Skip to main content

T cell avidity and tumor recognition: implications and therapeutic strategies

Abstract

In the last two decades, great advances have been made studying the immune response to human tumors. The identification of protein antigens from cancer cells and better techniques for eliciting antigen specific T cell responses in vitro and in vivo have led to improved understanding of tumor recognition by T cells. Yet, much remains to be learned about the intricate details of T cell – tumor cell interactions. Though the strength of interaction between T cell and target is thought to be a key factor influencing the T cell response, investigations of T cell avidity, T cell receptor (TCR) affinity for peptide-MHC complex, and the recognition of peptide on antigen presenting targets or tumor cells reveal complex relationships. Coincident with these investigations, therapeutic strategies have been developed to enhance tumor recognition using antigens with altered peptide structures and T cells modified by the introduction of new antigen binding receptor molecules. The profound effects of these strategies on T cell – tumor interactions and the clinical implications of these effects are of interest to both scientists and clinicians. In recent years, the focus of much of our work has been the avidity and effector characteristics of tumor reactive T cells. Here we review concepts and current results in the field, and the implications of therapeutic strategies using altered antigens and altered effector T cells.

T cell – tumor antigen interactions

Antigens recognized by tumor reactive T cells

One of the key advances in the study of tumor immunology has been the identification of specific protein antigens recognized by tumor reactive T cells. Both MHC class I and MHC class II-restricted peptides have been identified from tumor-associated antigens (TAA) on a variety of human cancers. The identification of TAA has dramatically improved our ability to study the interactions between tumor reactive T cells and their targets, and has been the foundation of new clinical strategies to treat cancer patients [1–4].

TAA can be classified into five groups based on their origin, structure, and tissue expression. Several of the earliest identified TAA were melanoma-melanocyte differentiation antigens [5–7]. These antigens, such as MART-1, gp100, and tyrosinase, are expressed exclusively by cells of the melanocyte lineage. They are considered to be shared TAA because they are expressed by the vast majority of melanomas tested [5–10]. A second group of antigens called cancer/testis antigens are expressed by normal testis and a variety of human tumors including cells from melanoma, breast, bladder, colon, lung, head and neck, gastric, ovarian, neuroblastoma, and prostate cancers [11–14]. These antigens are not universally expressed by tumors of a particular histology, but instead are seen in only a small fraction of any tumor type [15–22]. A third group of antigens are derived form normal viral proteins, and are found exclusively on tumors that are induced by viral infection of human cells [11–13]. This category includes antigens such as EBNA-3 on Epstein Barr virus-induced lymphomas and the E6 and E7 proteins on human papilloma virus-induced cervical cancers [23–25]. The fourth group of antigens is characterized by aberrant expression in tumors relative to normal tissues [12, 13]. Many of these proteins have been implicated in tumorigenesis or tumor growth and progression. Antigens such as Her-2/neu and p53, each of which may be highly over-expressed by tumor cells relative to normal tissues, fall into this category [26–32]. The final group of antigens is characterized by protein structures that contain mutations in the sequence [12, 13]. These mutations alter the processing, presentation, or recognition of the epitope by the immune system. Such mutations have been described for the β-catenin and CDK4 genes, as well as others [25, 33, 34]. With the wide variety of antigens available for recognition by the immune system, it is not surprising that proteins expressed by many common tumors can be targeted by T cells.

To date, tumor reactive T cells have been identified that recognize dozens to hundreds of different peptide epitopes. Epitopes may be presented by MHC class I for CD8 T cell recognition, or by class II molecules for CD4 T cell recognition. Epitopes for TAA restricted by HLA A, B, C, and DR alleles have been identified [11–13]. Epitopes with the most clinical relevance are those that are restricted by the most common MHC molecules (HLA-A2, C7, A1, B44, A3, B7, and DR4). These epitopes can be targeted in treatments for the greatest number of patients [35].

T cell avidity and tumor cell recognition

Avidity describes the strength of interaction between a T cell and its target antigen. Avidity is usually measured via T cell activation by a target cell, and is a sum of several contributing components, such as T cell receptor (TCR) expression levels, TCR/peptide/MHC binding affinity, co-stimulatory molecule expression, and the extracellular microenvironment. Experimental evidence suggests that avidity may exert fine control over the response of an activated T cell by influencing the binding and signaling of TCR complexes on the T cell surface. Certain T cell responses are extremely sensitive to activation by antigen. It has been reported that one TCR/peptide/MHC interaction can lead to activation of a T cell as measured by Ca+2 mobilization, three interactions lead to target cell lysis, and ten interactions lead to full activation as measured by T cell proliferation [36]. However, other more commonly used methods for measuring T cell function, such as cytokine secretion or cytolysis, fail to detect T cell responses unless far more peptide is encountered on the target. These assays are commonly performed using peptide loaded antigen presenting cells (APC) as targets in co-culture with T cells. The avidity of a T cell population can be defined by the concentration of antigen required to elicit a T cell response after target loading. In these assays, a high avidity T cell requires less antigen (< 1 nM peptide loaded on an APC) for activation than a moderate (1–100 nM peptide loaded on an APC) or low (>100 nM peptide loaded on an APC) avidity T cell [37].

Many investigators have demonstrated a correlation between T cell avidity and target recognition of T cell populations that recognize virally infected targets, murine tumor models, and human cancers. The first reported relationship between T cell avidity and target cell recognition examined interactions between polyclonal T cell populations and the protein antigen gp160 on HIV infected target cells [38]. In this study, immunization with high doses of antigen led to expansion of T cells with low avidity, whereas immunization with low doses of antigen led to expansion of T cells with high avidity. In a second report, it was shown that if high avidity T cells were exposed to high levels of antigen on targets, activation induced T cell death resulted [39]. These studies illustrated that T cell avidity plays an important role in both T cell priming and T cell response to antigen. Zeh et al. subsequently examined whether T cell avidity also influenced recognition of antigens expressed by tumor cells using a murine melanoma model [4]. In this study, high avidity T cells were raised to the antigens TRP-2 or p15E by stimulating T cells with very low amounts of antigenic peptide. In adoptive therapy experiments, the resultant high avidity T cells were more effective at eliminating lung metastases from B16 melanoma than low avidity T cells. Similar results have been seen with human T cells. Dudley et al. examined the response of individual T cell clones that recognized the melanoma TAA gp100:209–217 [40]. In co-culture with targets, the peptide load required for response by individual T cell clonotypes varied by several logs. Furthermore, there was a correlation between the relative avidity of the T cell clonotypes and their ability to recognize tumor cells. Taken together, these mouse and human results suggest that the relative sensitivity of a T cell to antigen influences its ability to recognize tumors, and that high avidity T cells are required for efficient anti-tumor immunity.

Though it is intuitive that high avidity T cells would better recognize tumors than low avidity T cells, there are reports of T cell populations which do not follow these avidity rules. Many T cells have been raised for TAA recognition through stimulation of naïve lymphocytes by peptides selected according to known MHC binding motifs [41]. The 369–376 peptide from Her-2/neu has generated conflicting reports regarding the relationship between T cell avidity and tumor target recognition. Several groups have identified T cells that recognize Her-2/neu:369–376 peptide as well as Her-2/neu+ tumor cells [26, 27, 42–44]. However, others have identified high avidity T cells that recognize peptide loaded targets but not tumors. Two clinical trials of immunization with the Her-2/neu:369–376 peptide resulted in the detection of T cells reactive with peptide loaded cells but not tumor cells [45, 46]. These contrasting results suggest that the relationship between avidity and target recognition in vivo is complex, and that it is likely under the influence of other significant factors. Identifying and controlling these other factors may be vital if T cells with the genetic capacity and sufficient avidity to recognize TAA are to function as potent anti-tumor effectors.

Experimental studies of TCR affinity and T cell avidity

TCR affinity is the strength of the molecular interaction between the receptor and peptide-MHC complex. TCR affinity has been proposed by some as the single most important component of T cell avidity, which is in agreement with current models of T cell activation that are based on the stability of TCR/peptide/MHC contact. However, experimental evidence can be found both supporting and opposing this point of view. For example, several groups have reported that bright tetramer staining, and thus high affinity TCR/peptide/MHC binding, correlates with high avidity T cell-target interactions [47, 48]. On the other hand, other groups have found no correlation between tetramer binding and T cell avidity [49, 50]. In several investigations, we have evaluated the avidity and affinity of T cells and TCR based on 1) recognition of APC's loaded with low concentrations of peptide, 2) recognition of tumor targets, or 3) an ability to signal without CD8 coreceptor binding. These studies, the results of which are detailed below, have shown that T cells with identical receptors may behave with different avidities in different circumstances.

T cell clones with identical TCR's may have different relative avidity for peptide loaded APC targets. T cell clones that recognize the HLA-A2 restricted TAA epitope gp100:209–217 were isolated from patients with malignant melanoma. DNA sequence analysis of the TCR subunits was performed on the clones, and several clones with identical receptors (sister clones) were identified. Assays measuring cytokine secretion by the sister clones after stimulation with peptide loaded APC targets or melanoma tumor targets revealed different relative avidities and differing abilities to recognize various tumor lines. These observations are not confined to melanoma reactive T cells or human TCR. Sister T cell clones recognizing the Her-2/neu:369–377 peptide have been isolated with different reactivities against the same Her-2/neu expressing target cells, and studies in animal models have also found T cells sharing the same TCR that have markedly different avidities [51].

High avidity T cells may have receptors that bind peptide-MHC complex with low affinity. A gp100:209–217 reactive T cell clone (R6C12) isolated from a patient with malignant melanoma was shown to have extremely high avidity and recognize HLA-A2+ gp100 positive tumor cells [52, 53]. Despite the high avidity of the R6C12 cells, they stained poorly with gp100:209–217 tetramers, suggesting that they had low affinity receptors. Tetramer staining by these cells was enhanced using a modified gp100 peptide that more tightly bound the HLA-A2 molecule [54]. Binding of modified tetramers was easily inhibited by anti-CD8 mAb, providing further evidence that despite the high avidity of CTL clone R6C12, its TCR had relatively low affinity. We have used gene transfer studies to characterize the R6C12 TCR in more detail [55, 56]. The R6C12 receptor was cloned, and the receptor was transferred to Jurkat cells using a retroviral construct. These cells, derived from a human T cell lymphoma, do not express the CD8 coreceptor. Transduced Jurkat cells recognized peptide antigen on loaded APC targets with high avidity yet failed to recognize tumor cells, suggesting that the affinity of the receptor for peptide-MHC was insufficient for T cell signaling without coreceptor binding. Subsequently, the R6C12 TCR was transferred to peripheral blood T cells from normal donors [57]. These cultures, in contrast to transduced Jurkat cells, demonstrated the high avidity of the original R6C12 T cell clone. In sum, these data showed that the high avidity of the R6C12 T cell was not due to a high affinity TCR.

