Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Jun 1.
Published in final edited form as: Curr Opin Immunol. 2010 May 4;22(3):402–410. doi: 10.1016/j.coi.2010.04.002

Designing CD8+ T Cell Vaccines: It's Not Rocket Science (Yet)

Jonathan W Yewdell 1
PMCID: PMC2908899  NIHMSID: NIHMS214997  PMID: 20447814

Abstract

CD8+ T cells play important roles in clearing viral infections and eradicating tumors. Designing vaccines that elicit effective CD8+ T cell responses requires a thorough knowledge of the pathways of antigen presentation in vivo. Here, I review recent progress in understanding the activation of naïve CD8+ T cells in vivo, with particular emphasis on cross-priming, the presentation of protein antigens acquired by dendritic cells from their environment. With the rapid advances in this area of research, the dawn of rational vaccine design is at hand.

Introduction

From the time of Jenner's introduction of scientific method-based vaccination until the present day, vaccination for viral diseases has been based on administration of modified intact or fragmented viruses. For most important human viral pathogens, this empiric approach has been sufficient, as viral vaccines are one of the greatest successes of modern medicine. For a substantial number of pathogenic viruses, however, the empiric approach of vaccine design has failed. Moreover, it is becoming increasingly likely that therapeutic vaccination can play an important role in treating established diseases, particularly cancer, where the immunosurveillance theory has made a Lazarus-like reappearance1.

Harnessing the full potential of the immune system to prevent and treat diseases will require rational vaccine design. Just as rocket science is rooted in Newton's laws of physics, engineering vaccines to precisely target pathogens and malignant cells requires establishing the laws of immunity. While there has been tremendous progress in understanding the immune system in all its complex glory, much remains to be learned before vaccines can be precisely engineered based on firmly established principles. Here, I review recent progress in understanding the induction of CD8+ T cell responses, which play important roles in clearing viruses and other pathogens, and in preventing and eradicating tumors. In keeping with the theme of this volume of Current Opinion in Immunology, I emphasize practical issues that impact vaccine design.

The Basics

CD8+ T cells typically express a clonally restricted αβ T cell receptor that recognize one of the “classical” MHC class I molecules bearing oligopeptides (normally 8 to 11 residues) in their binding groove. The cell presenting the MHC class I peptide complex (CIPC) is termed the antigen presenting cell (APC). To activate naïve CD8+ T cells to generate effector and particularly memory T cells, the goal of initial vaccination, APCs must express a combination of co-stimulatory cell surface and secreted molecules. In the absence of co-stimulation, CD8+ T cells are tolerized by interacting with cells expressing their cognate CIPCs, providing anti-vaccination, but potentially useful in treating or preventing autoimmunity.

To avoid tolerance induction, naïve T cells limit their peregrinations to lymphoid tissues where they will exclusively encounter foreign antigen presented by “professional” antigen presenting cells (pAPCs), i.e. bone marrow lineage cells that express/secrete the appropriate co-stimulatory molecules for T cell activation. A key question in vaccinology are the identities of pAPCs in different vaccination/infection scenarios, and how differences between pAPCs influence CD8+ T cell proliferation, effector function, and memory differentiation.

APCs can generate peptides from two potential sources: polypeptides they have synthesized on their own ribosomes, or polypeptides synthesized by other means. The former is termed direct presentation (or direct priming if the CD8+ T cell is naïve) while the latter is termed cross-presentation/priming. Gene based infectious agents/vectors are potentially presented by either route, while by definition, protein/peptide based immunogens are presented by cross-presentation. Peptides can be proteolytically processed for cross-presentation in two compartments: endolysosomes or cytosol. In the latter case, antigen is processed by the standard endogenous pathway proteases (including the proteasome) and liberated peptides are transported into the endoplasmic reticulum (ER) by the transporter associated with antigen processing (TAP). In the former case, loading occurs in an endolysosomes compartment and presentation is TAP- and proteasome-independent.

What Do Vaccinologists Need to Know?

Rational vaccine design has two components.

First, what is the desired response? What specificities (i.e. class I peptide complexes) should be induced? What types of CD8+ T cells provide optimal functionality? What anatomic locations should be the focus of the response?

Second, how should these responses be generated? What immunogens should be used? What dose? What route and site of immunization? How many boosts?

Designing the first vaccine component requires a thorough understanding of the interaction between the target agent and the host. For each system, it is necessary to understand what contributes to the effectiveness of the CD8+ T cell response. Many mysteries remain. Superficially, it would seem that vaccines should induce responses to as many immunogenic peptides as possible. But it appears likely that some CIPCs provide superior targets; than others for CD8+ T cell effector activity 2, 3. In this case, a narrower response is probably advantageous, since CD8+ T cells typically compete with each other in the priming, and particularly the boosting phase of vaccination 4. In this regard, vectors that elicit the minimal response to vector antigens should be greatly preferred over more complex vectors. Poxviruses, with hundreds of open reading frames, dozens of which are immunogenic 5, are particularly ill suited as CD8+ T cell vectors. Despite their present popularity, they should ultimately be replaced with less ornate vectors, unless immunity to pathogenic poxviruses is an associated goal of vaccination.

Designing the second vaccine component requires a thorough knowledge of the immune system. Basic understanding of immunity will come largely from insights generated by mouse model systems. The power of mouse systems grows at a much faster rate than other systems, due to multiple factors including cost, ease of genetic manipulation, availability of reagents, and not the least, the capacity to study immune events in real time by intravital imaging via multiphoton microscopy, which is being rapidly extended to microbial pathogens 6.

The half-joking phrase HIV vaccinologists employ to summarize decades of disappointments with animal vaccination models, “mice lie and monkeys mislead”, has a germ of truth, however. Mice and men are different creatures, and major differences in viral tropism and innate immune sensors, limit the predictive powers of mouse vaccine studies, particularly for cell-mediated responses. While studies in sub-human primates will provide a bridge to humans, human trials will remain just that: trials with a substantial chance of failure.

Routes of Natural Priming

A critical basic question in viral immunology with practical ramifications for vaccinology is the natural route of priming of anti-viral CD8+ T cells. Robust cross-priming of anti-viral CD8+ T cells is easy to demonstrate experimentally by simply injecting either protein immunogens or virus-infected cells incapable of producing infectious virions 7. Further, the robust mouse CD8+ T cell response to mouse cytomegalovirus (MCVM) in the face of multiple efficient mechanisms to block direct presentation, argues strongly for the physiological relevance of cross-priming 8-10.

