Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Nov 12.
Published in final edited form as: Curr Opin Organ Transplant. 2010 Aug;15(4):405–410. doi: 10.1097/MOT.0b013e32833b7916

Overcoming the memory barrier in tolerance induction: Molecular mimicry and functional heterogeneity among pathogen-specific T cell populations

Mandy L Ford 1, Christian P Larsen 1
PMCID: PMC4642449  NIHMSID: NIHMS734361  PMID: 20616729

Abstract

Purpose of Review

This review highlights recent advances in our understanding of the frequency and nature of alloreactivity among memory T cell populations, and discusses recent successes in experimentally targeting these populations in order to prolong graft survival.

Recent Findings

Recent studies suggest that not only is alloreactivity present within peripheral T cell compartments of normal healthy individuals, but cross-reactivity between viral-specific T cells and alloeptiopes may in fact be a very common occurrence. Furthermore, this cross-reactivity functions at the level of molecular mimicry of TCR recognition. Therapeutics that specifically target cell surface molecules or effector pathways used by memory T cells to mediate graft rejection will likely be required in order to attenuate the donor-reactive memory T cell response during transplantation.

Summary

A major challenge facing the field over the next decade is to define the heterogeneity that exists within memory T cell populations that impacts graft survival. Understanding the functional and phenotypic differences that modify the memory T cell barrier to tolerance induction might allow a strategy in which strength of immunosuppression could be tailored to fit the immunological history of a given transplant recipient in order to minimize non-immune toxicities, maximize protective immunity, and prolong graft survival.

Keywords: Transplant, costimulation, memory T cell, molecular mimicry

Introduction

T cell memory is a hallmark of adaptive immunity and plays a critical role in providing protective immunity to transplant recipients, who may lose the ability to mount de novo cellular immune responses while on immunosuppression. However, many studies over the last decades revealed the double-edged sword of memory T cells; that is, while providing protective immunity against the panoply of pathogens encountered on a daily basis, these cells may also pose a potent barrier to the attenuation of donor-specific immune responses and the induction of tolerance or a tolerant-like state. Herein, we discuss recent advances in our understanding of the frequency and nature of alloreactivity among memory T cell populations, and advances in therapeutically targeting these populations in the hopes of prolonging graft survival following transplantation.

Evidence for the existence of alloreactive T cells among memory

Perhaps the first and most fundamental question when contemplating the potential barrier posed by alloreactive memory T cells is to consider the evidence that alloreactivity exists among memory T cells. The issue of whether alloreactive T cell precursor frequencies are higher, lower, or equivalent among memory T cell populations as compared to naive T cells has been debated for at least two decades [1,2]. Early experiments addressed the alloreactivity of cord blood-derived T cell populations, which would putatively contain little to no memory T cells [2]. During the last year however, this issue has been largely put to rest, mainly with the publication of a study by Lakkis and Metes, which analyzed the frequencies of alloreactive T cells in naive (CD45ROCD62L+), central memory (CD45RO+ CD62L+), effector memory (CD45RO+ CD62L), and terminal effector memory (CD4RO CD62L) compartments, and found roughly equal frequencies of alloreactivity among all four compartments [3]. Importantly, however, differences in the ways in which alloreactivity manifested in these different T cell populations were noted. For example, in the CD8+ T cell compartment, naive T cells exhibited increased proliferation in response to allostimulation as compared to TEM, while TEM exhibited increased granzyme B/perforin expression as compared to naive T cells in response to alloantigen [3]. Thus, these data highlight that while the overall frequency of alloreactive T cells in memory vs. naive T cell compartments may be similar, the behavior of these alloreactive cells in vivo may be quite distinct, depending on their differentiation status.

Advances in our understanding in the generation of donor-reactive T cell memory

How do donor-reactive memory T cells arise in a previously untransplanted recipient? Existing ideas suggested that donor-reactive memory T cells can be generated via pregnancy, exposure to environmental pathogens, and frequent blood transfusions [4,5]. A recent report has conclusively demonstrated the latter phenomenon. Specifically, Zimring and colleagues found that immunity generated against transfused platelets, even across only minor histocompatibility antigens, was sufficient to induce rejection following a subsequent bone marrow transplantation in murine recipients [6]. These data indicate that patients receiving even leuko-reduced platelet products may be at an increased risk for memory T cell-mediated graft rejection following bone marrow transplantation; however further experiments examining the barriers posed by transfusion-derived minor antigens in experimental models of solid-organ transplantation are needed.