Finally, low avidity T cells may have receptors that exhibit characteristics of high affinity TCR/peptide/MHC binding. We have described a tyrosinase reactive T cell with low-moderate avidity characteristics in assays using peptide loaded APC targets, but with the high affinity TCR characteristic of CD8 independence. Of note, this T cell is also capable of recognizing tumor cell targets. A T cell clone recognizing a HLA-A2 restricted epitope from tyrosinase was isolated from the CD4+ population of a patient with malignant melanoma, and the receptor was used for TCR transfer studies like those described above. Both the original human T cell clone and transduced murine 58α-β-cells, which lack human CD8, were able to recognize HLA-A2+ tyrosinase+ tumor cells, even though greater than 100 ng/ml of peptide on targets was required to stimulate IL-2 secretion in APC co-culture assays. In direct contrast to the R6C12 TCR described above, this TCR from a low avidity T cell clone binds and signals in the absence of CD8 coreceptor. Taken together, our studies suggest to us that T cell avidity does not necessarily predict the affinity of the TCR, and that T cells are likely able to modulate their avidity independent of TCR affinity.

Other factors influencing T cell recognition of targets

If T cells have the capacity to alter their antigen responsiveness by factors independent of their antigen receptor, molecular mechanisms other than the TCR must be implicated. Investigations by others have described numerous mechanisms by which T cell function may be altered in cancer patients. Mizoguchi et al. reported that T cells from mice bearing MCA 38 colon carcinoma tumors had reduced expression of CD3ζ chain expression on their surface, and that they had reduced levels of the tyrosine kinases p56lckand p59fyn[58]. Given that CD3ζ chain, p56lckand p59fynare required for TCR-mediated signaling to occur [59], decreased expression of these molecules in tumor bearing hosts will result in impairment of T cell immunity. It was recently reported that the levels of L-arginine in the cell culture medium could regulate CD3ζ chain expression [60] and that the enzyme arginase I produced by macrophages may regulate the levels of L-arginine in cancer patients [61]. Other investigators have shown that tumor bearing mice have lower levels of the transcription factor NFκB [62]. These signaling defects have been confirmed in several mouse tumor models and in patients with colorectal carcinoma, renal cell cancer, head and neck cancers, and other malignancies [63–67]. Other metabolic pathways also appear to regulate T cell function, such as oxidative stress from hydrogen peroxide released by cells of the monocyte/macrophage lineage [68] and the level of tryptophan metabolites resulting from indoleamine 2,3-dioxygenase expression by macrophages [69, 70]. Clearly, the influence of tumors on the physiology of the host may impact the ability to mount an immune response to malignancy by myriad mechanisms.

The CD8 coreceptor and its influence on the recognition of T cell targets deserve special emphasis. The CD8 coreceptor plays a critical role in the activation of some CD8+ T cells by binding to the α3 domain of MHC class I and recruiting the kinase p56lckto the CD3 complex [71]. As discussed above, the dependence upon CD8 coreceptor function by a specific T cell clone is greatly influenced by the TCR/peptide/MHC binding characteristics of the cell. Classically, CD8 is described as a T cell membrane αβ heterodimer [72, 73]. Recently, a CD8 αα homodimer form has been described [74]. Transfection studies have shown that the CD8 αβ heterodimer has higher affinity for MHC class I and p56lckthan the CD8 αα homodimer, and that the αβ heterodimer more efficiently mediates T cell activation [74]. The ratio of CD8 αβ to CD8 αα as well as the ability for CD8 αβ to co-localize with the TCR to lipid rafts can have a profound impact on T cell avidity [51]. Future investigations will further clarify the role of coreceptor molecules in T cell tumor recognition, and may lead to new immunotherapy strategies based in part on T cell coreceptor function.

Enhancing tumor recognition with modified TAA

Enhancing the immunogenicity of TAA by enhancing MHC-peptide binding

Tumor antigen based clinical trials have led to relatively few clinical responses [75–80]. In addition, many cancer vaccine trials show little evidence of anti-tumor immunity in the peripheral blood of patients following vaccination [78, 81]. In an effort to enhance the immunogenicity of known tumor antigens, investigators have introduced modifications into the amino acid sequences of known epitopes. Amino acid substitutions at MHC anchor positions in the antigenic peptide can lead to enhanced peptide/MHC binding [82], and can enhance the immunogenicity of an otherwise weakly immunogenic peptide both in vitro and in vivo [83–86]. The melanoma epitope gp100:209-217-2M is a well-studied example of an anchor residue-substituted peptide. Substituting a methionine for the native threonine at position 2 enhances binding of this peptide to HLA-A2 9-fold. More importantly, this M substitution enhances the immunogenicity of the peptide in vitro and in vivo with the resulting T cells having the capacity to recognize tumor cells [75, 83].

Modifications of weakly immunogenic peptides at MHC anchor residues can result in other desirable effects, such as enhancing a peptide's stability in solution. The stability of the weakly immunogenic HLA-A2 restricted peptide antigen NY-ESO-1:155–163 is enhanced by an amino acid substitution at an MHC anchor residue [82]. A substitution of valine for cysteine at position 9 in the peptide not only enhances binding to HLA-A2, but also prevents disulfide bridge formation, thus eliminating dimerization of the peptide in solution [85]. Similarly, a substitution of a serine or alanine for the cysteine at position 2 of the HLA-A1 restricted tyrosinase:243–251 decreases the amount of peptide required to elicit T cell responses in vitro by two to three logs [87]. This simple approach of modifying the MHC binding residues of weakly antigenic peptides represents a powerful strategy for activating T cell populations that would otherwise be unresponsive to stimulation by the native antigen.

Enhancing the immunogenicity of TAA by altering TCR contact residues

It has been shown that immunization with xenogeneic proteins can lead to enhanced immunity to the native protein. The genes encoding the human or rodent homologs of several tumor antigens have been used to vaccinate mice [41, 88–90]. In these studies, the xenogeneic antigens routinely resulted in greater immune responses, leading to improved anti-tumor immunity. It was speculated that differences in the amino acid sequence between the xenogeneic antigen and the target antigen resulted in heteroclitic peptides (peptide analogs substituted at positions other than MHC contact residues) that were capable of inducing both effector and helper T cell responses. This hypothesis was directly tested using a peptide from the murine tumor antigen AH-1 [91]. Substituting an alanine for a valine at position 5 increased the binding to the TCR while having no impact on binding to the murine MHC I molecule. This substitution increased the ability of the AH-1 peptide to elicit CTL responses that protect mice from challenges with AH-1 expressing tumors [91]. These animal studies indicated that modifications to TCR contact residues can enhance the immunogenicity of peptide antigens.

Several investigations have also examined the response of human T cells to peptides modified at TCR contact residues [92–94]. One such study identified a heteroclitic peptide for the immunodominant HLA-A2 restricted epitope from human carcinoembryonic antigen, CEA:605–613. Substituting an aspartic acid for the asparagine at position 6, a TCR contact residue, enhances the capacity of this peptide to elicit CEA reactive T cells that can recognize CEA antigen on tumor cells [92]. Furthermore, clinical responses have been reported in colon cancer patients receiving a tumor vaccine comprised of autologous dendritic cells loaded with this heteroclitic CEA peptide [95]. Based on these promising results, other groups have evaluated modified peptides and identified heteroclitic peptides from several tumor antigens [82, 94, 96]. These modified peptides represent a promising approach for vaccinating cancer patients with otherwise weakly immunogenic antigens.

Influence of peptide modifications on the T cell repertoire

Despite the ability of modified peptides to elicit strong anti-tumor immune responses when used for vaccinating patients, these peptides have generally failed to induce effective anti-tumor immunity and tumor regression [75, 77]. Among several possible explanations for these results, one must consider whether modified peptides will optimally stimulate the TAA reactive T cell repertoire in vivo. The T cell repertoire has tremendous diversity due in part to the structure of the TCR molecule. TCR α and β chains consists of a variable (V) segment, a joining (J) segment, and a constant (C) region with the β chain also containing a diversity (D) region. Germline rearrangements occurring within the TCR α and β loci during T cell development randomly join different V-J or V-D-J regions into a single transcriptional unit. The majority of the TCR diversity is the result of the random insertion or deletion of nucleotides at the junctions between the V and J segments for the α chain, and between the V and D and the D and J segments for the β chain. It is these V-J and V-D-J junctions of the α and β chains respectively that encode the putative third complementarity determining region (CDR3), the structural feature of the TCR critical for antigen recognition [97, 98].

Though initial reports suggested that there was a limited TCR repertoire used by tumor reactive T cells [99–104], we and others have failed to find evidence of restricted TCR V gene usage [105–112]. When we performed a detailed analysis of the TCR V genes used by MART-1:27–35 and gp100:209–217 reactive T cells, we found that 19 (out of a possible 46) different TCR Vβ were used by the MART-1:27–35 reactive T cell clones [105, 108, 110, 113–116], and 16 different TCR Vβ were used by gp100:209–217 reactive T cell clones (unpublished). Further, no homology was found within the CDR3 regions of the TCR β chains of MART-1:27–35 or gp100:209–217 reactive T cell clones. These observations suggest that there is likely to be considerable TCR diversity among tumor reactive T cells.

Amino acid substitutions in peptides at the TCR contact residues can influence TCR binding and alter the TCR repertoire. This was elegantly demonstrated in a study using single TCR chain transgenic mice. Animals expressing the transgene for a single TCR subunit chain on all T cells were vaccinated with the native moth cytochrome C (MCC) peptides or peptides containing non-conservative amino acid substitutions at the TCR contact residues. MCC reactive T cell hybridomas were isolated from the T cell repertoire after vaccination. By introducing a positively charged amino acid residue into the immunizing peptide, the investigators could induce the presence of negatively charged amino acids in the non-transgenic TCR chains of reactive clones [117]. Thus, alterations in the immunizing peptide influenced the animals' T cell repertoire significantly. We have seen similar changes in the TCR repertoire of patients after vaccination with peptide antigens modified at MHC anchor residues. We found that after vaccination with a gp100:209–217 peptide containing methionine instead of a threonine at position 2, T cell clones could be isolated from patients that recognized the modified peptide but not the native peptide or tumor cells [118]. One patient was identified from whom gp100:209 specific tumor reactive T cell clones could be isolated prior to vaccination. After vaccination, none of the peptide reactive T cell clones isolated from his peripheral blood were able to recognize tumor cells. These results indicate that even changes in the antigenic peptide which do not face the TCR can impact on the TCR repertoire. Given these observations, the potential effects on the T cell repertoire must be considered when contemplating vaccine strategies using substituted peptides.

Enhancing tumor recognition by modifying T cells

Generating tumor reactive T cell populations by TCR transfer

Generating an effective anti-tumor response in vivo requires the presence of T cell precursors capable of recognizing TAA. In many cancer patients, TAA reactive precursors can not be expanded from harvested tumor tissue, lymphoid tissue, or peripheral blood samples. It is not clear whether this is due to the low frequency of T cells against self-antigens, which comprise the majority of shared TAA, or due to an inability to activate or induce proliferation of reactive cells in vitro. A potential solution for these patients is to engineer tumor reactive T cells from naïve lymphocytes using gene therapy techniques. The validity of this approach has been established in pre-clinical studies briefly described above: through the use of specially designed DNA constructs, gene modification of effector T cells in vitro has enabled investigators to re-direct the specificity of T cell populations and T cell clones toward TAA. The majority of work in this area has used single chain antibody constructs bound to intracellular T cell signaling domains, although several investigators have transferred naturally occurring two-chain TCR molecules with their associated activities.