Despite mounting evidence, the physiological significance of cross-priming continues to be questioned (though even Zinkernagel and acolytes, once highly skeptical 11, appear to have accepted its relevance 12). Kemball et al. 13, 14 reported that despite reaching enormous titers in multiple organs, Coxsackievirus B3 (CVB) induces barely detectable CD8+ T cell responses. CVB, like other picornaviruses, blocks exocytosis and thereby disrupts endogenous antigen presentation, which is dependent on export of CIPCs from the ER 15. Kemball et al. 13, 14 argued that given a potent mechanism to block direct priming, the poor immunogenicity of CVB undermines the physiological contribution of cross-priming to viruses in general.

Notably, however, the endogenous anti-CVB CD4+ T cell response was also deficient 13, and a transgenic TCR CD4+ T cell response was weak to an inserted antigen 14. Since cross-priming is well established as the major mechanism in generating MHC class II peptide complexes, this suggests that CVB suppresses T cell responses independently of the route of antigen presentation. Further, the failure to detect transgenic TCR CD8+ T cell responses to inserted determinants, may be related to the strategy for expressing the determinants, whereby the determinants are rapidly released as oligopeptides from viral proteins, and likely to be rapidly degraded, and hence, poor at cross-priming (see below).

In any event, these intriguing findings with CVB findings stress the need to extend mouse studies from the handful of heavily studied model virus systems (LCM, vaccinia, influenza, herpes) to a wider variety of viruses. They also underscore the importance of using new approaches for discriminating the contributions of direct- vs. cross-priming in vivo.

Fortunately, a number of new approaches have been described for tackling this question.

  1. Modifying class I trafficking. Lizee et al reported that modifying the cytoplasmic domain of class I molecules interferes with their trafficking to endosomal compartments, and when such class I molecules are expressed in transgenic mice, reduces cross-priming 16. Although this would not affect cross-priming via cytosolic delivery, it provides a means for gauging the contribution of endolysosomal processing, which appeared play a major role in CD8+ T cell responses to vesicular stomatitis virus, and particularly, inactivated Sendai virus.

  2. Knocking out or modifying APC subsets. Exploiting the selective shut down in cross-presentation that occurs upon DC activation in vitro, Wilson et al. showed that activating DCs in vivo by injecting TLR ligands blocks in vivo cross-priming to protein antigens 17. DC pre-activation reduced CD8+ T cell responses to herpes simplex and influenza A viruses, supporting an important contribution of cross-priming to these viruses, and demonstrating the potential of this approach. Indeed, this approach was soon exploited to demonstrate the importance of cross-priming to a modified (vaccinia) virus Ankara (MVA) encoded antigen 18.

    Lin et al. reported that injection of cytochrome C selectively ablated cross-presenting DCs, due to the pro-apoptotic activity of cytochrome C following its delivery to the cytosol 19 This is potentially a powerful approach, but is limited to the extent that cross-presenting cells also participate in direct-priming.

    Hildner et al. described a novel knockout mouse (BATf3) that lacks development of CD8αα homodimer expressing (CD8α+) and CD103+ (langerin + CD8α-) -DC subsets 20, together to play a central role in cross-priming 21. Depending on the importance of these DCs in direct priming, this could be a useful strain for weighing the contribution of direct vs. cross-priming.

  3. Drug-modulation of antigen processing. Barnaba and colleagues reported that chloroquine enhances cross-priming by retarding antigen degradation in endolysosomes and thereby enhancing cytosolic delivery 22, 23. Chloroquine-enhanced immunogenicity is therefore, potentially an indicator of cytosolic-based cross-priming. Importantly, from the practical standpoint, as chloroquine is a widely used and well tolerated drug, it has potential as an adjuvant for cross-priming vaccines.

    Conversely, Basler et al. reported that the proteasome inhibitor bortezomib (a.k.a. Velcade or PS341) reduces CD8+ T cell responses by blocking generation of CiPCs (and not by interfering with T cell proliferation)24. Bortezomib-resistant immunogenicity is therefore, potentially an indicator of endolysosomal based cross-priming.

  4. Genetic modulation of antigen processing. Saveanu et al. reported that knocking out mouse endosomal protease insulin regulated amino peptidase (IRAP) selectively interferes with cross-priming 25, implicating IRAP-trimming in endosomal dependent cross-presentation, and providing a new target for selectively modulating cross-presentation. Mice lacking ERAP (ER associated aminopeptidase, which trims TAP-transported peptides) are also available, but their antigen processing phenotype is complex 26-28.

  5. Caveat. Antigen processing pathways are complex, and in addition to “non-classical” connections between theoretically distinct pathways, there can be considerable cross-talk between the pathways (e.g. knocking out TAP decreases numbers of peptide receptive molecules available for endolysosomal loading 29, 30.) It is essential, therefore, in given experimental scenarios to perform functional control experiments that document the specificity of the manipulation for the given pathways.

Mechanisms of Cross Priming

Protein vs. Peptide?

For gene-based CD8+ T cell vaccines the physical nature of the cross-priming antigen is of paramount importance, since it dictates antigen expression strategy. In 2004, three groups simultaneously reported that the relevant form of antigen for cross-priming are proteins (i.e. proteasome substrates), rather than proteasome products 31-33, casting doubt on the importance of chaperoned peptides in cross-priming 34. Subsequent studies in a number of different systems confirmed that antigen stability in cells is a determining factor in cross-priming potency 18, 35-38. Lev et al. reinforced this conclusion by reporting that peptides, if metabolically stable, are capable of robust cross-priming 39. Thus, the poor immunogenicity of peptides in cross-priming appears to be attributable to the law of mass action, i.e. their amounts in donor cells are typically too low to be immunogenic.

Whatever the role of chaperoned-peptides in physiological cross-priming, they still might be effective vaccines. Indeed, Oizumi et al. reported that expression of a secreted form of gp96-fused to the mouse IgG1 Fc domain enormously enhances the immunogenicity of a nominal antigen synthesized by the same cell 40. These findings are partially consistent with those of Nicchitta and colleagues, who first reported the adjuvant effects of secreted gp96, but failed to detect and increase in immunogenicity of antigens from cells secreting gp96 41. Further, the evidence continues to mount that the immunogenicity of purified molecular chaperones is poor in the absence of contaminants introduced during their production and purification that trigger innate immune receptors and provide adjuvant activity42-44.