As mentioned above, donor-reactive memory T cells have also been purported to arise via exposure to environmental pathogens. Evidence for this exists from both human and mouse models. For example, studies by Burrows and colleagues demonstrated that T cells specific for EBV-EBNA3A restricted by HLA-B8 were cross-reactive with HLA-B44 (putatively presenting a self-peptide) [79]. Earlier studies showed that CD4+ T cells-specific for tetanus toxoid in the context of HLA-DR3 were cross-reactive with HLA-DR4 [10], and a T cell specific for HSV-VP13/14 presented by A2 has been shown to also cross-react with HLA-B44 [11]. Thus, scattered throughout the literature are examples of specific donor-recipient pairs eliciting cross-reactivity between pathogen-specific and alloreactive T cell populations. But just how common is this phenomenon? This year, a new study by Heemskerk and colleagues suggests that it is in fact quite common, perhaps much more so than originally thought. In this elegant series of experiments, Amir et al. analyzed pathogen-specific T cell lines and clones against a panel of HLA-typed target cells, and found that fully 80% of T cell lines and 45% of pathogen-specific T cell clones exhibited alloreactivity [12]. For the pathogen-specific T cell clones, this cross-reactivity was usually confined to a single HLA molecule. Importantly, TCR gene transfer experiments demonstrated that the pathogen and allo-reactivity was conferred by a single TCR, thus confirming cross-reactivity at the molecular level [12]. These results have important implications for the field of transplantation, in that they indicate that the phenomenon of heterologous immunity is perhaps much more common than originally anticipated. Thus, the question becomes not just whether donor reactive memory T cells exist in a given recipient, but rather to what degree their frequency, phenotype, and functionality determines the relative barrier posed by donor-reactive memory T cell populations present in a given individual.

On the nature of T cell allorecognition via molecular mimicry

While the existence of heterolgous immunity in transplantation has been recognized at the cellular level for more than a decade [13], the molecular mechanisms underlying this phenomenon are just beginning to be elucidated. Heterologous immunity as a broader term refers to the observation that one stimulus can lead to a immune response, either cell-mediated or humoral, that affects the response to a second, distinct stimulus [14]. For T cell response, one could conceive of least two possible mechanisms that could contribute to this observation: bystander activation and molecular mimicry. Recently, important experimental evidence supporting the existence of molecular mimicry as an underlying cause of alloreactivity among heterologous immune responses has emerged. For instance, Allen and colleagues published a seminal study analyzing the molecular basis underlying the specificity of alloreactive T cells for peptide:MHC complexes, and found that alloreactive T cells have the ability to respond to multiple, distinct peptide epitopes that share no sequence homology [15]. Thus, this work demonstrated that alloreactive T cells are in fact “poly-specific” and may have the ability to recognize many unrelated peptide sequences [16]. These conclusions were extended by the findings of McCluskey and colleagues, who reported on the molecular mechanisms underlying the observed cross-reactivity of EBV-EBNA3A/HLA-B08 restricted TCR with HLA-B44 molecules [17]. The results of their analyses suggested that dual recognition of unrelated cognate and allogeneic peptide:MHC complexes by a single TCR involved binding modes that were almost identical. Interestingly, only following TCR ligation were the viral and allopeptides forced into this identical conformation, suggesting an induced-fit mechanism of TCR recognition in these instances [17,18]. These seminal studies have therefore definitively demonstrated that heterolgous immunity between pathogen-derived and transplant antigens can function at the level of molecular mimicry.

Cross-reactive allospecific memory T cell repertoire is likely to be more focused than the naïve alloreactive T cell repertoire

While alloreactivity is equally present in naive and memory T cell compartments, the spectrum of specificities within these repertoires is likely to be quite different. Due to the restricted TCR repertoire of pathogen-specific memory T cells [19,20], the alloreactive T cell repertoire among memory is likely to be confined to fewer discrete TCR clones. However, the frequency of any given clone is much higher within the memory pool as compared to the naive T cell repertoire. Thus, given T cell clones within the memory T cell pool are likely to be responding at higher frequency than given clones within the naive T cell pool. Because high T cell precursor frequency is known to constitute a barrier to long-term graft survival induced via costimulation blockade [2123], this may be another method by which donor-reactive memory T cells constitute a barrier to tolerance induction.