Redirecting T cell specificity through TCR gene therapy requires the transfer of naturally occurring TCR α and β chains to alternate effectors. TCR gene therapy has potential advantages over other adoptive immunotherapy strategies, such as the relative uniformity of the therapeutic agent and the precision with which the transduced T cell population can be measured before and after treatment. The feasibility of redirecting T cell specificity by TCR gene transfer was demonstrated by Dembic et al. in 1986 [119]. With the identification of the first shared tumor antigens for human melanoma in the early 1990's [120], we set out to transfer TAA recognition to a naïve lymphocyte population using this strategy. A TCR recognizing the melanoma antigenic peptide MART-1:27–35 was chosen for initial studies, since MART-1 is expressed by most melanomas and the epitope is restricted by the predominant MHC allele expressed in the United States, HLA-A2. The unique TCR α and β chain sequences from two HLA-A2 restricted, MART-1/Melan A reactive T cell clones were identified [105]. The Jurkat cell line was co-transfected with plasmids containing the α and β chain genes, and transfected cells were cloned in limiting dilution. Expression of the introduced TCR was confirmed, and the functional capacity of transfected clones with varied levels of TCR expression was determined by co-culturing the transduced population with peptide loaded target cells. Transfected Jurkat clones secreted IL-2 in response to culture with MART-1 loaded targets but not targets loaded with an irrelevant peptide. Furthermore, the functional avidity of the transfected clones correlated with the expression level of the transferred TCR. This was the first demonstration that a TAA specific TCR could be transferred with its characteristic antigen recognition to alternate T cells. Since these studies, Jurkat cells have been used to evaluate the transfer of other TCR's, including an HLA-A1 restricted TCR specific for MAGE-3 [121, 122].

Next, we attempted to transfer the MART-1:27–35 reactive TCR to primary human T cells from peripheral blood [123, 124]. A retroviral vector, designated A7, was constructed for transducing lymphocytes with the MART-1 receptor. To facilitate incorporation of retrovirus into the target cell genome, peripheral blood lymphocytes (PBL) were stimulated to proliferate with anti-CD3 antibody and IL-2 [125]. Transduced primary T cells were able to recognize peptide loaded targets as well as HLA-A2+ melanoma cells. Clones generated from these cultures had varied effector functions in response to co-culture with target cells. Further analysis revealed that only those that expressed the CD8 coreceptor were capable of recognizing tumor cells. Clones which expressed only the CD4 coreceptor could only recognize targets loaded with an excess of exogenous peptide, suggesting that the transferred receptor was dependent upon CD8 for full receptor function. This study verified that T cells suitable for adoptive immunotherapy could be re-directed to recognize tumor cells by TCR gene transfer. TCR's specific for a number of TAA and viral antigens associated with tumor development have been successfully introduced into T cells via retroviral gene transfer. These include TCR's specific for melanoma antigens MAGE-3, gp100, tyrosinase and CAMEL, the widely expressed oncoprotein MDM2, and the Epstein-Barr Virus protein LMP2 expressed by Hodgkin's lymphoma [57, 121, 122, 126–130]. Recently, the transfer of a TCR into T cells with known specificity has been shown to result in individual cells reactive to both antigens [131, 132]. It is therefore conceivable to engineer individual T cells with the ability to recognize multiple TAA.

The two-chain approach to TCR transfer has been modified by other investigators to address inherent problems of the approach with TCR subunit expression and pairing. When full-length TCR genes are introduced into normal T cells the native TCR α and β chains may pair with the exogenous TCR β and α chains respectively. This serves to dilute the number of functionally paired TCR's on the cell surface [133, 134], and it raises the possibility that TCR's with unknown specificity could be generated, possibly leading to unexpected autoimmunity. To counter these problems, chimeric TCR genes have been generated by fusing the cytoplasmic signaling domain of CD3ζ to MAGE-1 reactive TCR α and β genes [135]. The chimeric TCR gene successfully conferred MAGE-1 reactive function to T cells following retroviral transfer. Notably, subunit genes were shown to pair exclusively to each other following retroviral transfer to T cells, preventing both dilution of functional transferred TCR and generation of TCR's with unknown specificity.

Viral vectors for TCR transfer

Several viral vectors have been investigated for human gene therapy. Adenoviruses were the first viral vectors used due to their abilities to infect both dividing and non-dividing cells and to generate very high titer viral stocks. However, adenoviral vectors lack the ability to provide long-term transgene expression and are highly immunogenic. The viral vector of choice for many gene therapy studies, particularly in haematopoietic cells, is the retrovirus. Retroviruses infect only dividing cells and incorporate into the host cell genome, resulting in long-term transgene expression. They have low immunogenicity, providing a combination of beneficial properties for their use in gene therapies. Removal of the structural genes (gag), gene encoding enzymes for nucleic acid metabolism (pol), and the envelope encoding genes (env) serves to prevent self replication of the retrovirus following infection of target cells. The transgene TCR subunits and, commonly, a gene for cell selection encoding antibiotic resistance or a cell surface marker are then inserted under the control of the LTR and internal promoters. Our laboratory now employs a vector in which segments of the LTR have been replaced with elements of the cytomegalovirus (CMV) immediate early gene promoter. This hybrid promoter allows higher transcription levels in packaging cells leading to higher retroviral titer. We use both an internal promoter and IRES to allow for transcription of TCR genes and a selectable marker [136]. Other retroviruses that have been used for transfer of genes to human cells include murine stem-cell viruses and lentiviruses. Lentiviruses are a subset of retroviruses that are more genetically complex than MMLV. Their low immunogenic properties coupled with the capability of infecting non-dividing cells have made them a candidate for use in gene therapy. Recently, several groups have demonstrated lentiviral based gene transfer to primary human T cells [137–140]. While transduction of non-dividing T cells is possible, it has been repeatedly shown that T cell activation is still necessary for high level transfer and expression of the transgene. Furthermore, while use of retroviral based gene therapy is clinically established, lentiviral based therapies are not yet approved for clinical use.

Generating tumor reactive T cell populations with chimeric antibody-receptors

Chimeric antibody receptors are another single chain alternative to TCR for redirecting T cell specificity to TAA. Chimeric immunoglobulin (cIg) receptors are composed of the heavy and light chain variable regions of an antibody fused to the transmembrane/intracellular portion of a lymphocyte signaling molecule. The most commonly used transmembrane/intracellular portions are from the Fc εRI-γ chain and the CD3-ζ chain. cIg receptors, described shortly after the development of single chain Ab molecules in the 1980's, are attractive constructs for modifying T cell specificity because their binding is not MHC-restricted, and because cIg can recognize intact surface proteins without the need for antigen processing and presentation by the target cell [141]. TCR transduced T cells, on the other hand, are more likely to demonstrate normal antigen binding and signaling behavior, which may be important for eliciting optimal CTL responses.

In 1993, Stancovski et al. reported anti Her-2/neu activity by T cell hybridomas transduced with Her-2/neu specific cIg fused to the Fc εRI-γ chain [142]. Subsequent studies by other investigators have demonstrated the efficacy of cIg receptor constructs specific for the breast cancer antigens Her3 and Her4 [143, 144]. Ovarian cancer, lung cancer, melanoma, prostate cancer, and renal cell carcinoma are among the tumors that have been targeted with cIg receptor retroviral constructs by various groups [145–149]. Several groups have targeted glycoprotein molecules such as carcinoembryonic antigen (CEA) and GA733-2 that are expressed by a majority of colorectal cancers and other tumors of gastro-intestinal origin, and their cIg transduced T cells have shown efficacy in vitro and in murine models [150–154]. A comparison of CEA-directed cIg fused to the Fc εRI-γ chain or the CD3-ζ chain found that despite similar levels of transgene expression, CD3-ζ-linked cIg were able to better control the growth of CEA-expressing tumors in murine models [155]. Recently, these constructs have been further engineered to incorporate a costimulatory signaling mechanism [156–159]. Constructs containing the heavy and light chain variable regions of an antibody, the CD28 signaling domain, and the CD3-ζ chain in series were first described by Finney et al in 1998. T cells transduced with cIg containing the CD28 signaling domain have shown enhanced ability to control the growth of CEA-expressing tumors in murine models [158, 160].

In summary, techniques for altering T cell-tumor interactions through gene transfer are being widely investigated. At the present time, several groups of investigators are addressing the methodologic and regulatory hurdles that must be overcome in preparing these agents for clinical use. The first clinical trials of TCR gene therapy have recently been initiated. If promising, the early scientific and clinical results of these studies may soon stimulate broad interest in TCR gene therapy for cancer and associated areas of investigation.

Conclusion

Despite the wealth of information that has been acquired pertaining to T cell recognition of tumors, we are left with far more questions than answers regarding ways by which the immune response might be manipulated to improve cancer treatment. Though the T cell repertoire is expansive, the repertoire of tumor reactive cells in any individual may be very limited, or may be difficult to activate and expand either in vitro or in vivo. The relationship between TCR affinity, T cell avidity, and T cell effector function is complex. This may account for the disparity between our success in stimulating antigen reactive precursor T cells through immunization and generating cells for adoptive therapy in vitro, and our inability to achieve a high rate of durable clinical responses. A universal approach to immunization against tumor antigens or adoptive immunotherapy may not be possible for any tumor type. Instead, combined therapeutic approaches or therapy optimized for the individual may be necessary. Current and future investigations of specific T cell – tumor interactions and novel therapeutics will determine whether broadly effective immune therapies are to be realized.

References

  1. Bhattacharya-Chatterjee M, Chatterjee SK, Foon KA: The anti-idiotype vaccines for immunotherapy. Curr Opin Mol Ther. 2001, 3 (1): 63-69.

    CAS  PubMed  Google Scholar 

  2. Haigh PI, Difronzo LA, Gammon G, Morton DL: Vaccine therapy for patients with melanoma. Oncology (Huntingt). 1999, 13 (11): 1561-74; discussion 1574 passim..

    CAS  Google Scholar 

  3. Nestle FO: Dendritic cell vaccination for cancer therapy. Oncogene. 2000, 19 (56): 6673-6679. 10.1038/sj.onc.1204095.

    CAS  PubMed  Google Scholar 

  4. Zeh HJIII, Perry-Lalley D, Dudley ME, Rosenberg SA, Yang JC: High avidity CTLs for two self-antigens demonstrate superior in vitro and in vivo antitumor efficacy. JImmunol. 1999, 162 (2): 989-

    CAS  Google Scholar 

  5. Bakker AB, Schreurs MW, de Boer AJ, Kawakami Y, Rosenberg SA, Adema GJ, Figdor CG: Melanocyte lineage-specific antigen gp100 is recognized by melanoma-derived tumor-infiltrating lymphocytes. JExpMed. 1994, 179 (3): 1005-

    CAS  Google Scholar 

  6. Coulie PG, Brichard V, Van Pel A, Wolfel T, Schneider J, Traversari C, Mattei S, De Plaen E, Lurquin C, Szikora JP: A new gene coding for a differentiation antigen recognized by autologous cytolytic T lymphocytes on HLA-A2 melanomas [see comments]. JExpMed. 1994, 180 (1): 35-

    CAS  Google Scholar 

  7. Kawakami Y, Eliyahu S, Delgado CH, Robbins PF, Rivoltini L, Topalian SL, Miki T, Rosenberg SA: Cloning of the gene coding for a shared human melanoma antigen recognized by autologous T cells infiltrating into tumor. Proc Natl Acad Sci U S A. 1994, 91 (9): 3515-3519.