It appears that with few exceptions 45, the affinity for molecular chaperones for peptides is insufficient to preserve sufficient quantities for cross-priming in the natural setting 46. The potency of chaperones-based vaccines is likely largely dependent on their affinity with their antigenic cargo. A general strategy to achieve high affinity interaction is to stably attach (via chemical cross-linking or producing a genetic/synthetic fusion protein) the immunogen to a high affinity ligand for the relevant chaperone 47, 48.

Antigen Acquisition for Cross-Priming

Cross-priming DCs can potentially obtain their antigen via multiple routes. For cell derived-antigens, antigen can be transferred to DCs by nibbling from live antigen expressing cells 49, or by phagocytosis from dead antigen expressing cells. It appears, however, that cross-priming antigen cannot be salvaged from every cellular compartment. Tewalt et al. reported that vaccinia virus encoded proteins sequestered in viral factories (i.e. viral assembly sites) are not available for CD8+ T cell cross-priming 50. Since CD4+ T cell cross-priming was not inhibited, Tewalt et al. concluded that cross-priming DCs were able to acquire factory antigens, but could only process them in endolysosomes and not export them to the cytosol for class I processing.

Much remains to be learned about the DC antigen acquisition process, which is also critical in tolerizing T cells to self antigens in the absence of inflammatory signals. While there is considerable literature regarding the cross-priming immunogenicity of live vs. apoptotic vs. necrotic cells 51, many findings are contradictory and the message for optimizing gene-based vaccines is clouded. Plesa et al. generated recombinant rabies viruses that differ markedly in their cytopathic effect based on just two amino acid differences in an inserted protein, and found that cytopathicity increased cross-priming upon injection of infected cells 52. There was little effect on immunogenicity of infectious virus, however, leading Please et al. to conclude that direct priming dominates in this system. Still, co-expression of death inducing/preventing gene products represents a viable strategy for optimizing cytopathogenicity in cross-priming of other gene-based vectors.

More broadly, manipulating cell death signals may also enhance protein based cross-priming. Sancho et al. identified CLEC9A as necrotic cell detector that enhances the cross-presentation by a still to be established mechanism 53. This builds on Carminschi et al.'s original demonstration that CLEC9A is selectively expressed on mouse CD8α+ and plasmacytoid DCs, and that targeting protein immunogens via CLEC9 greatly enhances their immunogenicity 54.

DCs can also acquire pre-formed CIPCs from other APCs. “Trogocytosis” was coined by Joly and Hudrisier to connote the intercellular transfer of plasma membrane proteins during cellular interactions 55. I coined “cross-dressing” to describe DCs trogocytosis of CIPC from other cells 56 as a potential means for amplifying CD8+ T cell responses, as originally suggested by Fazekas de St Groth and colleagues 57. Using a mouse tumor model, Dolan et al. initially described in vivo cross-dressing 58. In a further, fascinating twist, Qu et al. demonstrated cross-dressing of priming DCs from monocytes that generated CIPCs from phagocytosed antigen obtained from dead cells 59 (i.e. cross-dressing of cross-presented antigen!). (Note that this information transfer probably also extends to co-stimulatory and inhibitory molecules. Indeed, it appears the even TCRs can be exchanged between activated and naïve CD8+ T cells by trogocytosis, recruiting additional effector cells and adding an unexpected wrinkle to the clonal selection theory of lymphocyte function 60).

More wonders abound: Neefjes and colleagues described peptide transfer to DCs from donor cells via gap junctions 61, and recently expanded the relevance to multiple systems 62, 63. Gap junctions form between connexin 43-expressing DCs and other connexin 43-expressing cells. Connexins form six sided membrane channels that connect the cytosol of communicating cells, enabling the passage of flexible peptides and other small diameter molecules. The contribution of GAP junctional transfer to in vivo priming remains to be established. It is more likely to contribute to direct priming than cross-priming since direct priming in most cases generates greater quantities of peptides suitable for gap junctional transfer. In any event, it would be of interest to examine the adjuvant effect of connexin 43 expression in gene-based vaccines.

A New Cross-Presentation Compartment

It is beyond the scope of this review to discuss the recent progress in unraveling the cell biological mechanisms that enable cross-presentation, except for a recent set of findings with important implications for protein-based vaccines. Kurts and colleagues described a novel endosomal compartment accessed by the mannose receptor that participates in cross presentation in bone marrow derived CD8α+DCs and in cross-priming in vivo 64-66. Remarkably, TLR-activation recruits TAP to endosomes, providing a pathogen-specific mechanism for regulating cross-presentation. Presentation of a model antigen was proteasome dependent, and evidence suggested the export of the antigen to proteasomes associated with the cytosolic face of endosomes, with TAP-mediated re-import of peptides into the originating endosome, where they associate with peptide receptive class I molecules. A similar process was previously proposed for phagocytosed material 67. Such a mechanism could greatly increase the efficiency of cross-presentation by targeting locally generated, pathogen-derived peptides to TAP and limiting competition from cellular peptides. Peptides might also be directly generated in the mannose receptor compartment, particularly since IRAP is reported to be present 25.

If a similar compartment exists in human DCs that cross-prime in vivo 68, targeting protein antigens to this compartment through interaction with the mannose receptor (or potentially other yet to be discovered receptors), could enable efficient protein-immunogen based cross-priming.

Who's Priming In vivo?

DCs rule (for now)

A large number of experiments in mice point to the central importance of DCs in both direct and cross-priming. Cross-priming appears to be particularly dependent on CD8α+ and CD103+ DCs (whose relative importance likely varies with circumstances of immunization and between immune organs 69), and, it will be important to see if future studies with the Batf3 knockout mice affirm this conclusion. CD8α+DCs have a number of adaptations that may account for their enhanced cross-priming ability. These include optimizing endolysosomal pH and composition 70 and CLEC9 regulated-cross-presentation71. The importance of CD103+ DCs in cross-priming has only recently become clear 72 73, and much remains to be learned about their special adaptations for cross-priming. CD103+ DCs are a migrating subset that transports antigens from the periphery to the draining nodes, and they are likely to be of central importance in cross-priming of immunogens delivered by local injection with limited access to the circulation.