Stimulation history profoundly impacts antigen-specific memory T cell quantity, quality, and recall requirements

In recent years, it has become increasingly apparent that a large degree of heterogeneity exists among memory T cells in mouse and man. For example, many groups now have characterized the existence of central (TCM) vs effector (TEM) memory T cells which express different homing receptors that allow them to traffic to the lymphoid organs or peripheral tissues, respectively [24]. These subsets are therefore thought to be responsible for the initiation of recall responses and the surveillance of peripheral tissue sites for invading pathogens, respectively. The functional phenotypes of memory cells can be influenced by a variety of different factors. Current thinking holds that the route of exposure, dose, replication rate, recurrence, and tropism of the infectious challenge may impact qualitative aspects of memory T cell development [25]. For example, there is evidence that repeated exposure to a given antigen results in not only increased quantity of antigen-specific memory T cells, but also altered quality in that the cells became increasingly TEM-like [25. ]. Therefore, donor-reactive memory T cells that are cross-reactive to antigens presented by recurrent infections, such as the common cold, might be present at a higher precursor frequency and be more likely to exhibit a TEM phenotype than donor-reactive memory T cells generated via exposure to a single infection. Are these cells differentially distributed within the body? Are the mobile or sessile? Do they have distinct homing molecules and recirculation patterns? Do they differ with regard to expression of costimulatory and adhesion molecules? Do they possess altered cytokine or cytolytic function? These questions remain to be answered.

Likewise, donor-reactive memory T cells generated following a latent viral infection, such as a herpesvirus infection, might also possess unique phenotypes and functional characteristics. This is likely due to the fact that, unlike acute infections (measles, influenza) where virus is cleared, latent or persistent infections such as EBV, CMV and the polyoma viruses maintain reservoirs of virus within host cells that can periodically reactivate and cause recurrent clinical disease, resulting in populations of persistently activated, viral-specific memory T cells that possess a predominantly TEM phenotype [26]. Due to this recurring antigen exposure and TEM phenotype, these cells may exhibit altered trafficking patterns, obviate the need for initiation of a recall response in secondary lymphoid tissues [27], and demonstrate increased resistance to both tolerance-induction strategies [2830] and common immunosuppressive agents [31].

Thus, understanding the relative barrier posed by different subsets of cells within these heterogeneous populations, particularly in terms of resistance to new costimulation blockade-based therapies for transplantation, remains an important goal.

Effector mechanisms used by memory T cells to mediate rejection

As discussed above, it is likely not just the presence of donor-reactive memory T cells, but their functional phenotypes, that likely determine the potency of the relative barrier they pose to transplantation tolerance induction. Evidence characterizing the varied subsets of memory T cells is mounting, and it is becoming increasingly clear that these cells may mediate their alloreactivity via different effector functions [3].

For example, effector memory T cells are poised in peripheral tissues, with the potential to rapidly respond and infiltrate allografts within 12–24 hours [32]. It was previously unknown as to whether these cells remain static residents of peripheral tissues or continuously recirculate throughout peripheral tissues, and a study published this year now suggests that they in fact recirculate to peripheral tissues and are not static [33]. This finding potentially explains the fact that memory T cells are able to infiltrate transplants so quickly, and also suggests that tissue-resident effector memory T cells could encounter monoclonal antibodies or other therapeutics in the bloodstream, and suggests that achieving high levels of therapeutics in peripheral tissues may not be necessary in order for these reagents to be effective against memory T cells which home there.

Memory T cells are also heterogeneous with regard to their cytokine producing capabilities. Studies of pathogen-specific immune responses and vaccine development first suggested that multi-cytokine producing cells might be more effective at providing protection from reinfection than single cytokine producers [34]. However, it was not known whether this was also true during memory T cell-mediated rejection of transplanted tissue. In 2009, Kirk and colleagues published a study examining the cytokine producing potential of different memory T cell subsets, and found that those T cells that formed the most potent barrier to long-term graft survival (CD2hi TEM cells) were also multi-cytokine producing effector cells [35]. In addition, these cells showed evidence of cytolytic function in that they expressed the cytolytic effector molecule granzyme and showed evidence of degranulation in vitro [35]. Thus, possessing a spectrum of effector functions, each of which could contribute to graft destruction, may determine the relative potency of a given population of memory T cells with regard to their ability to mediate graft rejection.

