NEJM197905103001902. [DOI] [PubMed] [Google Scholar]
  • 62.Weiden PL, Sullivan KM, Flournoy N, Storb R, Thomas ED the Seattle Marrow Transplant Team. Antileukemic effect of chronic graft-versus-host disease. Contribution to improved survival after allogeneic marrow transplantation. N Engl J Med. 1981;304:1529–33. doi: 10.1056/NEJM198106183042507. [DOI] [PubMed] [Google Scholar]
  • 63.Maraninchi D, Gluckman E, Blaise D, et al. Impact of T cell depletion on outcome of allogeneic bone-marrow transplantation for standard risk leukaemias. Lancet. 1987;2:175–8. doi: 10.1016/s0140-6736(87)90763-x. [DOI] [PubMed] [Google Scholar]
  • 64.Kolb HJ, Mittermuller J, Clemm C, et al. Donor leukocyte transfusions for treatment of recurrent chronic myelogenous leukaemia in marrow transplant patients. Blood. 1990;76:2462–5. [PubMed] [Google Scholar]
  • 65.Porter DL, Roth MS, McGarigle C, Ferrara J, Antin JH. Induction of graft-versus-host disease as immunotherapy for relapsed chronic myeloid leukemia. N Engl J Med. 1994;330:100–6. doi: 10.1056/NEJM199401133300204. [DOI] [PubMed] [Google Scholar]
  • 66.Dazzi F, Szydlo RM, Cross NCP, et al. Durability of responses following donor lymphocyte infusions for patients who relapse after allogeneic stem cell transplantation for chronic myeloid leukemia. Blood. 2000;96:2712–6. [PubMed] [Google Scholar]
  • 67.Guglielmi C, Arcese W, Dazzi F, et al. Donor lymphocyte infusion for relapsed chronic myelogenous leukemia: prognostic relevance of the initial cell dose. Blood. 2002;100:397–405. doi: 10.1182/blood.v100.2.397. [DOI] [PubMed] [Google Scholar]
  • 68.Mackinnon S, Papadopolous EB, Carabasi MH, et al. Adoptive immunotherapy evaluating escalating doses of donor leukocytes for relapse of chronic myeloid leukemia after bone marrow transplantation: separation of graft-versus-leukemia responses from graft-versus-host disease. Blood. 1995;86:1261–8. [PubMed] [Google Scholar]
  • 69.Mackinnon S. Who may benefit from donor leucocyte infusions after allogeneic stem cell transplantation? Br J Haematol. 2000;110:12–7. doi: 10.1046/j.1365-2141.2000.02075.x. [DOI] [PubMed] [Google Scholar]
  • 70.Peggs KS, Mackinnon S. Cellular therapy: donor lymphocyte infusion. Curr Opin Hematol. 2001;8:349–54. doi: 10.1097/00062752-200111000-00006. [DOI] [PubMed] [Google Scholar]
  • 71.Ringden O, Hermans J, Labopin M, Apperley J, Gorin NC, Gratwohl A. The highest leukaemia-free survival after allogeneic bone marrow transplantation is seen in patients with grade I acute graft-versus-host disease. Acute and Chronic Leukaemia Working Parties of the European Group for Blood and Marrow Transplantation (EBMT) Leuk Lymph. 1996;24:71–9. doi: 10.3109/10428199609045715. [DOI] [PubMed] [Google Scholar]
  • 72.Zikos P, Van Lint MT, Lamparelli T, et al. Allogeneic hemopoietic stem cell transplantation for patients with high risk acute lymphoblastic leukemia: favorable impact of chronic graft-versus-host disease on survival and relapse. Haematologica. 1998;83:896–903. [PubMed] [Google Scholar]
  • 73.Goulmy E, Schipper R, Pool J, et al. Mismatches of minor histocompatibility antigens between HLA-identical donors and recipients and the development of graft-versus-host disease after bone marrow transplantation. N Engl J Med. 1996;334:281–5. doi: 10.1056/NEJM199602013340501. [DOI] [PubMed] [Google Scholar]
  • 74.Mutis T, Gillespie G, Schrama E, Falkenburg JH, Moss P, Goulmy E. Tetrameric HLA class I-minor histocompatibility antigen peptide complexes demonstrate minor histocompatibility antigen-specific cytotoxic T lymphocytes in patients with graft-versus-host disease. Nat Med. 1999;5:839–42. doi: 10.1038/10563. [DOI] [PubMed] [Google Scholar]