To translate the mouse model findings to human immunology it is critical to establish whether there is are equivalent DC subsets in humans (which do not have a CD8α+ DC subset) particularly adept at cross-priming 74. Galibert et al. provided the initial evidence for the equivalence of human BDCA3+ DCs with mouse CD8α+ DCs 75, and the conserved expression of CLEC9 54 and CLEC12 76 supports the relationship. It is of obvious importance to characterize the functional characteristics of BDCA3+ DCs, and to determine whether they are present in sub human primates, where experimental in vivo manipulation is possible.

pAPC Wannabes

Although thousands of studies point to the central importance of DCs in cross-priming CD8+ T cells, other cell types have been identified that are capable of efficient cross-priming, including in recent years, plasmacytoid DCs 68, interferon producing killer dendritic cells 77 and neutrophils 78. None of these cell types are thought to be present in significant quantities when and where priming occurs in immune organs, undermining their potential participation in priming. On the other hand, although it is assumed that priming exclusively occurs in draining lymph nodes and spleen, effective (though delayed) anti-viral CD8+ T cell priming can occur in lymphotoxin-α -/- mice, which lack lymph nodes and have a disorganized spleen 79 (indeed, the gp96-Ig fusion protein discussed above highly immunogenic in these mice 40).

Intravital Microscopy to the Rescue

In any event, conclusive definition of naturally priming APCs must include anatomical evidence for the interaction between naïve CD8+ T cells and the priming APCs. The recent advances in intravital microscopy, coupled with the generation of transgenic mice and microbial vectors expressing fluorescent proteins under tissue specific promoters, provide an unprecedented opportunity for characterizing immune activation in real time in something approaching to natural conditions 6. This enabled Hickman et al. to demonstrate that direct priming to viruses introduced subcutaneously occurs in a newly defined anatomic region in the draining lymph node, termed the peripheral interfollicular region 80. Intravital microscopy also revealed that lymph-borne viruses are transported from the subcapsular sinus into the lymph node parenchyma by macrophages that sample the subcapsular fluid 81.

Intravital microcopy is poised to rewrite the rules of immunogenicity, and surprises regarding the APCs involved and the anatomic sites of T cell priming are likely. A key challenge in the future is to adapt intravital microscopy to non-human primate models.

Practical cross-priming –optimizing antigen, route, conditions

Gene Based Immunogens

For gene-based priming, if the route of priming is known, there is a simple rule for selecting the form of the expressed antigen. Direct priming is optimized by expressing rapidly degraded polypeptides (exception: some minigene products are degraded so rapidly that immunogenicity is reduced relative to even the full-length source polypeptide 82). Conversely, cross-priming is optimized by expressing long lived antigens. If the route of priming is uncertain, it is best to express long-lived antigens. This will optimize cross-priming, and will be reasonably effective in direct priming, since even directing 100% of nascent antigen to proteasomes only increases peptide production by two- to three-fold, due to the efficiency of the DRiP pathway in generating peptides from stable proteins 83.

Polypeptide based Immunogens

Extended Peptides

Optimally sized synthetic peptides (for class I affinity) are generally ineffective immunogens in humans. Wei and Sherman reported that adding just 3 residues to the amino terminus of three different optimal 8- or 9-mer peptides greatly increased their cross-presentation capacity 84. Presentation was TAP-dependent, implying cytosolic delivery, and the enhancing effect was dependent on the trimer extension sequence, an important consideration in designing such peptide immunogens. Relating these findings to Lev et al.'s report of stable cytosolic peptides 39 could provide great insight into the intersection of cross-priming and direct priming pathways, and shed light on the metabolic stabilities of oligopeptides in the cytosol.

DC cross-presentation of proteins 85 and extended peptides 86 lasts for days after antigen acquisition, due to the storage of antigen in a lysosome-like depot compartment in matured DCs 86. The increased immunogenicity of extended peptides is likely due to two factors: resistance to protease destruction (any proteolysis will drastically diminish binding of minimal peptides to class I molecules), and restricting presentation to cross-presenting DCs, since minimal peptides will generate large number of CIPCs on nearly every cell type they accesses, leading to tolerance induction 87.

Success!

Melief and colleagues have spectacularly demonstrated the vaccine potential of long peptides. Immunizing cervical carcinoma patients with a incomplete Freund's adjuvant containing a mixture of 13 different 20-30+ mer peptides corresponding to human papilloma virus 16 transforming proteins, they found remarkable clinical responses related to induction of anti-viral CD8+ T cell responses 88, 89. A key advantage of this approach is that multiple HLA class I and II allomorphs are likely to be covered by the mixed peptide immunogen.

Encapsulated Antigens

Advances in material science offer great promise for polypeptide based vaccines introduced in particulate form. It will take time to optimize the parameters that influence immunogenicity, including route of immunization 90 method of antigen attachment, particle size 91 and composition (synthetic 92, 93, or self-assembling, e.g. virus like particles 94). A general principle is that immunogenicity is greatly enhanced by delivering antigen and TLR-(or other innate immune receptor) activating substances in the same particle 95 96. In one virus like particle system, the increased immunogenicity was due to enhanced DC co-stimulatory capacity, and not antigen presentation 97. Evidence suggests that innate immune activating substances should directly or indirectly activate the type I IFN receptor, which enhances both cross-presenting DC-co-stimulation and T cell activation 98. Combining two approaches, it would be interesting to target particulate immunogens to optimal human DC subsets by coupling antigen containing particles to subset-specific ligands (e.g. anti-CLEC9 antibodies).

Conclusion

Practical people, like vaccinologists and Presidents (“Give me a one armed economist”… attributed to Harry Truman, weary of receiving “on the other hand..” answers) prefer clear advice. The immune system, however, is even more complicated than the economy, and more insight into the workings of the immune system is necessary for vaccinology to become rocket science, and for antigen processingologists to shed an arm.

Fortunately, a combination of successes (T cell cancer immunotherapy 89, 99) and failures (HIV vaccines) provides a strong carrot and stick argument for robust funding of basic research into antigen presentation and T cell activation. Perhaps the words of Churchill best describe the current state of affairs regarding rational vaccine design.