Potential Strategies for Attenuation of Donor-Reactive Memory T Cell Responses

While a few studies have pointed to an increased risk of rejection in calcineurin-inhibitor-treated recipients possessing an initial high frequency of donor-reactive memory T cells, it is likely that as a general rule calcineurin inhibitors may effectively inhibit the reactivation and effector function of memory T cells [31]. However, in order to improve the side-effect profile of immunosuppressive regimens, many investigators over the past 15 years have contributed to the development of belatacept [36], a second-generation CD28 blocker that has shown immense promise in recent clinical trials [3739]. For example, in a Phase III multi-center clinical trial in renal transplantation, patients treated with belatacept exhibited reduced nephrotoxicity, reduced hyperlipidemia, and a decrease in cardiovascular events as compared to the control group, which received a standard cyclosporine-based immunosuppressive regimen [38]. While these results were certainly promising, this study also revealed that belatacept-treated patients receiving the moderate intensity dosing regimen exhibited a signficant increase in both frequency and severity of acute rejection episodes [38]. From studies by many groups in murine models, it is clear that the pre-existence of donor-reactive memory T cells is a potent barrier to long-term graft survival induced by costimulation blockade [28,40]. Thus, it is interesting to speculate that the pre-existence of donor-reactive memory in some patients may be one risk factor contributing to higher incidence and severity of acute rejection following costimulation blockade-based therapy. Thus, as use of more selective immunosuppressive therapies such as the CD28 costimulation blockers are increased clinically, the need to simultaneously attenuate donor-reactive memory T cell responses by targeting memory cell-specific pathways becomes more pressing.

Over the last year, several such novel pathways have been suggested in the literature. First, as mentioned above, Kirk and colleagues observed the ability of CD2hi TEM cells to mediate costimulation blockade-resistant rejection, and targeted these cells using the CD2 blocker LFA-3-Ig (alefacept) in a non-human primate model of renal transplantation [35]. Blockade of CD2, when used as part of a regimen consisting also of CTLA-4 Ig (CD28 blockade) and sirolimus (mTOR blockade), resulted in renal allograft survival beyond the period of treatment (>90 days) in five out of eight rhesus macaques [35]. These results suggested that alefacept, which is currently FDA-approved for use in plaque psoriasis, may be an appropriate adjunct therapy for use in combination with CD28 blockers such as belatacept.

As discussed above, memory T cells are known to rapidly traffic into grafts and mediate rejection by exacting effector functions and recruiting in other inflammatory mediators. Thus, one potential strategy to limit their pathogenicity might be to block the ability of memory T cells to migrate into the transplanted tissue. Fairchild and colleagues demonstrated that treatment with anti-LFA-1 reduced the early infiltration of memory T cells into donor tissue [41], and we have shown that anti-LFA-1 mAb was effective at inhibiting donor-reactive memory T cell responses when combined with traditional costimulation blockade in murine models [42]. Translational studies in non-human primate models demonstrated that anti-LFA-1 used as an induction agent worked synergistically with belatacept in inhibiting graft rejection in a model of islet transplantation (Badell and Larsen, manuscript in preparation). Furthermore, investigator-initiated exploratory phase II studies using the anti-LFA-1 mAb, efalizumab have suggested efficacy in clinical islet cell transplantation (Larsen, submitted and Peter Stock and Andrew Poselt (UCSF), personal communication). These data suggest that targeting LFA-1, and perhaps more broadly the adhesion pathways used by memory T cells to infiltrate grafted tissue, may be a potent method of attenuating memory T cell-mediated allograft rejection.

If donor-reactive memory T cells are able to infiltrate into the graft, what are the pathways necessary for their in situ effector function? Valujskikh and colleagues assessed the role of inducible costimulatory molecule (ICOS, CD278) in eliciting effector function from early graft-infiltrating memory T cells [43]. Their results revealed that while ICOS was not constitutively expressed on memory T cells, its expression was upregulated in situ following proliferation of memory T cells within the graft itself. Importantly, blockade of ICOS signaling via an anti-ICOS mAb resulted in a significant decrease in the early expression of IFN-γ, perforin, granzyme B, and FasL mRNA within the graft [43].

In addition, reagents which block proteasome degradation and thereby inhibit NFkB nuclear translocation are increasingly showing promise as inhibitors of donor-reactive memory T cell responses. For example, a recent in vitro study demonstrated the ability of bortezomib, one such proteasome inhibitor, to suppress activation of resistant memory T cells in vitro [44]. Importantly, use of bortezomib was shown to preserve regulatory T cell function while inhibiting the activation of donor-reactive memory T cells [44].