    PubMed Central  CAS  PubMed  Google Scholar 

  8. Kawakami Y, Eliyahu S, Delgado CH, Robbins PF, Sakaguchi K, Appella E, Yannelli JR, Adema GJ, Miki T, Rosenberg SA: Identification of a human melanoma antigen recognized by tumor-infiltrating lymphocytes associated with in vivo tumor rejection. ProcNatlAcadSciUSA. 1994, 91 (14): 6458-

    CAS  Google Scholar 

  9. Robbins PF, el Gamil M, Li YF, Topalian SL, Rivoltini L, Sakaguchi K, Appella E, Kawakami Y, Rosenberg SA: Cloning of a new gene encoding an antigen recognized by melanoma-specific HLA-A24-restricted tumor-infiltrating lymphocytes. JImmunol. 1995, 154 (11): 5944-

    CAS  Google Scholar 

  10. Wolfel T, Hauer M, Klehmann E, Brichard V, Ackermann B, Knuth A, Boon T, Meyer zum Buschenfelde KH: Analysis of antigens recognized on human melanoma cells by A2- restricted cytolytic T lymphocytes (CTL). IntJCancer. 1993, 55 (2): 237-

    CAS  Google Scholar 

  11. Coulie PG: Human tumour antigens recognized by T cells: new perspectives for anti- cancer vaccines?. MolMedToday. 1997, 3 (6): 261-

    CAS  Google Scholar 

  12. Rosenberg SA: Progress in human tumour immunology and immunotherapy. Nature. 2001, 411 (6835): 380-10.1038/35077246.

    CAS  PubMed  Google Scholar 

  13. Boon T, Coulie PG, Van den EB: Tumor antigens recognized by T cells. ImmunolToday. 1997, 18 (6): 267-

    CAS  Google Scholar 

  14. Chen YT, Old LJ: Cancer-testis antigens: targets for cancer immunotherapy [comment]. Cancer J Sci Am. 1999, 5 (1): 16-10.1111/j.1349-7006.1999.tb00660.x.

    CAS  PubMed  Google Scholar 

  15. Chen YT, Scanlan MJ, Sahin U, Tureci O, Gure AO, Tsang S, Williamson B, Stockert E, Pfreundschuh M, Old LJ: A testicular antigen aberrantly expressed in human cancers detected by autologous antibody screening. ProcNatlAcadSciUSA. 1997, 94 (5): 1914-

    CAS  Google Scholar 

  16. Jager D, Stockert E, Scanlan MJ, Gure AO, Jager E, Knuth A, Old LJ, Chen YT: Cancer-testis antigens and ING1 tumor suppressor gene product are breast cancer antigens: characterization of tissue-specific ING1 transcripts and a homologue gene. Cancer Res. 1999, 59 (24): 6197-

    CAS  PubMed  Google Scholar 

  17. van der BP, Traversari C, Chomez P, Lurquin C, De Plaen E, Van den EB, Knuth A, Boon T: A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma. Science. 1991, 254 (5038): 1643-

    Google Scholar 

  18. Van den EB, Peeters O, De Backer O, Gaugler B, Lucas S, Boon T: A new family of genes coding for an antigen recognized by autologous cytolytic T lymphocytes on a human melanoma. JExpMed. 1995, 182 (3): 689-

    Google Scholar 

  19. Ikeda H, Lethe B, Lehmann F, van Baren N, Baurain JF, De Smet C, Chambost H, Vitale M, Moretta A, Boon T, Coulie PG: Characterization of an antigen that is recognized on a melanoma showing partial HLA loss by CTL expressing an NK inhibitory receptor. Immunity. 1997, 6 (2): 199-10.1016/S1074-7613(00)80426-4.

    CAS  PubMed  Google Scholar 

  20. Gaugler B, Van den EB, van der BP, Romero P, Gaforio JJ, De Plaen E, Lethe B, Brasseur F, Boon T: Human gene MAGE-3 codes for an antigen recognized on a melanoma by autologous cytolytic T lymphocytes. JExpMed. 1994, 179 (3): 921-

    CAS  Google Scholar 

  21. Van Den Eynde BJ, Gaugler B, Probst-Kepper M, Michaux L, Devuyst O, Lorge F, Weynants P, Boon T: A new antigen recognized by cytolytic T lymphocytes on a human kidney tumor results from reverse strand transcription [see comments]. JExpMed. 1999, 190 (12): 1793-

    CAS  Google Scholar 

  22. van der BP, Bastin J, Gajewski T, Coulie PG, Boel P, De Smet C, Traversari C, Townsend A, Boon T: A peptide encoded by human gene MAGE-3 and presented by HLA-A2 induces cytolytic T lymphocytes that recognize tumor cells expressing MAGE-3. EurJImmunol. 1994, 24 (12): 3038-

    Google Scholar 

  23. Wallace LE, Rickinson AB, Rowe M, Epstein MA: Epstein-Barr virus-specific cytotoxic T-cell clones restricted through a single HLA antigen. Nature. 1982, 297 (5865): 413-10.1038/297413a0.

    CAS  PubMed  Google Scholar 

  24. Burrows SR, Sculley TB, Misko IS, Schmidt C, Moss DJ: An Epstein-Barr virus-specific cytotoxic T cell epitope in EBV nuclear antigen 3 (EBNA 3). JExpMed. 1990, 171 (1): 345-

    CAS  Google Scholar 

  25. Kast WM, Brandt RM, Drijfhout JW, Melief CJ: Human leukocyte antigen-A2.1 restricted candidate cytotoxic T lymphocyte epitopes of human papillomavirus type 16 E6 and E7 proteins identified by using the processing-defective human cell line T2. JImmunother. 1993, 14 (2): 115-

    CAS  Google Scholar 

  26. Ioannides CG, Fisk B, Fan D, Biddison WE, Wharton JT, O'Brian CA: Cytotoxic T cells isolated from ovarian malignant ascites recognize a peptide derived from the HER-2/neu proto-oncogene. Cell Immunol. 1993, 151 (1): 225-10.1006/cimm.1993.1233.

    PubMed  Google Scholar 

  27. Lustgarten J, Theobald M, Labadie C, LaFace D, Peterson P, Disis ML, Cheever MA, Sherman LA: Identification of Her-2/Neu CTL epitopes using double transgenic mice expressing HLA-A2.1 and human CD.8. HumImmunol. 1997, 52 (2): 109-

    CAS  Google Scholar 

  28. Rongcun Y, Salazar-Onfray F, Charo J, Malmberg KJ, Evrin K, Maes H, Kono K, Hising C, Petersson M, Larsson O, Lan L, Appella E, Sette A, Celis E, Kiessling R: Identification of new HER2/neu-derived peptide epitopes that can elicit specific CTL against autologous and allogeneic carcinomas and melanomas. JImmunol. 1999, 163 (2): 1037-

    CAS  Google Scholar 

  29. Theobald M, Biggs J, Dittmer D, Levine AJ, Sherman LA: Targeting p53 as a general tumor antigen. ProcNatlAcadSciUSA. 1995, 92 (26): 11993-

    CAS  Google Scholar 

  30. Houbiers JG, Nijman HW, van der Burg SH, Drijfhout JW, Kenemans P, van de Velde CJ, Brand A, Momburg F, Kast WM, Melief CJ: In vitro induction of human cytotoxic T lymphocyte responses against peptides of mutant and wild-type p53. EurJImmunol. 1993, 23 (9): 2072-

    CAS  Google Scholar 

  31. Gnjatic S, Cai Z, Viguier M, Chouaib S, Guillet JG, Choppin J: Accumulation of the p53 protein allows recognition by human CTL of a wild-type p53 epitope presented by breast carcinomas and melanomas. JImmunol. 1998, 160 (1): 328-

    CAS  Google Scholar 

  32. Brossart P, Stuhler G, Flad T, Stevanovic S, Rammensee HG, Kanz L, Brugger W: Her-2/neu-derived peptides are tumor-associated antigens expressed by human renal cell and colon carcinoma lines and are recognized by in vitro induced specific cytotoxic T lymphocytes. Cancer Res. 1998, 58 (4): 732-

    CAS  PubMed  Google Scholar 

  33. Stauss HJ, Van Waes C, Fink MA, Starr B, Schreiber H: Identification of a unique tumor antigen as rejection antigen by molecular cloning and gene transfer. J Exp Med. 1986, 164 (5): 1516-1530. 10.1084/jem.164.5.1516.

    CAS  PubMed  Google Scholar 

  34. Wolfel T, Hauer M, Schneider J, Serrano M, Wolfel C, Klehmann-Hieb E, De Plaen E, Hankeln T, Meyer zum Buschenfelde KH, Beach D: A p16INK4a-insensitive CDK4 mutant targeted by cytolytic T lymphocytes in a human melanoma. Science. 1995, 269 (5228): 1281-

    CAS  PubMed  Google Scholar 

  35. Cao K, Hollenbach J, Shi X, Shi W, Chopek M, Fernandez-Vina MA: Analysis of the frequencies of HLA-A, B, and C alleles and haplotypes in the five major ethnic groups of the United States reveals high levels of diversity in these loci and contrasting distribution patterns in these populations. Hum Immunol. 2001, 62 (9): 1009-1030. 10.1016/S0198-8859(01)00298-1.

    CAS  PubMed  Google Scholar 

  36. Huppa JB, Davis MM: T-cell-antigen recognition and the immunological synapse. Nature Reviews Immunology. 2003, 3 (12): 973-10.1038/nri1245.

    CAS  PubMed  Google Scholar 

  37. Snyder JT, Alexander-Miller MA, Berzofsky JA, Belyakov IM: Molecular mechanisms and biological significance of CTL avidity. Current Hiv Research. 2003, 1 (3): 287-10.2174/1570162033485230.

    CAS  PubMed  Google Scholar 

  38. Alexander-Miller MA, Leggatt GR, Berzofsky JA: Selective expansion of high- or low-avidity cytotoxic T lymphocytes and efficacy for adoptive immunotherapy. ProcNatlAcadSciUSA. 1996, 93 (9): 4102-

    CAS  Google Scholar 

  39. Alexander-Miller MA, Leggatt GR, Sarin A, Berzofsky JA: Role of antigen, CD8, and cytotoxic T lymphocyte (CTL) avidity in high dose antigen induction of apoptosis of effector CTL. JExpMed. 1996, 184 (2): 485-

    CAS  Google Scholar 

  40. Dudley ME, Nishimura MI, Holt AK, Rosenberg SA: Antitumor immunization with a minimal peptide epitope (G9-209-2M) leads to a functionally heterogeneous CTL response. JImmunother. 1999, 22 (4): 288-

    CAS  Google Scholar 

  41. Sette A, Fikes J: Epitope-based vaccines: an update on epitope identification, vaccine design and delivery. Current Opinion in Immunology. 2003, 15 (4): 461-10.1016/S0952-7915(03)00083-9.