Now this is not the end. It is not even the beginning of the end. But it is, perhaps, the end of the beginning.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Teng MW, et al. Immune-mediated dormancy: an equilibrium with cancer. J Leukoc Biol. 2008;84:988–993. doi: 10.1189/jlb.1107774. [DOI] [PubMed] [Google Scholar]
  • 2.Crowe SR, et al. Vaccination with an acidic polymerase epitope of influenza virus elicits a potent antiviral T cell response but delayed clearance of an influenza virus challenge. J Immunol. 2005;174:696–701. doi: 10.4049/jimmunol.174.2.696. [DOI] [PubMed] [Google Scholar]
  • 3.Yang OO, et al. Impacts of avidity and specificity on the antiviral efficiency of HIV-1-specific CTL. J Immunol. 2003;171:3718–3724. doi: 10.4049/jimmunol.171.7.3718. [DOI] [PubMed] [Google Scholar]
  • 4.Kastenmuller W, et al. Cross-competition of CD8+ T cells shapes the immunodominance hierarchy during boost vaccination. The Journal of experimental medicine. 2007;204:2187–2198. doi: 10.1084/jem.20070489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Oseroff C, et al. HLA class I-restricted responses to vaccinia recognize a broad array of proteins mainly involved in virulence and viral gene regulation. Proc Natl Acad Sci U S A. 2005;102:13980–13985. doi: 10.1073/pnas.0506768102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Hickman HD, et al. Caught in the act: intravital multiphoton microscopy of host-pathogen interactions. Cell Host Microbe. 2009;5:13–21. doi: 10.1016/j.chom.2008.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Chen W, et al. Cross-priming of CD8+ T cells by viral and tumor antigens is a robust phenomenon. Eur J Immunol. 2004;34:194–199. doi: 10.1002/eji.200324257. [DOI] [PubMed] [Google Scholar]
  • 8.Holtappels R, et al. The efficacy of antigen processing is critical for protection against cytomegalovirus disease in the presence of viral immune evasion proteins. Journal of Virology. 2009;83:9611–9615. [Google Scholar]
  • 9.Munks MW, et al. Viral interference with antigen presentation does not alter acute or chronic CD8 T cell immunodominance in murine cytomegalovirus infection. J Immunol. 2007;178:7235–7241. doi: 10.4049/jimmunol.178.11.7235. [DOI] [PubMed] [Google Scholar]
  • 10.Bohm V, et al. The immune evasion paradox: immunoevasins of murine cytomegalovirus enhance priming of CD8 T cells by preventing negative feedback regulation. J Virol. 2008;82:11637–11650. doi: 10.1128/JVI.01510-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Zinkernagel RM. On cross-priming of MHC class I-specific CTL: rule or exception? Eur J Immunol. 2002;32:2385–2392. doi: 10.1002/1521-4141(200209)32:9<2385::AID-IMMU2385>3.0.CO;2-V. [DOI] [PubMed] [Google Scholar]
  • 12.Freigang S, et al. A lymphocytic choriomeningitis virus glycoprotein variant that is retained in the endoplasmic reticulum efficiently cross-primes CD8(+) T cell responses. Proc Natl Acad Sci U S A. 2007;104:13426–13431. doi: 10.1073/pnas.0704423104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • ••13.Kemball CC, et al. Enumeration and functional evaluation of virus-specific CD4+ and CD8+ T cells in lymphoid and peripheral sites of coxsackievirus B3 infection. J Virol. 2008;82:4331–4342. doi: 10.1128/JVI.02639-07. [DOI] [PMC free article] [PubMed] [Google Scholar]; Demonstration that a virus can replicate to astounding levels and fail to induce a detectable primary T cell response. Raises important questions about relevance of cross-priming of viral antigens. Findings should be expanded to defined metabolically stable antigens better suited for cross-priming.
  • 14.Kemball CC, et al. Coxsackievirus B3 Inhibits Antigen Presentation In Vivo, Exerting a Profound and Selective Effect on the MHC Class I Pathway. PLoS pathogens. 2009;5:17. doi: 10.1371/journal.ppat.1000618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Deitz SB, et al. MHC I-dependent antigen presentation is inhibited by poliovirus protein 3A. Proc Natl Acad Sci U S A. 2000;97:13790–13795. doi: 10.1073/pnas.250483097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Lizee G, et al. Control of dendritic cell cross-presentation by the major histocompatibility complex class I cytoplasmic domain. Nature Immunology. 2003;4:1065–1073. doi: 10.1038/ni989. [DOI] [PubMed] [Google Scholar]
  • 17.Wilson NS, et al. Systemic activation of dendritic cells by Toll-like receptor ligands or malaria infection impairs cross-presentation and antiviral immunity. Nat Immunol. 2006;7:165–172. doi: 10.1038/ni1300. [DOI] [PubMed] [Google Scholar]
  • ••18.Gasteiger G, et al. Cross-priming of cytotoxic T cells dictates antigen requisites for modified vaccinia virus Ankara vector vaccines. J Virol. 2007;81:11925–11936. doi: 10.1128/JVI.00903-07. [DOI] [PMC free article] [PubMed] [Google Scholar]; In contrast to reference 13, provides strong evidence for the relevance of cross-priming of anti-viral CTLs. Important clinical implications for a common vaccine vector platfor
  • •19.Lin ML, et al. Selective suicide of cross-presenting CD8+ dendritic cells by cytochrome c injection shows functional heterogeneity within this subset. Proc Natl Acad Sci USA. 2008;105:3029–3034. doi: 10.1073/pnas.0712394105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • •20.Hildner K, et al. Batf3 deficiency reveals a critical role for CD8alpha+ dendritic cells in cytotoxic T cell immunity. Science (New York, N Y. 2008;322:1097–1100. doi: 10.1126/science.1164206. [DOI] [PMC free article] [PubMed] [Google Scholar]; With reference 19, describes tools likely to be useful in dissecting in vivo priming.
  • 21.Heath WR, Carbone FR. Dendritic cell subsets in primary and secondary T cell responses at body surfaces. Nat Immunol. 2009;10:1237–1244. doi: 10.1038/ni.1822. [DOI] [PubMed] [Google Scholar]