  • 75.Dickinson AM, Wang XN, Sviland L, et al. In situ dissection of the graft-versus-host activities of cytotoxic T cells specific for minor histocompatibility antigens. Nat Med. 2002;8:410–4. doi: 10.1038/nm0402-410. [DOI] [PubMed] [Google Scholar]
  • 76.Mutis T, Blokland E, Kester M, Schrama E, Goulmy E. Generation of minor histocompatibility antigen HA-1-specific cytotoxic T cells restricted by nonself HLA molecules: a potential strategy to treat relapsed leukemia after HLA-mismatched stem cell transplantation. Blood. 2002;100:547–52. doi: 10.1182/blood-2002-01-0024. [DOI] [PubMed] [Google Scholar]
  • 77.Warren EH, Gavin M, Greenberg PD, Riddell SR. Minor histocompatibility antigens as targets for T-cell therapy after bone marrow transplantation. Curr Opin Hematol. 1998;5:429–33. doi: 10.1097/00062752-199811000-00013. [DOI] [PubMed] [Google Scholar]
  • 78.Shimizu J, Yamazaki S, Sakaguchi S. Induction of tumor immunity by removing CD25+CD4+ T cells: a common basis between tumor immunity and autoimmunity. J Immunol. 1999;163:5211–8. [PubMed] [Google Scholar]
  • 79.Woo EY, Yeh H, Chu CS, et al. Regulatory T cells from lung cancer patients directly inhibit autologous T cell proliferation. J Immunol. 2002;168:4272–6. doi: 10.4049/jimmunol.168.9.4272. [DOI] [PubMed] [Google Scholar]
  • 80.Rammensee HG, Bevan MJ. Evidence from in vitro studies that tolerance to self antigens is MHC-restricted. Nature. 1984;308:741–4. doi: 10.1038/308741a0. [DOI] [PubMed] [Google Scholar]
  • 81.Sadovnikova E, Stauss H. Peptide-specific cytotoxic T lymphocytes restricted by nonself major histocompatibility complex class I molecules: reagents for tumor immunotherapy. Proc Natl Acad Sci USA. 1996;93:13114–8. doi: 10.1073/pnas.93.23.13114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Sadovnikova E, Jopling L, Soo KS, Stauss HJ. Generation of human tumor-reactive cytotoxic T cells against peptides presented by non-self HLA class I molecules. Eur J Immunol. 1998;28:193–200. doi: 10.1002/(SICI)1521-4141(199801)28:01<193::AID-IMMU193>3.0.CO;2-K. [DOI] [PubMed] [Google Scholar]
  • 83.Gao L, Bellantuono I, Elasser A, et al. Selective elimination of leukemic CD34+ progenitor cells by cytotoxic T lymphocytes specific for WT1. Blood. 2000;95:2198–203. [PubMed] [Google Scholar]
  • 84.Bellantuono I, Gao L, Parry S, et al. Two distinct HLA-A0201-presented epitopes of the Wilm's tumor antigen 1 can function as targets for leukemia-reactive CTL. Blood. 2002;100:36835–7. doi: 10.1182/blood.V100.10.3835. [DOI] [PubMed] [Google Scholar]
  • 85.Stanislawski T, Voss R-H, Lotz C, et al. Circumventing tolerance to a human MDM2-derived tumor antigen by TCR gene transfer. Nat Immunol. 2001;2:962–70. doi: 10.1038/ni1001-962. [DOI] [PubMed] [Google Scholar]
  • 86.Kessels HW, van Den Boom MD, Spits H, Hooijberg E, Schumacher TN. Changing T cell specificity by retroviral T cell receptor display. Proc Natl Acad Sci USA. 2000;97:14578–83. doi: 10.1073/pnas.97.26.14578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Kessells HW, de Visser KE, Kruisbeek AM, Schumacher TNM. Circumventing T-cell tolerance to tumour antigens. Biologicals. 2001;29:277–83. doi: 10.1006/biol.2001.0300. [DOI] [PubMed] [Google Scholar]
  • 88.Chames P, Willemsen RA, Rojas G, et al. TCR-like human antibodies expressed on human CTL mediate antibody affinity-dependent cytolytic activity. J Immunol. 2002;169:1110–8. doi: 10.4049/jimmunol.169.2.1110. [DOI] [PubMed] [Google Scholar]
  • 89.Yang L, Qin X-F, Baltimore D, Van Parijs L. Generation of functional antigen-specific T cells in defined genetic backgrounds by retrovirus-mediated expression of TCR cDNAs in hematopoietic precursor cells. Proc Natl Acad Sci USA. 2002;99:6204–9. doi: 10.1073/pnas.092154599. [DOI] [PMC free article] [PubMed] [Google Scholar]

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

RESOURCES