    CAS  PubMed  Google Scholar 

  42. Peoples GE, Smith RC, Linehan DC, Yoshino I, Goedegebuure PS, Eberlein TJ: Shared T-Cell Epitopes in Epithelial Tumors. Cellular Immunology. 1995, 164 (2): 279-10.1006/cimm.1995.1171.

    CAS  PubMed  Google Scholar 

  43. Kono K, Rongcun Y, Charo J, Ichihara F, Celis E, Sette A, Appella E, Sekikawa T, Matsumoto Y, Kiessling R: Identification of HER2/neu-derived peptide epitopes recognized by gastric cancer-specific cytotoxic T lymphocytes. IntJCancer. 1998, 78 (2): 202-

    CAS  Google Scholar 

  44. Disis ML, Smith JW, Murphy AE, Chen W, Cheever MA: In vitro generation of human cytolytic T-cells specific for peptides derived from the HER-2/neu protooncogene protein. Cancer Res. 1994, 54 (4): 1071-

    CAS  PubMed  Google Scholar 

  45. Zaks TZ, Rosenberg SA: Immunization with a peptide epitope (p369-377) from HER-2/neu leads to peptide-specific cytotoxic T lymphocytes that fail to recognize HER-2/neu+ tumors. Cancer Res. 1998, 58 (21): 4902-

    CAS  PubMed  Google Scholar 

  46. Knutson KL, Schiffman K, Cheever MA, Disis ML: Immunization of cancer patients with a HER-2/neu, HLA-A2 peptide, p369- 377, results in short-lived peptide-specific immunity. ClinCancer Res. 2002, 8 (5): 1014-

    CAS  Google Scholar 

  47. Kerry SE, Buslepp J, Cramer LA, Maile R, Hensley LL, Nielsen AI, Kavathas P, Vilen BJ, Collins EJ, Frelinger JA: Interplay between TCR affinity and necessity of coreceptor ligation: high-affinity peptide-MHC/TCR interaction overcomes lack of CD8 engagement. JImmunol. 2003, 171 (9): 4493-

    CAS  Google Scholar 

  48. Dutoit V, Rubio-Godoy V, Dietrich PY, Quiqueres AL, Schnuriger V, Rimoldi D, Lienard D, Speiser D, Guillaume P, Batard P, Cerottini JC, Romero P, Valmori D: Heterogeneous T-cell response to MAGE-A10(254-262): High avidity-specific cytolytic T lymphocytes show superior antitumor activity. Cancer Research. 2001, 61 (15): 5850-

    CAS  PubMed  Google Scholar 

  49. Derby MA, Wang J, Margulies DH, Berzofsky JA: Two intermediate-avidity cytotoxic T lymphocyte clones with a disparity between functional avidity and MHC tetramer staining. IntImmunol. 2001, 13 (6): 817-

    CAS  Google Scholar 

  50. Dutoit V, Guillaume P, Cerottini JC, Romero P, Valmori D: Dissecting TCR-MHC/peptide complex interactions with A2/peptide multimers incorporating tumor antigen peptide variants: crucial role of interaction kinetics on functional outcomes. European Journal of Immunology. 2002, 32 (11): 3285-10.1002/1521-4141(200211)32:11<3285::AID-IMMU3285>3.0.CO;2-9.

    CAS  PubMed  Google Scholar 

  51. Cawthon AG, Lu HP, Alexander-Miller MA: Peptide requirement for CTL activation reflects the sensitivity to CD3 engagement: Correlation with CD8 alpha beta versus CD8 alpha alpha expression. Journal of Immunology. 2001, 167 (5): 2577-

    CAS  Google Scholar 

  52. Dudley ME, Wunderlich J, Nishimura MI, Yu D, Yang JC, Topalian SL, Schwartzentruber DJ, Hwu P, Marincola FM, Sherry R, Leitman SF, Rosenberg SA: Adoptive transfer of cloned melanoma-reactive T lymphocytes for the treatment of patients with metastatic melanoma. J Immunother. 2001, 24 (4): 363-373. 10.1097/00002371-200107000-00012.

    CAS  PubMed  Google Scholar 

  53. Dudley ME, Ngo LT, Westwood J, Wunderlich JR, Rosenberg SA: T-cell clones from melanoma patients immunized against an anchor-modified gp100 peptide display discordant effector phenotypes. Cancer J. 2000, 6 (2): 69-

    CAS  PubMed  Google Scholar 

  54. Denkberg G, Klechevsky E, Reiter Y: Modification of a tumor-derived peptide at an HLA-A2 anchor residue can alter the conformation of the MHC-peptide complex: Probing with TCR-like recombinant antibodies. Journal of Immunology. 2002, 169 (8): 4399-

    CAS  Google Scholar 

  55. Cole DJ, Weil DP, Shilyansky J, Custer M, Kawakami Y, Rosenberg SA, Nishimura MI: Characterization of the functional specificity of a cloned T-cell receptor heterodimer recognizing the MART-1 melanoma antigen. Cancer Res. 1995, 55 (4): 748-

    CAS  PubMed  Google Scholar 

  56. Roszkowski JJ, Yu DC, McKee MD, Cole DJ, McKee MD, Nishimura MI: CD8 Independent Tumor Cell Recognition is a Property of the T Cell Receptor and not the T Cell. Submitted, JImmunol. 2002

    Google Scholar 

  57. Morgan RA, Dudley ME, Yu YYL, Zheng ZL, Robbins PF, Theoret MR, Wunderlich JR, Hughes MS, Restifo NP, Rosenberg SA: High efficiency TCR gene transfer into primary human lymphocytes affords avid recognition of melanoma tumor antigen glycoprotein 100 and does not alter the recognition of autologous melanoma antigens. Journal of Immunology. 2003, 171 (6): 3287-

    CAS  Google Scholar 

  58. Mizoguchi H, O'Shea JJ, Longo DL, Loeffler CM, McVicar DW, Ochoa AC: Alterations in signal transduction molecules in T lymphocytes from tumor-bearing mice. Science. 1992, 258 (5089): 1795-

    CAS  PubMed  Google Scholar 

  59. Boniface JJ, Davis MM: T-cell recognition of antigen. A process controlled by transient intermolecular interactions. AnnNYAcadSci. 1995, 766: 62-

    CAS  Google Scholar 

  60. Rodriguez PC, Zea AH, Culotta KS, Zabaleta J, Ochoa JB, Ochoa AC: Regulation of T cell receptor CD3 chain expression by L-arginine. Journal of Biological Chemistry. 2002, 277 (24): 21123-10.1074/jbc.M110675200.

    CAS  PubMed  Google Scholar 

  61. Rodriguez PC, Zea AH, DeSalvo J, Culotta KS, Zabaleta J, Quiceno DG, Ochoa JB, Ochoa AC: L-arginine consumption by macrophages modulates the expression of CD3 xi chain in T lymphocytes. Journal of Immunology. 2003, 171 (3): 1232-

    CAS  Google Scholar 

  62. Correa MR, Ochoa AC, Ghosh P, Mizoguchi H, Harvey L, Longo DL: Sequential development of structural and functional alterations in T cells from tumor-bearing mice. Journal of Immunology. 1997, 158 (11): 5292-

    CAS  Google Scholar 

  63. Nakagomi H, Petersson M, Magnusson I, Juhlin C, Matsuda M, Mellstedt H, Taupin JL, Vivier E, Anderson P, Kiessling R: Decreased expression of the signal-transducing zeta chains in tumor- infiltrating T-cells and NK cells of patients with colorectal carcinoma. Cancer Res. 1993, 53 (23): 5610-

    CAS  PubMed  Google Scholar 

  64. Finke JH, Zea AH, Stanley J, Longo DL, Mizoguchi H, Tubbs RR, Wiltrout RH, Oshea JJ, Kudoh S, Klein E, Bukowski RM, Ochoa AC: Loss of T-Cell Receptor Zeta-Chain and P56(Lck) in T-Cells Infiltrating Human Renal-Cell Carcinoma. Cancer Research. 1993, 53 (23): 5613-

    CAS  PubMed  Google Scholar 

  65. Fu EJ, Arca MJ, Hain JM, Krinock R, Rado J, Cameron MJ, Chang AE, Sondak VK: Tumor-induced suppression of antitumor reactivity and depression of TCR zeta expression in tumor-draining lymph node lymphocytes: Possible relationship to the Th2 pathway. Journal of Immunotherapy. 1997, 20 (2): 111-

    CAS  PubMed  Google Scholar 

  66. Noda S, Nagatanarumiya T, Kosugi A, Narumiya S, Ra C, Fujiwara H, Hamaoka T: Do Structural-Changes of T-Cell Receptor Complex Occur in Tumor-Bearing State. Japanese Journal of Cancer Research. 1995, 86 (4): 383-

    CAS  PubMed  Google Scholar 

  67. Rabinowich H, Suminami Y, Reichert TE, CrowleyNowick P, Bell M, Edwards R, Whitesrde TL: Expression of cytokine genes or proteins and signaling molecules in lymphocytes associated with human ovarian carcinoma. International Journal of Cancer. 1996, 68 (3): 276-10.1002/(SICI)1097-0215(19961104)68:3<276::AID-IJC2>3.0.CO;2-Z.

    CAS  Google Scholar 

  68. Kono K, Salazar-Onfray F, Petersson M, Hansson J, Masucci G, Wasserman K, Nakazawa T, Anderson P, Kiessling R: Hydrogen peroxide secreted by tumor-derived macrophages down-modulates signal-transducing zeta molecules and inhibits tumor-specific T cell- and natural killer cell-mediated cytotoxicity. EurJImmunol. 1996, 26 (6): 1308-

    CAS  Google Scholar 

  69. Friberg M, Jennings R, Alsarraj M, Dessureault S, Cantor A, Extermann M, Mellor AL, Munn DH, Antonia SJ: Indoleamine 2,3-dioxygenase contributes to tumor cell evasion of T cell-mediated rejection. International Journal of Cancer. 2002, 101 (2): 151-10.1002/ijc.10645.

    CAS  Google Scholar 

  70. Uyttenhove C, Pilotte L, Theate I, Stroobant V, Colau D, Parmentier N, Boon T, Van Den Eynde BJ: Evidence for a tumoral immune resistance mechanism based on tryptophan degradation by indoleamine 2,3-dioxygenase. Nature Medicine. 2003, 9 (10): 1269-10.1038/nm934.

    CAS  PubMed  Google Scholar 

  71. Salazar-Fontana LI, Bierer BE: T-lymphocyte coactivator molecules. Current Opinion in Hematology. 2001, 8 (1): 5-10.1097/00062752-200101000-00002.