  • 22.Accapezzato D, et al. Chloroquine enhances human CD8(+) T cell responses against soluble antigens in vivo. Journal of Experimental Medicine. 2005;202:817–828. doi: 10.1084/jem.20051106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Garulli B, et al. Primary CD8+ T-cell response to soluble ovalbumin is improved by chloroquine treatment in vivo. Clin Vaccine Immunol. 2008;15:1497–1504. doi: 10.1128/CVI.00166-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Basler M, et al. The proteasome inhibitor bortezomib enhances the susceptibility to viral infection. J Immunol. 2009;183:6145–6150. doi: 10.4049/jimmunol.0901596. [DOI] [PubMed] [Google Scholar]
  • 25.Saveanu L, et al. IRAP identifies an endosomal compartment required for MHC class I cross-presentation. Science (New York, N Y. 2009;325:213–217. doi: 10.1126/science.1172845. [DOI] [PubMed] [Google Scholar]
  • 26.Yan J, et al. In vivo role of ER-associated peptidase activity in tailoring peptides for presentation by MHC class Ia and class Ib molecules. The Journal of experimental medicine. 2006;203:647–659. doi: 10.1084/jem.20052271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.York IA, et al. Endoplasmic reticulum aminopeptidase 1 (ERAP1) trims MHC class I-presented peptides in vivo and plays an important role in immunodominance. Proceedings of the National Academy of Sciences of the United States of America. 2006;103:9202–9207. doi: 10.1073/pnas.0603095103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Hammer GE, et al. In the absence of aminopeptidase ERAAP, MHC class I molecules present many unstable and highly immunogenic peptides. Nature Immunology. 2007;8:101–108. doi: 10.1038/ni1409. [DOI] [PubMed] [Google Scholar]
  • 29.Day PM, et al. Effect of TAP on the generation and intracellular trafficking of peptide-receptive major histocompatibility complex class I molecules. Immunity. 1995;2:137–147. doi: 10.1016/s1074-7613(95)80014-x. [DOI] [PubMed] [Google Scholar]
  • 30.Song R, et al. Peptide-receptive class I major histocompatibility complex molecules on TAP-deficient and wild-type cells and their roles in the processing of exogenous antigens. Immunology. 1999;97:316–324. doi: 10.1046/j.1365-2567.1999.00759.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Norbury CC, et al. CD8+ T cell cross-priming via transfer of proteasome substrates. Science (New York, N Y. 2004;304:1318–1321. doi: 10.1126/science.1096378. [DOI] [PubMed] [Google Scholar]
  • 32.Shen LJ, Rock KL. Cellular protein is the source of cross-priming antigen in vivo. Proceedings of the National Academy of Sciences of the United States of America. 2004;101:3035–3040. doi: 10.1073/pnas.0308345101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Wolkers MC, et al. Antigen bias in T cell cross-priming. Science (New York, N Y. 2004;304:1314–1317. doi: 10.1126/science.1096268. [DOI] [PubMed] [Google Scholar]
  • 34.Murshid A, et al. Heat-shock proteins in cancer vaccines: agents of antigen cross-presentation. Expert review of vaccines. 2008;7:1019–1030. doi: 10.1586/14760584.7.7.1019. [DOI] [PubMed] [Google Scholar]
  • 35.Basta S, et al. Cross-presentation of the long-lived lymphocytic choriomeningitis virus nucleoprotein does not require neosynthesis and is enhanced via heat shock proteins. J Immunol. 2005;175:796–805. doi: 10.4049/jimmunol.175.2.796. [DOI] [PubMed] [Google Scholar]
  • 36.Fluet ME, et al. Effects of rapid antigen degradation and VEE glycoprotein specificity on immune responses induced by a VEE replicon vaccine. Virology. 2008;370:22–32. doi: 10.1016/j.virol.2007.08.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Bins AD, et al. In Vivo Antigen Stability Affects DNA Vaccine Immunogenicity. J Immunol. 2007;179:2126–2133. doi: 10.4049/jimmunol.179.4.2126. [DOI] [PubMed] [Google Scholar]
  • 38.Donohue KB, et al. Cross-priming utilizes antigen not available to the direct presentation pathway. Immunology. 2006;119:63–73. doi: 10.1111/j.1365-2567.2006.02406.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • •39.Lev A, et al. The exception that reinforces the rule: crosspriming by cytosolic peptides that escape degradation. Immunity. 2008;28:787–798. doi: 10.1016/j.immuni.2008.04.015. [DOI] [PMC free article] [PubMed] [Google Scholar]; Demonstrates that poor cross-priming activity of biosynthesized peptides is due to their rapid degradation, and not intrinsic failure to access cross-priming pathway. Also shows that cells spare some peptides from rapid degradation, suggesting a biological function for select oligopeptides.
  • 40.Oizumi S, et al. Molecular and cellular requirements for enhanced antigen cross-presentation to CD8 cytotoxic T lymphocytes. J Immunol. 2007;179:2310–2317. doi: 10.4049/jimmunol.179.4.2310. [DOI] [PubMed] [Google Scholar]
  • 41.Nicchitta CV. Re-evaluating the role of heat-shock protein-peptide interactions in tumour immunity. Nat Rev Immunol. 2003;3:427–432. doi: 10.1038/nri1089. [DOI] [PubMed] [Google Scholar]
  • 42.Marincek BC, et al. Heat shock protein-antigen fusions lose their enhanced immunostimulatory capacity after endotoxin depletion. Mol Immunol. 2008;46:181–191. doi: 10.1016/j.molimm.2008.07.039. [DOI] [PubMed] [Google Scholar]
  • 43.Bendz H, et al. Human heat shock protein 70 enhances tumor antigen presentation through complex formation and intracellular antigen delivery without innate immune signaling. J Biol Chem. 2007;282:31688–31702. doi: 10.1074/jbc.M704129200. [DOI] [PubMed] [Google Scholar]
  • 44.Ye Z, Gan YH. Flagellin contamination of recombinant heat shock protein 70 is responsible for its activity on T cells. J Biol Chem. 2007;282:4479–4484. doi: 10.1074/jbc.M606802200. [DOI] [PubMed] [Google Scholar]