Conclusions

Studies published within the last year have definitively demonstrated that not only is alloreactivity present within peripheral T cell compartments of normal healthy individuals, but cross-reactivity between viral-specific T cells and alloeptiopes may in fact be a very common occurrence. Thus, the challenge facing the field over the next decade will be to define the functional phenotypes that exist within memory T cell populations, as a result of the type and frequency of pathogen exposure, that results in a greater or lesser barrier to tolerance induction. For example, allo-crossreactive memory in HLA-B08/B44 donor- recipient pairs is related to a latent pathogen (EBV), while the allo-crossreactive memory found in HLA-DR3/DR4 combinations is related to a single or multi-acute exposure to a cleared antigen (tetanus toxoid). Because these memory T cell type responses generated against these distinct types of exposures may pose different barriers and thus require different therapies, knowing the number and quality of these types of cells for individual patients may be important. Ultimately, one might foresee a strategy in which strength and type of immunosuppression could be tailored to fit the immunological history of a given transplant recipient in order to minimize non-immune toxicities, maximize protective immunity, and prolong graft survival.

Acknowledgments

The authors would like to acknowledge Drs. Allan D. Kirk, William H. Kitchens, and John D. Shires for helpful discussions. C.P.L. is supported by the National Institutes of Health (AI40519, AI073707), the Juvenile Diabetes Research Foundation, and the Mason Trust. M.L.F. is supported by the National Institutes of Health (AI-81789, AI-079409). C.P.L. is a consultant of Bristol-Myers Squibb. M.L.F. has nothing to disclose.

Funding: C.P.L. is supported by the National Institutes of Health (AI40519, AI073707), the Juvenile Diabetes Research Foundation, and the Mason Trust. M.L.F. is supported by the National Institutes of Health (AI-81789, AI-079409).