    CAS  PubMed  Google Scholar 

  72. Norment AM, Littman DR: A 2Nd Subunit of Cd8 Is Expressed in Human T-Cells. Embo Journal. 1988, 7 (11): 3433-

    PubMed Central  CAS  PubMed  Google Scholar 

  73. Disanto JP, Knowles RW, Flomenberg N: The Human Lyt-3 Molecule Requires Cd8 for Cell-Surface Expression. Embo Journal. 1988, 7 (11): 3465-

    PubMed Central  CAS  PubMed  Google Scholar 

  74. Moebius U, Kober G, Griscelli AL, Hercend T, Meuer SC: Expression of Different Cd8 Isoforms on Distinct Human Lymphocyte Subpopulations. European Journal of Immunology. 1991, 21 (8): 1793-

    CAS  PubMed  Google Scholar 

  75. Rosenberg SA, Yang JC, Schwartzentruber DJ, Hwu P, Marincola FM, Topalian SL, Restifo NP, Dudley ME, Schwarz SL, Spiess PJ, Wunderlich JR, Parkhurst MR, Kawakami Y, Seipp CA, Einhorn JH, White DE: Immunologic and therapeutic evaluation of a synthetic peptide vaccine for the treatment of patients with metastatic melanoma. NatMed. 1998, 4 (3): 321-

    CAS  Google Scholar 

  76. Staib L, Birebent B, Somasundaram R, Purev E, Braumuller H, Leeser C, Kuttner N, Li W, Zhu D, Diao J, Wunner W, Speicher D, Beger HG, Song H, Herlyn D: Immunogenicity of recombinant GA733-2E antigen (CO17-1A, EGP, KS1-4, KSA, Ep-CAM) in gastro-intestinal carcinoma patients. IntJCancer. 2001, 92 (1): 79-

    CAS  Google Scholar 

  77. Cole DJ, Wilson MC, Baron PL, O'Brien P, Reed C, Tsang KY, Schlom J: Phase I study of recombinant CEA vaccinia virus vaccine with post vaccination CEA peptide challenge. HumGene Ther. 1996, 7 (11): 1381-

    CAS  Google Scholar 

  78. Eder JP, Kantoff PW, Roper K, Xu GX, Bubley GJ, Boyden J, Gritz L, Mazzara G, Oh WK, Arlen P, Tsang KY, Panicali D, Schlom J, Kufe DW: A phase I trial of a recombinant vaccinia virus expressing prostate- specific antigen in advanced prostate cancer. ClinCancer Res. 2000, 6 (5): 1632-

    CAS  Google Scholar 

  79. Khleif SN, Abrams SI, Hamilton JM, Bergmann-Leitner E, Chen A, Bastian A, Bernstein S, Chung Y, Allegra CJ, Schlom J: A phase I vaccine trial with peptides reflecting ras oncogene mutations of solid tumors. JImmunother. 1999, 22 (2): 155-

    CAS  Google Scholar 

  80. Marshall JL, Gulley JL, Arlen PM, Beetham PK, Tsang KY, Slack R, Hodge JW, Doren S, Grosenbach DW, Hwang J, Fox E, Odogwu L, Park S, Panicali D, Schlom J: Phase I study of sequential vaccinations with fowlpox-CEA(6D)-TRICOM alone and sequentially with vaccinia-CEA(6D)-TRICOM, with and without granulocyte-macrophage colony-stimulating factor, in patients with carcinoembryonic antigen-expressing carcinomas. J Clin Oncol. 2005, 23 (4): 720-731. 10.1006/jabr.2000.8623.

    CAS  PubMed  Google Scholar 

  81. Murphy GP, Tjoa BA, Simmons SJ, Ragde H, Rogers M, Elgamal A, Kenny GM, Troychak MJ, Salgaller ML, Boynton AL: Phase II prostate cancer vaccine trial: report of a study involving 37 patients with disease recurrence following primary treatment. Prostate. 1999, 39 (1): 54-10.1002/(SICI)1097-0045(19990401)39:1<54::AID-PROS9>3.0.CO;2-U.

    CAS  PubMed  Google Scholar 

  82. Loftus DJ, Squarcina P, Nielsen MB, Geisler C, Castelli C, Odum N, Appella E, Parmiani G, Rivoltini L: Peptides derived from self-proteins as partial agonists and antagonists of human CD8(+) T-cell clones reactive to melanoma/melanocyte epitope MART1(27-35). Cancer Research. 1998, 58 (11): 2433-

    CAS  PubMed  Google Scholar 

  83. Parkhurst MR, Salgaller ML, Southwood S, Robbins PF, Sette A, Rosenberg SA, Kawakami Y: Improved induction of melanoma-reactive CTL with peptides from the melanoma antigen gp100 modified at HLA-A*0201-binding residues. JImmunol. 1996, 157 (6): 2539-

    CAS  Google Scholar 

  84. Vertuani S, Sette A, Sidney J, Southwood S, Fikes J, Keogh E, Lindencrona JA, Ishioka G, Levitskaya J, Kiessling R: Improved immunogenicity of an immunodominant epitope of the Her-2/neu protooncogene by alterations of MHC contact residues. Journal of Immunology. 2004, 172 (6): 3501-

    CAS  Google Scholar 

  85. Bownds S, Tong-On P, Rosenberg SA, Parkhurst M: Induction of tumor-reactive cytotoxic T-lymphocytes using a peptide from NY-ESO-1 modified at the carboxy-terminus to enhance HLA-A2.1 binding affinity and stability in solution. JImmunother. 2001, 24 (1): 1-10.1097/00002371-200101000-00001.

    CAS  Google Scholar 

  86. Valmori D, Levy F, Miconnet I, Zajac P, Spagnoli GC, Rimoldi D, Lienard D, Cerundolo V, Cerottini JC, Romero P: Induction of potent antitumor CTL responses by recombinant vaccinia encoding a Melan-A peptide analogue. Journal of Immunology. 2000, 164 (2): 1125-

    CAS  Google Scholar 

  87. Kittlesen DJ, Thompson LW, Gulden PH, Skipper JCA, Colella TA, Shabanowitz J, Hunt DF, Engelhard VH, Slingluff CL: Human melanoma patients recognize an HLA-A1-restricted CTL epitope from tyrosinase containing two cysteine residues: Implications for tumor vaccine development (vol 160, pg 2099, 1998). Journal of Immunology. 1999, 162 (5): 3106-

    CAS  Google Scholar 

  88. Colella TA, Bullock TN, Russell LB, Mullins DW, Overwijk WW, Luckey CJ, Pierce RA, Restifo NP, Engelhard VH: Self-tolerance to the murine homologue of a tyrosinase-derived melanoma antigen: implications for tumor immunotherapy. JExpMed. 2000, 191 (7): 1221-

    CAS  Google Scholar 

  89. Disis ML, Shiota FM, Cheever MA: Human HER-2/neu protein immunization circumvents tolerance to rat neu: a vaccine strategy for 'self' tumour antigens. Immunology. 1998, 93 (2): 192-10.1046/j.1365-2567.1998.00424.x.

    PubMed Central  CAS  PubMed  Google Scholar 

  90. Fong L, Brockstedt D, Benike C, Breen JK, Strang G, Ruegg CL, Engleman EG: Dendritic cell-based xenoantigen vaccination for prostate cancer immunotherapy. J Immunol. 2001, 167 (12): 7150-7156.

    CAS  PubMed  Google Scholar 

  91. Slansky JE, Rattis FM, Boyd LF, Fahmy T, Jaffee EM, Schneck JP, Margulies DH, Pardoll DM: Enhanced antigen-specific antitumor immunity with altered peptide ligands that stabilize the MHC-peptide-TCR complex. Immunity. 2000, 13 (4): 529-10.1016/S1074-7613(00)00052-2.

    CAS  PubMed  Google Scholar 

  92. Zaremba S, Barzaga E, Zhu MZ, Soares N, Tsang KY, Schlom J: Identification of an enhancer agonist cytotoxic T lymphocyte peptide from human carcinoembryonic antigen. Cancer Research. 1997, 57 (20): 4570-

    CAS  PubMed  Google Scholar 

  93. Castilleja A, Carter D, Efferson CL, Ward NE, Kawano K, Fisk B, Kudelka AP, Gershenson DM, Murray JL, O'Brian CA, Ioannides CG: Induction of tumor-reactive CTL by C-side chain variants of the CTL epitope HER-2/neu protooncogene (369-377) selected by molecular modeling of the peptide: HLA-A2 complex. Journal of Immunology. 2002, 169 (7): 3545-

    CAS  Google Scholar 

  94. Rivoltini L, Squarcina P, Loftus DJ, Castelli C, Tarsini P, Mazzocchi A, Rini F, Viggiano V, Belli F, Parmiani G: A superagonist variant of peptide MART1/Melan A27-35 elicits anti- melanoma CD8+ T cells with enhanced functional characteristics: implication for more effective immunotherapy. Cancer Res. 1999, 59 (2): 301-

    CAS  PubMed  Google Scholar 

  95. Fong L, Hou YF, Rivas A, Benike C, Yuen A, Fisher GA, Davis MM, Engleman EG: Altered peptide ligand vaccination with Flt3 ligand expanded dendritic cells for tumor immunotherapy. Proceedings of the National Academy of Sciences of the United States of America. 2001, 98 (15): 8809-10.1073/pnas.141226398.

    PubMed Central  CAS  PubMed  Google Scholar 

  96. Tangri S, Ishioka GY, Huang XQ, Sidney J, Southwood S, Fikes S, Sette A: Structural features of peptide analogs of human histocompatibility leukocyte antigen class I epitopes that are more potent and immunogenic than wild-type peptide. Journal of Experimental Medicine. 2001, 194 (6): 833-10.1084/jem.194.6.833.