  • 45.Kunisawa J, Shastri N. Hsp90 alpha chaperones large C-terminally extended proteolytic intermediates in the MHC class I antigen processing pathway. Immunity. 2006;24:523–534. doi: 10.1016/j.immuni.2006.03.015. [DOI] [PubMed] [Google Scholar]
  • 46.Bleifuss E, et al. Differential capacity of chaperone-rich lysates in cross-presenting human endogenous and exogenous melanoma differentiation antigens. International Journal of Hyperthermia. 2008;24:623–637. doi: 10.1080/02656730802213384. [DOI] [PubMed] [Google Scholar]
  • 47.Moroi Y, et al. Induction of cellular immunity by immunization with novel hybrid peptides complexed to heat shock protein 70. Proc Natl Acad Sci U S A. 2000;97:3485–3490. doi: 10.1073/pnas.070550797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Castellino F, et al. Receptor-mediated uptake of antigen/heat shock protein complexes results in major histocompatibility complex class I antigen presentation via two distinct processing pathways. The Journal of experimental medicine. 2000;191:1957–1964. doi: 10.1084/jem.191.11.1957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Harshyne LA, et al. A Role for Class A Scavenger Receptor in Dendritic Cell Nibbling from Live Cells. J Immunol. 2003;170:2302–2309. doi: 10.4049/jimmunol.170.5.2302. [DOI] [PubMed] [Google Scholar]
  • 50.Tewalt EF, et al. Viral sequestration of antigen subverts cross presentation to CD8(+) T cells. PLoS pathogens. 2009;5:e1000457. doi: 10.1371/journal.ppat.1000457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Kepp O, et al. Immunogenic cell death modalities and their impact on cancer treatment. Apoptosis. 2009;14:364–375. doi: 10.1007/s10495-008-0303-9. [DOI] [PubMed] [Google Scholar]
  • 52.Plesa G, et al. Immunogenicity of cytopathic and noncytopathic viral vectors. J Virol. 2006;80:6259–6266. doi: 10.1128/JVI.00084-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • ••53.Sancho D, et al. Identification of a dendritic cell receptor that couples sensing of necrosis to immunity. Nature. 2009;458:899–903. doi: 10.1038/nature07750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • ••54.Caminschi I, et al. The dendritic cell subtype-restricted C-type lectin Clec9A is a target for vaccine enhancement. Blood. 2008;112:3264–3273. doi: 10.1182/blood-2008-05-155176. [DOI] [PMC free article] [PubMed] [Google Scholar]; Provides critical insight into the special cross-priming capacity of CD8αα and along with ref 53 provides a potential strategy for highly efficient cross-priming in humans.
  • 55.Joly E, Hudrisier D. What is trogocytosis and what is its purpose? Nat Immunol. 2003;4:815. doi: 10.1038/ni0903-815. [DOI] [PubMed] [Google Scholar]
  • 56.Yewdell JW, Haeryfar SM. Understanding Presentation of Viral Antigens to CD8(+) T Cells In Vivo: The Key to Rational Vaccine Design * 1. Annu Rev Immunol. 2005;23:651–682. doi: 10.1146/annurev.immunol.23.021704.115702. [DOI] [PubMed] [Google Scholar]
  • 57.Smith AL, Fazekas De St GB. Antigen-pulsed CD8alpha+ dendritic cells generate an immune response after subcutaneous injection without homing to the draining lymph node. Journal of Experimental Medicine. 1999;189:593–598. doi: 10.1084/jem.189.3.593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Dolan BP, et al. Dendritic cells cross-dressed with peptide MHC class I complexes prime CD8+ T cells. J Immunol. 2006;177:6018–6024. doi: 10.4049/jimmunol.177.9.6018. [DOI] [PubMed] [Google Scholar]
  • 59.Qu C, et al. MHC class I/peptide transfer between dendritic cells overcomes poor cross-presentation by monocyte-derived APCs that engulf dying cells. J Immunol. 2009;182:3650–3659. doi: 10.4049/jimmunol.0801532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Chaudhri G, et al. T cell receptor sharing by cytotoxic T lymphocytes facilitates efficient virus control. Proc Natl Acad Sci U S A. 2009;106:14984–14989. doi: 10.1073/pnas.0906554106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Neijssen J, et al. Cross-presentation by intercellular peptide transfer through gap junctions. Nature. 2005;434:83–88. doi: 10.1038/nature03290. [DOI] [PubMed] [Google Scholar]
  • 62.Benlalam H, et al. Gap Junction Communication between Autologous Endothelial and Tumor Cells Induce Cross-Recognition and Elimination by Specific CTL. Journal of Immunology. 2009;182:2654–2664. doi: 10.4049/jimmunol.0800815. [DOI] [PubMed] [Google Scholar]
  • 63.Pang B, et al. Direct antigen presentation and gap junction mediated cross-presentation during apoptosis. J Immunol. 2009;183:1083–1090. doi: 10.4049/jimmunol.0900861. [DOI] [PubMed] [Google Scholar]
  • ••64.Burgdorf S, et al. Spatial and mechanistic separation of cross-presentation and endogenous antigen presentation. Nat Immunol. 2008;9:558–566. doi: 10.1038/ni.1601. [DOI] [PubMed] [Google Scholar]
  • ••65.Burgdorf S, et al. Distinct pathways of antigen uptake and intracellular routing in CD4 and CD8 T cell activation. Science (New York, NY. 2007;316:612–616. doi: 10.1126/science.1137971. [DOI] [PubMed] [Google Scholar]; With ref 64, provides a potential strategy for targeting protein immunogens to highly effective cross-priming DC subsets.
  • 66.Burgdorf S, et al. The mannose receptor mediates uptake of soluble but not of cell-associated antigen for cross-presentation. J Immunol. 2006;176:6770–6776. doi: 10.4049/jimmunol.176.11.6770. [DOI] [PubMed] [Google Scholar]
  • 67.Guermonprez P, et al. ER-phagosome fusion defines an MHC class I cross-presentation compartment in dendritic cells. Nature. 2003;425:397–402. doi: 10.1038/nature01911. [DOI] [PubMed] [Google Scholar]