References

  • 1.Lechler R, Batchelor R, Lombardi G. The relationship between MHC restricted and allospecific T cell recognition. Immunol Lett. 1991;29:41–50. doi: 10.1016/0165-2478(91)90197-i. [DOI] [PubMed] [Google Scholar]
  • 2.Lombardi G, Sidhu S, Daly M, Batchelor JR, Makgoba W, Lechler RI. Are primary alloresponses truly primary? Int Immunol. 1990;2:9–13. doi: 10.1093/intimm/2.1.9. [DOI] [PubMed] [Google Scholar]
  • 3**.Macedo C, Orkis EA, Popescu I, Elinoff BD, Zeevi A, Shapiro R, Lakkis FG, Metes D. Contribution of naive and memory T-cell populations to the human alloimmune response. Am J Transplant. 2009;9:2057–2066. doi: 10.1111/j.1600-6143.2009.02742.x. This article provides evidence that a high precursor frequency of alloreactive T cells exists in the memory T cell pools of normal individuals. [DOI] [PubMed] [Google Scholar]
  • 4.Bingaman AW, Farber DL. Memory T cells in transplantation: generation, function, and potential role in rejection. Am J Transplant. 2004;4:846–852. doi: 10.1111/j.1600-6143.2004.00453.x. [DOI] [PubMed] [Google Scholar]
  • 5.Ford ML, Kirk AD, Larsen CP. Donor-reactive T-cell stimulation history and precursor frequency: barriers to tolerance induction. Transplantation. 2009;87:S69–74. doi: 10.1097/TP.0b013e3181a2a701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Patel SR, Cadwell CM, Medford A, Zimring JC. Transfusion of minor histocompatibility antigen-mismatched platelets induces rejection of bone marrow transplants in mice. J Clin Invest. 2009;119:2787–2794. doi: 10.1172/JCI39590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Burrows SR, Khanna R, Burrows JM, Moss DJ. An alloresponse in humans is dominated by cytotoxic T lymphocytes (CTL) cross-reactive with a single Epstein-Barr virus CTL epitope: implications for graft-versus-host disease. J Exp Med. 1994;179:1155–1161. doi: 10.1084/jem.179.4.1155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Burrows SR, Silins SL, Moss DJ, Khanna R, Misko IS, Argaet VP. T cell receptor repertoire for a viral epitope in humans is diversified by tolerance to a background major histocompatibility complex antigen. J Exp Med. 1995;182:1703–1715. doi: 10.1084/jem.182.6.1703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Burrows SR, Silins SL, Khanna R, Burrows JM, Rischmueller M, McCluskey J, Moss DJ. Cross-reactive memory T cells for Epstein-Barr virus augment the alloresponse to common human leukocyte antigens: degenerate recognition of major histocompatibility complex-bound peptide by T cells and its role in alloreactivity. Eur J Immunol. 1997;27:1726–1736. doi: 10.1002/eji.1830270720. [DOI] [PubMed] [Google Scholar]
  • 10.Umetsu DT, Yunis EJ, Matsui Y, Jabara HH, Geha RS. HLA-DR-4-associated alloreactivity of an HLA-DR-3-restricted human tetanus toxoid-specific T cell clone: inhibition of both reactivities by an alloantiserum. Eur J Immunol. 1985;15:356–361. doi: 10.1002/eji.1830150410. [DOI] [PubMed] [Google Scholar]
  • 11.Koelle DM, Chen HB, McClurkan CM, Petersdorf EW. Herpes simplex virus type 2-specific CD8 cytotoxic T lymphocyte cross-reactivity against prevalent HLA class I alleles. Blood. 2002;99:3844–3847. doi: 10.1182/blood.v99.10.3844. [DOI] [PubMed] [Google Scholar]
  • 12**.Amir AL, D’Orsogna LJ, Roelen DL, van Loenen MM, Hagedoorn RS, de Boer R, van der Hoorn MA, Kester MG, Doxiadis II, Falkenburg JH, et al. Allo-HLA reactivity of virus-specific memory T-cells is common. Blood. 2010 doi: 10.1182/blood-2009-07-234906. Feb 16 Epub ahead of print. This study suggests that T cell cross-reactivity between pathogen-derived epitopes and alloantigens is more common than originally appreciated. [DOI] [PubMed] [Google Scholar]
  • 13.Adams AB, Pearson TC, Larsen CP. Heterologous immunity: an overlooked barrier to tolerance. Immunol Rev. 2003;196:147–160. doi: 10.1046/j.1600-065x.2003.00082.x. [DOI] [PubMed] [Google Scholar]
  • 14.Selin LK, Brehm MA, Naumov YN, Cornberg M, Kim SK, Clute SC, Welsh RM. Memory of mice and men: CD8+ T-cell cross-reactivity and heterologous immunity. Immunol Rev. 2006;211:164–181. doi: 10.1111/j.0105-2896.2006.00394.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Feldman HI, Kobrin S, Wasserstein A. Hemodialysis vascular access morbidity. Journal of the American Society of Nephrology. 1996;7:523–535. doi: 10.1681/ASN.V74523. [DOI] [PubMed] [Google Scholar]