    PubMed Central  CAS  PubMed  Google Scholar 

  97. Kaye J, Vasquez NJ, Hedrick SM: Involvement of the same region of the T cell antigen receptor in thymic selection and foreign peptide recognition. JImmunol. 1992, 148 (11): 3342-

    CAS  Google Scholar 

  98. Sorger SB, Paterson Y, Fink PJ, Hedrick SM: T cell receptor junctional regions and the MHC molecule affect the recognition of antigenic peptides by T cell clones. JImmunol. 1990, 144 (3): 1127-

    CAS  Google Scholar 

  99. Sensi M, Parmiani G: Analysis of TCR usage in human tumors: a new tool for assessing tumor- specific immune responses. ImmunolToday. 1995, 16 (12): 588-

    CAS  Google Scholar 

  100. Nitta T, Oksenberg JR, Rao NA, Steinman L: Predominant expression of T cell receptor V alpha 7 in tumor- infiltrating lymphocytes of uveal melanoma. Science. 1990, 249 (4969): 672-

    CAS  PubMed  Google Scholar 

  101. Ferradini L, Mackensen A, Genevee C, Bosq J, Duvillard P, Avril MF, Hercend T: Analysis of T-Cell Receptor Variability in Tumor-Infiltrating Lymphocytes from A Human Regressive Melanoma - Evidence for Insitu T-Cell Clonal Expansion. Journal of Clinical Investigation. 1993, 91 (3): 1183-

    PubMed Central  CAS  PubMed  Google Scholar 

  102. Straten PT, Scholler J, Houjensen K, Zeuthen J: Preferential Usage of T-Cell Receptor-Alpha-Beta Variable Regions Among Tumor-Infiltrating Lymphocytes in Primary Human-Malignant Melanomas. International Journal of Cancer. 1994, 56 (1): 78-

    Google Scholar 

  103. Weidmann E, Logan TF, Yasumura S, Kirkwood JM, Trucco M, Whiteside TL: Evidence for Oligoclonal T-Cell Response in A Metastasis of Renal-Cell Carcinoma Responding to Vaccination with Autologous Tumor-Cells and Transfer of In-Vitro-Sensitized Vaccine-Draining Lymph-Node Lymphocytes. Cancer Research. 1993, 53 (20): 4745-

    CAS  PubMed  Google Scholar 

  104. Bennett WT, Pandolfi F, Grove BH, Hawes GE, Boyle LA, Kradin RL, Kurnick JT: Dominant Rearrangements Among Human Tumor-Infiltrating Lymphocytes - Analysis of T-Cells Derived from 32 Patients with Melanoma, Lung, and Renal-Cell Carcinoma. Cancer. 1992, 69 (9): 2379-

    CAS  PubMed  Google Scholar 

  105. Cole DJ, Weil DP, Shamamian P, Rivoltini L, Kawakami Y, Topalian S, Jennings C, Eliyahu S, Rosenberg SA, Nishimura MI: Identification of MART-1-specific T-cell receptors: T cells utilizing distinct T-cell receptor variable and joining regions recognize the same tumor epitope [published erratum appears in Cancer Res 1994 Nov 15;54(22):6014]. Cancer Res. 1994, 54 (20): 5265-

    CAS  PubMed  Google Scholar 

  106. Nishimura MI, Custer MC, Schwarz SL, Parker LL, Mixon A, Clay TM, Yannelli JR, Rosenberg SA: T cell-receptor V gene use by CD4+ melanoma-reactive clonal and oligoclonal T-cell lines. J Immunother. 1998, 21 (5): 352-362.

    CAS  PubMed  Google Scholar 

  107. Nishimura MI, Kawakami Y, Charmley P, O'Neil B, Shilyansky J, Yannelli JR, Rosenberg SA, Hood L: T-cell receptor repertoire in tumor-infiltrating lymphocytes. Analysis of melanoma-specific long-term lines. JImmunotherEmphasisTumor Immunol. 1994, 16 (2): 85-

    CAS  Google Scholar 

  108. Shilyansky J, Nishimura MI, Yannelli JR, Kawakami Y, Jacknin LS, Charmley P, Rosenberg SA: T-cell receptor usage by melanoma-specific clonal and highly oligoclonal tumor-infiltrating lymphocyte lines. ProcNatlAcadSciUSA. 1994, 91 (7): 2829-

    CAS  Google Scholar 

  109. Romero P, Pannetier C, Herman J, Jongeneel CV, Cerottini JC, Coulie PG: Multiple specificities in the repertoire of a melanoma patient's cytolytic T lymphocytes directed against tumor antigen MAGE-1.A1. JExpMed. 1995, 182 (4): 1019-

    CAS  Google Scholar 

  110. Dietrich PY, Le Gal FA, Dutoit V, Pittet MJ, Trautman L, Zippelius A, Cognet I, Widmer V, Walker PR, Michielin O, Guillaume P, Connerotte T, Jotereau F, Coulie PG, Romero P, Cerottini JC, Bonneville M, Valmori D: Prevalent role of TCR alpha-chain in the selection of the preimmune repertoire specific for a human tumor-associated self-antigen. Journal of Immunology. 2003, 170 (10): 5103-

    CAS  Google Scholar 

  111. Ferradini L, Roman-Roman S, Azocar J, Avril MF, Viel S, Triebel F, Hercend T: Analysis of T-cell receptor alpha/beta variability in lymphocytes infiltrating a melanoma metastasis. Cancer Res. 1992, 52 (17): 4649-

    CAS  PubMed  Google Scholar 

  112. Albertini MR, Nicklas JA, Chastenay BF, Hunter TC, Albertini RJ, Clark SS, Hank JA, Sondel PM: Analysis of T-Cell Receptor Beta- and Gamma-Genes from Peripheral-Blood, Regional Lymph-Node and Tumor-Infiltrating Lymphocyte Clones from Melanoma Patients. Cancer Immunology Immunotherapy. 1991, 32 (5): 325-10.1007/BF01789051.

    CAS  PubMed  Google Scholar 

  113. Dufour E, Carcelain G, Gaudin C, Flament C, Avril MF, Faure F: Diversity of the cytotoxic melanoma-specific immune response: some CTL clones recognize autologous fresh tumor cells and not tumor cell lines. JImmunol. 1997, 158 (8): 3787-

    CAS  Google Scholar 

  114. Maeurer MJ, Martin DM, Storkus WJ, Lotze MT: Tcr Usage in Ctls Recognizing Melanoma Melanocyte Antigens. Immunology Today. 1995, 16 (12): 603-10.1016/0167-5699(95)80084-0.

    CAS  PubMed  Google Scholar 

  115. Sensi M, Salvi S, Castelli C, Maccalli C, Mazzocchi A, Mortarini R, Nicolini G, Herlyn M, Parmiani G, Anichini A: T cell receptor (TCR) structure of autologous melanoma-reactive cytotoxic T lymphocyte (CTL) clones: tumor-infiltrating lymphocytes overexpress in vivo the TCR beta chain sequence used by an HLA-A2- restricted and melanocyte-lineage-specific CTL clone. JExpMed. 1993, 178 (4): 1231-

    CAS  Google Scholar 

  116. Sensi M, Traversari C, Radrizzani M, Salvi S, Maccalli C, Mortarini R, Rivoltini L, Farina C, Nicolini G, Wolfel T: Cytotoxic T-lymphocyte clones from different patients display limited T- cell-receptor variable-region gene usage in HLA-A2-restricted recognition of the melanoma antigen Melan-A/MART-1. ProcNatlAcadSciUSA. 1995, 92 (12): 5674-

    CAS  Google Scholar 

  117. Jorgensen JL, Esser U, Destgroth BF, Reay PA, Davis MM: Mapping T-Cell Receptor Peptide Contacts by Variant Peptide Immunization of Single-Chain Transgenics. Nature. 1992, 355 (6357): 224-10.1038/355224a0.

    CAS  PubMed  Google Scholar 

  118. Clay TM, Custer MC, McKee MD, Parkhurst M, Robbins PF, Kerstann K, Wunderlich J, Rosenberg SA, Nishimura MI: Changes in the fine specificity of gp100(209-217)-reactive T cells in patients following vaccination with a peptide modified at an HLA-A2.1 anchor residue. JImmunol. 1999, 162 (3): 1749-

    CAS  Google Scholar 

  119. Dembic Z, Haas W, Weiss S, McCubrey J, Kiefer H, von Boehmer H, Steinmetz M: Transfer of specificity by murine alpha and beta T-cell receptor genes. Nature. 1986, 320 (6059): 232-238. 10.1038/320232a0.

    CAS  PubMed  Google Scholar 

  120. Kawakami Y, Eliyahu S, Sakaguchi K, Robbins PF, Rivoltini L, Yannelli JR, Appella E, Rosenberg SA: Identification of the immunodominant peptides of the MART-1 human melanoma antigen recognized by the majority of HLA-A2-restricted tumor infiltrating lymphocytes. JExpMed. 1994, 180 (1): 347-

    CAS  Google Scholar 

  121. Aarnoudse CA, Kruse M, Konopitzky R, Brouwenstijn N, Schrier PI: TCR reconstitution in Jurkat reporter cells facilitates the identification of novel tumor antigens by cDNA expression cloning. IntJCancer. 2002, 99 (1): 7-

    CAS  Google Scholar 

  122. Calogero A, Hospers GA, Kruse KM, Schrier PI, Mulder NH, Hooijberg E, de Leij LF: Retargeting of a T cell line by anti MAGE-3/HLA-A2 alpha beta TCR gene transfer. Anticancer Res. 2000, 20 (3A): 1793-1799.

    CAS  PubMed  Google Scholar 

  123. Clay TM, Custer MC, Sachs J, Hwu P, Rosenberg SA, Nishimura MI: Efficient transfer of a tumor antigen-reactive TCR to human peripheral blood lymphocytes confers anti-tumor reactivity. J Immunol. 1999, 163 (1): 507-513.

    CAS  PubMed  Google Scholar 

  124. Clay TM, Custer MC, Spiess PJ, Nishimura MI: Potential use of T cell receptor genes to modify hematopoietic stem cells for the gene therapy of cancer. Pathol Oncol Res. 1999, 5 (1): 3-15. 10.1053/paor.1999.0003.

    CAS  PubMed  Google Scholar 

  125. Miller DG, Adam MA, Miller AD: Gene transfer by retrovirus vectors occurs only in cells that are actively replicating at the time of infection. MolCell Biol. 1990, 10 (8): 4239-

    CAS  Google Scholar 

  126. Schaft N, Willemsen RA, de Vries J, Lankiewicz B, Essers BW, Gratama JW, Figdor CG, Bolhuis RL, Debets R, Adema GJ: Peptide fine specificity of anti-glycoprotein 100 CTL is preserved following transfer of engineered TCR alpha beta genes into primary human T lymphocytes. JImmunol. 2003, 170 (4): 2186-

    CAS  Google Scholar 

  127. Orentas RJ, Roskopf SJ, Nolan GP, Nishimura MI: Retroviral transduction of a T cell receptor specific for an Epstein-Barr virus-encoded peptide. ClinImmunol. 2001, 98 (2): 220-

    CAS  Google Scholar 

  128. Roszkowski JJ, Eiben GE, Kast WM, Yee C, Van Besien K, Nishimura MI: Simultaneous generation of CD8+ and CD4+ melanoma reactive T cells by retroviral mediated transfer of a single T cell receptor. 2005, 65(4):

    Google Scholar 

  129. Roszkowski JJ, Yu DC, Rubinstein MP, McKee MD, Cole DJ, Nishimura MI: CD8-independent tumor cell recognition is a property of the T cell receptor and not the T cell. J Immunol. 2003, 170 (5): 2582-2589.

    CAS  PubMed  Google Scholar 

  130. Stanislawski T, Voss RH, Lotz C, Sadovnikova E, Willemsen RA, Kuball J, Ruppert T, Bolhuis RL, Melief CJ, Huber C, Stauss HJ, Theobald M: Circumventing tolerance to a human MDM2-derived tumor antigen by TCR gene transfer. Nat Immunol. 2001, 2 (10): 962-970. 10.1038/ni1001-962.