  • 16.Felix NJ, Allen PM. Specificity of T-cell alloreactivity. Nat Rev Immunol. 2007;7:942–953. doi: 10.1038/nri2200. [DOI] [PubMed] [Google Scholar]
  • 17**.Macdonald WA, Chen Z, Gras S, Archbold JK, Tynan FE, Clements CS, Bharadwaj M, Kjer-Nielsen L, Saunders PM, Wilce MC, et al. T cell allorecognition via molecular mimicry. Immunity. 2009;31:897–908. doi: 10.1016/j.immuni.2009.09.025. This study suggests that one of the major mechanisms underlying heterologous immunity is cross-reactivity at the molecular level. [DOI] [PubMed] [Google Scholar]
  • 18.Beddoe T, Chen Z, Clements CS, Ely LK, Bushell SR, Vivian JP, Kjer-Nielsen L, Pang SS, Dunstone MA, Liu YC, et al. Antigen ligation triggers a conformational change within the constant domain of the alphabeta T cell receptor. Immunity. 2009;30:777–788. doi: 10.1016/j.immuni.2009.03.018. [DOI] [PubMed] [Google Scholar]
  • 19.Sourdive DJ, Murali-Krishna K, Altman JD, Zajac AJ, Whitmire JK, Pannetier C, Kourilsky P, Evavold B, Sette A, Ahmed R. Conserved T cell receptor repertoire in primary and memory CD8 T cell responses to an acute viral infection. Journal of Experimental Medicine. 1998;188:71–82. doi: 10.1084/jem.188.1.71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.van Leeuwen EM, Remmerswaal EB, Heemskerk MH, ten Berge IJ, van Lier RA. Strong selection of virus-specific cytotoxic CD4+ T-cell clones during primary human cytomegalovirus infection. Blood. 2006;108:3121–3127. doi: 10.1182/blood-2006-03-006809. [DOI] [PubMed] [Google Scholar]
  • 21.Ford ML, Floyd TL, Wagener ME, Hanna SS, Stempora L, Hendrix R, Kirk AD, Larsen CP. Impact of Donor-reactive precurosr frequency on the differentiation of costimulation independent CD8+ memory T cells. Am J Transplant. 2009;9:333. [Google Scholar]
  • 22.Ford ML, Wagener ME, Hanna SS, Pearson TC, Kirk AD, Larsen CP. A critical precursor frequency of donor-reactive CD4+ T cell help is required for CD8+ T cell-mediated CD28/CD154-independent rejection. J Immunol. 2008;180:7203–7211. doi: 10.4049/jimmunol.180.11.7203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Koehn BH, Ford ML, Ferrer IR, Borom K, Gangappa S, Kirk AD, Larsen CP. PD-1-dependent mechanisms maintain peripheral tolerance of donor-reactive CD8+ T cells to transplanted tissue. J Immunol. 2008;181:5313–5322. doi: 10.4049/jimmunol.181.8.5313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Sallusto F, Geginat J, Lanzavecchia A. Central memory and effector memory T cell subsets: function, generation, and maintenance. Annu Rev Immunol. 2004;22:745–763. doi: 10.1146/annurev.immunol.22.012703.104702. [DOI] [PubMed] [Google Scholar]
  • 25.Masopust D, Ha SJ, Vezys V, Ahmed R. Stimulation history dictates memory CD8 T cell phenotype: implications for prime-boost vaccination. J Immunol. 2006;177:831–839. doi: 10.4049/jimmunol.177.2.831. [DOI] [PubMed] [Google Scholar]
  • 26.Amyes E, Hatton C, Montamat-Sicotte D, Gudgeon N, Rickinson AB, McMichael AJ, Callan MF. Characterization of the CD4+ T cell response to Epstein-Barr virus during primary and persistent infection. J Exp Med. 2003;198:903–911. doi: 10.1084/jem.20022058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Obhrai JS, Oberbarnscheidt MH, Hand TW, Diggs L, Chalasani G, Lakkis FG. Effector T cell differentiation and memory T cell maintenance outside secondary lymphoid organs. J Immunol. 2006;176:4051–4058. doi: 10.4049/jimmunol.176.7.4051. [DOI] [PubMed] [Google Scholar]
  • 28.Adams AB, Williams MA, Jones TR, Shirasugi N, Durham MM, Kaech SM, Wherry EJ, Onami T, Lanier JG, Kokko KE, et al. Heterologous immunity provides a potent barrier to transplantation tolerance. J Clin Invest. 2003;111:1887–1895. doi: 10.1172/JCI17477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Brook MO, Wood KJ, Jones ND. The impact of memory T cells on rejection and the induction of tolerance. Transplantation. 2006;82:1–9. doi: 10.1097/01.tp.0000226082.17507.da. [DOI] [PubMed] [Google Scholar]
  • 30.Wu Z, Bensinger SJ, Zhang J, Chen C, Yuan X, Huang X, Markmann JF, Kassaee A, Rosengard BR, Hancock WW, et al. Homeostatic proliferation is a barrier to transplantation tolerance. Nat Med. 2004;10:87–92. doi: 10.1038/nm965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Pearl JP, Parris J, Hale DA, Hoffmann SC, Bernstein WB, McCoy KL, Swanson SJ, Mannon RB, Roederer M, Kirk AD. Immunocompetent T-cells with a memory-like phenotype are the dominant cell type following antibody-mediated T-cell depletion. Am J Transplant. 2005;5:465–474. doi: 10.1111/j.1600-6143.2005.00759.x. [DOI] [PubMed] [Google Scholar]