    CAS  PubMed  Google Scholar 

  131. Heemskerk MH, Hoogeboom M, Hagedoorn R, Kester MG, Willemze R, Falkenburg JH: Reprogramming of virus-specific T cells into leukemia-reactive T cells using T cell receptor gene transfer. JExpMed. 2004, 199 (7): 885-

    CAS  Google Scholar 

  132. Langerman A, Callender GG, Nishimura MI: Retroviral transduction of peptide stimulated T cells can generate dual T cell receptor-expressing (bifunctional) T cells reactive with two defined antigens. JTranslMed. 2004, 2 (1): 42-

    Google Scholar 

  133. Blichfeldt E, Munthe LA, Rotnes JS, Bogen B: Dual T cell receptor T cells have a decreased sensitivity to physiological ligands due to reduced density of each T cell receptor. EurJImmunol. 1996, 26 (12): 2876-

    CAS  Google Scholar 

  134. Munthe LA, Blichfeldt E, Sollien A, Dembic Z, Bogen B: T cells with two Tcrbeta chains and reactivity to both MHC/idiotypic peptide and superantigen. Cell Immunol. 1996, 170 (2): 283-10.1006/cimm.1996.0162.

    PubMed  Google Scholar 

  135. Willemsen RA, Weijtens ME, Ronteltap C, Eshhar Z, Gratama JW, Chames P, Bolhuis RL: Grafting primary human T lymphocytes with cancer-specific chimeric single chain and two chain TCR. Gene Ther. 2000, 7 (16): 1369-10.1038/sj.gt.3301253.

    CAS  PubMed  Google Scholar 

  136. Vagner S, Galy B, Pyronnet S: Irresistible IRES. Attracting the translation machinery to internal ribosome entry sites. EMBO Rep. 2001, 2 (10): 893-10.1093/embo-reports/kve208.

    PubMed Central  CAS  PubMed  Google Scholar 

  137. Cavalieri S, Cazzaniga S, Geuna M, Magnani Z, Bordignon C, Naldini L, Bonini C: Human T lymphocytes transduced by lentiviral vectors in the absence of TCR activation maintain an intact immune competence. Blood. 2003, 102 (2): 497-505. 10.1182/blood-2003-01-0297.

    CAS  PubMed  Google Scholar 

  138. Gyobu H, Tsuji T, Suzuki Y, Ohkuri T, Chamoto K, Kuroki M, Miyoshi H, Kawarada Y, Katoh H, Takeshima T, Nishimura T: Generation and targeting of human tumor-specific Tc1 and Th1 cells transduced with a lentivirus containing a chimeric immunoglobulin T-cell receptor. Cancer Res. 2004, 64 (4): 1490-10.1158/0008-5472.CAN-03-2780.

    CAS  PubMed  Google Scholar 

  139. Maurice M, Verhoeyen E, Salmon P, Trono D, Russell SJ, Cosset FL: Efficient gene transfer into human primary blood lymphocytes by surface-engineered lentiviral vectors that display a T cell-activating polypeptide. Blood. 2002, 99 (7): 2342-10.1182/blood.V99.7.2342.

    CAS  PubMed  Google Scholar 

  140. Zhou X, Cui Y, Huang X, Yu Z, Thomas AM, Ye Z, Pardoll DM, Jaffee EM, Cheng L: Lentivirus-mediated gene transfer and expression in established human tumor antigen-specific cytotoxic T cells and primary unstimulated T cells. HumGene Ther. 2003, 14 (11): 1089-10.1089/104303403322124800.

    CAS  Google Scholar 

  141. Gross G, Waks T, Eshhar Z: Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc Natl Acad Sci U S A. 1989, 86 (24): 10024-10028.

    PubMed Central  CAS  PubMed  Google Scholar 

  142. Stancovski I, Schindler DG, Waks T, Yarden Y, Sela M, Eshhar Z: Targeting of T lymphocytes to Neu/HER2-expressing cells using chimeric single chain Fv receptors. J Immunol. 1993, 151 (11): 6577-6582.

    CAS  PubMed  Google Scholar 

  143. Muniappan A, Banapour B, Lebkowski J, Talib S: Ligand-mediated cytolysis of tumor cells: use of heregulin-zeta chimeras to redirect cytotoxic T lymphocytes. Cancer Gene Ther. 2000, 7 (1): 128-134. 10.1038/sj.cgt.7700100.

    CAS  PubMed  Google Scholar 

  144. Altenschmidt U, Klundt E, Groner B: Adoptive transfer of in vitro-targeted, activated T lymphocytes results in total tumor regression. J Immunol. 1997, 159 (11): 5509-5515.

    CAS  PubMed  Google Scholar 

  145. Hwu P, Yang JC, Cowherd R, Treisman J, Shafer GE, Eshhar Z, Rosenberg SA: In vivo antitumor activity of T cells redirected with chimeric antibody/T-cell receptor genes. Cancer Res. 1995, 55 (15): 3369-3373.

    CAS  PubMed  Google Scholar 

  146. Mezzanzanica D, Canevari S, Mazzoni A, Figini M, Colnaghi MI, Waks T, Schindler DG, Eshhar Z: Transfer of chimeric receptor gene made of variable regions of tumor-specific antibody confers anticarbohydrate specificity on T cells. Cancer Gene Ther. 1998, 5 (6): 401-407.

    CAS  PubMed  Google Scholar 

  147. Parker LL, Do MT, Westwood JA, Wunderlich JR, Dudley ME, Rosenberg SA, Hwu P: Expansion and characterization of T cells transduced with a chimeric receptor against ovarian cancer. Hum Gene Ther. 2000, 11 (17): 2377-2387. 10.1089/104303400750038480.

    CAS  PubMed  Google Scholar 

  148. Weijtens ME, Willemsen RA, Valerio D, Stam K, Bolhuis RL: Single chain Ig/gamma gene-redirected human T lymphocytes produce cytokines, specifically lyse tumor cells, and recycle lytic capacity. J Immunol. 1996, 157 (2): 836-843.

    CAS  PubMed  Google Scholar 

  149. Yun CO, Nolan KF, Beecham EJ, Reisfeld RA, Junghans RP: Targeting of T lymphocytes to melanoma cells through chimeric anti-GD3 immunoglobulin T-cell receptors. Neoplasia. 2000, 2 (5): 449-459. 10.1038/sj.neo.7900108.

    PubMed Central  CAS  PubMed  Google Scholar 

  150. Paul S, Bizouarne N, Dott K, Ruet L, Dufour P, Acres RB, Kieny MP: Redirected cellular cytotoxicity by infection of effector cells with a recombinant vaccinia virus encoding a tumor-specific monoclonal antibody. Cancer Gene Ther. 2000, 7 (4): 615-623. 10.1038/sj.cgt.7700161.

    CAS  PubMed  Google Scholar 

  151. Daly T, Royal RE, Kershaw MH, Treisman J, Wang G, Li W, Herlyn D, Eshhar Z, Hwu P: Recognition of human colon cancer by T cells transduced with a chimeric receptor gene. Cancer Gene Ther. 2000, 7 (2): 284-291. 10.1038/sj.cgt.7700121.

    CAS  PubMed  Google Scholar 

  152. Darcy PK, Kershaw MH, Trapani JA, Smyth MJ: Expression in cytotoxic T lymphocytes of a single-chain anti-carcinoembryonic antigen antibody. Redirected Fas ligand-mediated lysis of colon carcinoma. Eur J Immunol. 1998, 28 (5): 1663-1672. 10.1002/(SICI)1521-4141(199805)28:05<1663::AID-IMMU1663>3.0.CO;2-L.

    CAS  PubMed  Google Scholar 

  153. Nolan KF, Yun CO, Akamatsu Y, Murphy JC, Leung SO, Beecham EJ, Junghans RP: Bypassing immunization: optimized design of "designer T cells" against carcinoembryonic antigen (CEA)-expressing tumors, and lack of suppression by soluble CEA. Clin Cancer Res. 1999, 5 (12): 3928-3941.

    CAS  PubMed  Google Scholar 

  154. Schirrmann T, Pecher G: Human natural killer cell line modified with a chimeric immunoglobulin T-cell receptor gene leads to tumor growth inhibition in vivo. Cancer Gene Ther. 2002, 9 (4): 390-398. 10.1038/sj.cgt.7700453.

    CAS  PubMed  Google Scholar 

  155. Haynes NM, Snook MB, Trapani JA, Cerruti L, Jane SM, Smyth MJ, Darcy PK: Redirecting mouse CTL against colon carcinoma: superior signaling efficacy of single-chain variable domain chimeras containing TCR-zeta vs Fc epsilon RI-gamma. J Immunol. 2001, 166 (1): 182-187.

    CAS  PubMed  Google Scholar 

  156. Maher J, Brentjens RJ, Gunset G, Riviere I, Sadelain M: Human T-lymphocyte cytotoxicity and proliferation directed by a single chimeric TCRzeta /CD28 receptor. Nat Biotechnol. 2002, 20 (1): 70-75. 10.1038/nbt0102-70.

    CAS  PubMed  Google Scholar 

  157. Haynes NM, Trapani JA, Teng MW, Jackson JT, Cerruti L, Jane SM, Kershaw MH, Smyth MJ, Darcy PK: Single-chain antigen recognition receptors that costimulate potent rejection of established experimental tumors. Blood. 2002, 100 (9): 3155-3163. 10.1182/blood-2002-04-1041.

    CAS  PubMed  Google Scholar 

  158. Finney HM, Lawson AD, Bebbington CR, Weir AN: Chimeric receptors providing both primary and costimulatory signaling in T cells from a single gene product. J Immunol. 1998, 161 (6): 2791-2797.

    CAS  PubMed  Google Scholar 

  159. Hombach A, Wieczarkowiecz A, Marquardt T, Heuser C, Usai L, Pohl C, Seliger B, Abken H: Tumor-specific T cell activation by recombinant immunoreceptors: CD3 zeta signaling and CD28 costimulation are simultaneously required for efficient IL-2 secretion and can be integrated into one combined CD28/CD3 zeta signaling receptor molecule. J Immunol. 2001, 167 (11): 6123-6131.

    CAS  PubMed  Google Scholar 

  160. Haynes NM, Trapani JA, Teng MW, Jackson JT, Cerruti L, Jane SM, Kershaw MH, Smyth MJ, Darcy PK: Rejection of syngeneic colon carcinoma by CTLs expressing single-chain antibody receptors codelivering CD28 costimulation. J Immunol. 2002, 169 (10): 5780-5786.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Funding for the studies and authors were supplied by NIH grant CA096775 (MDM) and by NIH grants CA090873 and CA102280 (MIN). Thanks to Sandra Cleek for helping with preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Mark D McKee.

Additional information

Competing interests

The author(s) declare that they have no competing interests.

Authors' contributions

MDM conceived the design and organization for the review, participated in the immunological research included, and drafted the manuscript. JJR performed the immunological research included and participated in drafting the manuscript. MIN conceived, designed, and coordinated the immunological research included and participated in drafting the manuscript. All authors read and approved the final manuscript.

Rights and permissions

Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://creativecommons.org/licenses/by/2.0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

McKee, M.D., Roszkowski, J.J. & Nishimura, M.I. T cell avidity and tumor recognition: implications and therapeutic strategies. J Transl Med 3, 35 (2005). https://doi.org/10.1186/1479-5876-3-35

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1479-5876-3-35

Keywords