  • 32.Schenk AD, Nozaki T, Rabant M, Valujskikh A, Fairchild RL. Donor-reactive CD8 memory T cells infiltrate cardiac allografts within 24-h posttransplant in naive recipients. Am J Transplant. 2008;8:1652–1661. doi: 10.1111/j.1600-6143.2008.02302.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Liu L, Zhong Q, Tian T, Dubin K, Athale SK, Kupper TS. Epidermal injury and infection during poxvirus immunization is crucial for the generation of highly protective T cell-mediated immunity. Nat Med. 2010;16:224–227. doi: 10.1038/nm.2078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Seder RA, Darrah PA, Roederer M. T-cell quality in memory and protection: implications for vaccine design. Nat Rev Immunol. 2008;8:247–258. doi: 10.1038/nri2274. [DOI] [PubMed] [Google Scholar]
  • 35**.Weaver TA, Charafeddine AH, Agarwal A, Turner AP, Russell M, Leopardi FV, Kampen RL, Stempora L, Song M, Larsen CP, et al. Alefacept promotes co-stimulation blockade based allograft survival in nonhuman primates. Nat Med. 2009;15:746–749. doi: 10.1038/nm.1993. These data provide evidence that CD2 blockade can attenuate alloreactive memory T cells and prolong renal graft survival in non-human primates. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Larsen CP, Pearson TC, Adams AB, Tso P, Shirasugi N, Strobertm E, Anderson D, Cowan S, Price K, Naemura J, et al. Rational development of LEA29Y (belatacept), a high-affinity variant of CTLA4-Ig with potent immunosuppressive properties. Am J Transplant. 2005;5:443–453. doi: 10.1111/j.1600-6143.2005.00749.x. [DOI] [PubMed] [Google Scholar]
  • 37.Vincenti F, Larsen C, Durrbach A, Wekerle T, Nashan B, Blancho G, Lang P, Grinyo J, Halloran PF, Solez K, et al. Costimulation blockade with belatacept in renal transplantation. N Engl J Med. 2005;353:770–781. doi: 10.1056/NEJMoa050085. [DOI] [PubMed] [Google Scholar]
  • 38*.Vincenti F, Charpentier B, Vanrenterghem Y, Rostaing L, Bresnahan B, Darji P, Massari P, Mondragon-Ramirez GA, Agarwal M, Russo GD, et al. A Phase III Study of Belatacept-based Immunosuppression Regimens versus Cyclosporine in Renal Transplant Recipients (BENEFIT Study) Am J Transplant. 2010;10:535–546. doi: 10.1111/j.1600-6143.2009.03005.x. This study and the one following demonstrated that a CD28 blockade-based regimen can inhibit graft rejection in renal transplant patients with fewer associated non-immune toxicities than calcineurin inhibitor-based regimens. [DOI] [PubMed] [Google Scholar]
  • 39*.Durrbach A, Pestana JM, Pearson T, Vincenti F, Garcia VD, Campistol J, Rial MdC, Florman S, Block A, Russo GD, et al. A Phase III Study of Belatacept Versus Cyclosporine in Kidney Transplants from Extended Criteria Donors (BENEFIT-EXT Study) Am J Transplant. 2010;10:547–557. doi: 10.1111/j.1600-6143.2010.03016.x. [DOI] [PubMed] [Google Scholar]
  • 40.Pantenburg B, Heinzel F, Das L, Heeger PS, Valujskikh A. T cells primed by Leishmania major infection cross-react with alloantigens and alter the course of allograft rejection. J Immunol. 2002;169:3686–3693. doi: 10.4049/jimmunol.169.7.3686. [DOI] [PubMed] [Google Scholar]
  • 41.Setoguchi K, Schenk A, Ishii D, Tanabe K, Fairchild RL. Anti-Lymphocyte Function-Associated Antigen-1 (LFA-1) Monoclonal Antibody Prevents Graft-Infiltrating CD8 Memory T Cells Early Post-Transplant. Am J Transplant. 2009;9:209. [Google Scholar]
  • 42.Ford ML, Steeds CM, Wagener ME, Robertson JM, Hanna SS, Kirk AD, Larsen CP. Anti-LFA-1 Synergizes with CD28/CD154 Blockade To Inhibit Anti-Donor Cytotoxicity and Promote Graft Survival in Recipients Possessing Donor-Reactive Memory T Cells. Am J Transplant. 2010;10(s2):9. [Google Scholar]
  • 43.Schenk AD, Gorbacheva V, Rabant M, Fairchild RL, Valujskikh A. Effector functions of donor-reactive CD8 memory T cells are dependent on ICOS induced during division in cardiac grafts. Am J Transplant. 2009;9:64–73. doi: 10.1111/j.1600-6143.2008.02460.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Kim JS, Lee JI, Shin JY, Kim SY, Shin JS, Lim JH, Cho HS, Yoon IH, Kim KH, Kim SJ, et al. Bortezomib can suppress activation of rapamycin-resistant memory T cells without affecting regulatory T-cell viability in non-human primates. Transplantation. 2009;88:1349–1359. doi: 10.1097/TP.0b013e3181bd7b3a. [DOI] [PubMed] [Google Scholar]

RESOURCES