  • 68.Di Pucchio T, et al. Direct proteasome-independent cross-presentation of viral antigen by plasmacytoid dendritic cells on major histocompatibility complex class I. Nat Immunol. 2008;9:551–557. doi: 10.1038/ni.1602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Chung Y, et al. Anatomic location defines antigen presentation by dendritic cells to T cells in response to intravenous soluble antigens. Eur J Immunol. 2007;37:1453–1462. doi: 10.1002/eji.200636544. [DOI] [PubMed] [Google Scholar]
  • 70.Savina A, et al. The Small GTPase Rac2 Controls Phagosomal Alkalinization and Antigen Crosspresentation Selectively in CD8(+) Dendritic Cells. Immunity. 2009 doi: 10.1016/j.immuni.2009.01.013. [DOI] [PubMed] [Google Scholar]
  • 71.Sancho D, et al. Tumor therapy in mice via antigen targeting to a novel, DC-restricted C-type lectin. J Clin Invest. 2008;118:2098–2110. doi: 10.1172/JCI34584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.GeurtsvanKessel CH, et al. Clearance of influenza virus from the lung depends on migratory langerin+CD11b- but not plasmacytoid dendritic cells. The Journal of experimental medicine. 2008;205:1621–1634. doi: 10.1084/jem.20071365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Bedoui S, et al. Cross-presentation of viral and self antigens by skin-derived CD103+ dendritic cells. Nat Immunol. 2009;10:488–495. doi: 10.1038/ni.1724. [DOI] [PubMed] [Google Scholar]
  • 74.Irina C, et al. Enhancing immune responses by targeting antigen to DC. European Journal of Immunology. 2009;39:931–938. doi: 10.1002/eji.200839035. [DOI] [PubMed] [Google Scholar]
  • 75.Galibert L, et al. Nectin-like Protein 2 Defines a Subset of T-cell Zone Dendritic Cells and Is a Ligand for Class-I-restricted T-cell-associated Molecule. Journal of Biological Chemistry. 2005;280:21955–21964. doi: 10.1074/jbc.M502095200. [DOI] [PubMed] [Google Scholar]
  • 76.Lahoud MH, et al. The C-Type Lectin Clec12A Present on Mouse and Human Dendritic Cells Can Serve as a Target for Antigen Delivery and Enhancement of Antibody Responses. J Immunol. 2009;182:7587–7594. doi: 10.4049/jimmunol.0900464. [DOI] [PubMed] [Google Scholar]
  • 77.Pletneva M, et al. IFN-producing killer dendritic cells are antigen-presenting cells endowed with T-cell cross-priming capacity. Cancer Res. 2009;69:6607–6614. doi: 10.1158/0008-5472.CAN-09-0508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Beauvillain C, et al. Neutrophils efficiently cross-prime naive T cells in vivo. Blood. 2007;110:2965–2973. doi: 10.1182/blood-2006-12-063826. [DOI] [PubMed] [Google Scholar]
  • 79.Lund FE, et al. Lymphotoxin-{alpha}-Deficient Mice Make Delayed, But Effective, T and B Cell Responses to Influenza. J Immunol. 2002;169:5236–5243. doi: 10.4049/jimmunol.169.9.5236. [DOI] [PubMed] [Google Scholar]
  • •80.Hickman HD, et al. Direct priming of antiviral CD8+ T cells in the peripheral interfollicular region of lymph nodes. Nat Immunol. 2008;9:155–165. doi: 10.1038/ni1557. [DOI] [PubMed] [Google Scholar]
  • •81.Junt T, et al. Subcapsular sinus macrophages in lymph nodes clear lymph-borne viruses and present them to antiviral B cells. Nature. 2007;450:110–114. doi: 10.1038/nature06287. [DOI] [PubMed] [Google Scholar]; Along with ref. 80, demonstrates the importance of intravital microscopy to understanding activation of T cell responses to vaccines.
  • 82.Fu TM, et al. An endoplasmic reticulum-targeting signal sequence enhances the immunogenicity of an immunorecessive simian virus 40 large T antigen cytotoxic T-lymphocyte epitope. J Virol. 1998;72:1469–1481. doi: 10.1128/jvi.72.2.1469-1481.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Yewdell JW. Plumbing the sources of endogenous MHC class I peptide ligands. Curr Opin Immunol. 2007;19:79–86. doi: 10.1016/j.coi.2006.11.010. [DOI] [PubMed] [Google Scholar]
  • 84.Wei CH, Sherman LA. N-Terminal Trimer Extension of Nominal CD8 T Cell Epitopes Is Sufficient to Promote Cross-Presentation to Cognate CD8 T Cells In Vivo. J Immunol. 2007;179:8280–8286. doi: 10.4049/jimmunol.179.12.8280. [DOI] [PubMed] [Google Scholar]
  • 85.van Montfoort N, et al. Antigen storage compartments in mature dendritic cells facilitate prolonged cytotoxic T lymphocyte cross-priming capacity. Proc Natl Acad Sci USA. 2009;106:6730–6735. doi: 10.1073/pnas.0900969106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Faure F, et al. Long-lasting cross-presentation of tumor antigen in human DC. Eur J Immunol. 2009;39:380–390. doi: 10.1002/eji.200838669. [DOI] [PubMed] [Google Scholar]
  • 87.Bijker MS, et al. Superior induction of anti-tumor CTL immunity by extended peptide vaccines involves prolonged, DC-focused antigen presentation. Eur J Immunol. 2008;38:1033–1042. doi: 10.1002/eji.200737995. [DOI] [PubMed] [Google Scholar]
  • 88.Welters MJP, et al. Induction of Tumor-Specific CD4+ and CD8+ T-Cell Immunity in Cervical Cancer Patients by a Human Papillomavirus Type 16 E6 and E7 Long Peptides Vaccine. Clinical Cancer Research. 2008;14:178–187. doi: 10.1158/1078-0432.CCR-07-1880. [DOI] [PubMed] [Google Scholar]
  • ••89.Kenter GG, et al. Vaccination against HPV-16 Oncoproteins for Vulvar Intraepithelial Neoplasia. The New England journal of medicine. 2009;361:1838–1847. doi: 10.1056/NEJMoa0810097. [DOI] [PubMed] [Google Scholar]; Potential breakthrough study that demonstrates the therapeutic potential of cross-priming vaccines.
  • 90.Vogt A, et al. Transcutaneous anti-influenza vaccination promotes both CD4 and CD8 T cell immune responses in humans. J Immunol. 2008;180:1482–1489. doi: 10.4049/jimmunol.180.3.1482. [DOI] [PubMed] [Google Scholar]
  • 91.Tran KK, Shen H. The role of phagosomal pH on the size-dependent efficiency of cross-presentation by dendritic cells. Biomaterials. 2009;30:1356–1362. doi: 10.1016/j.biomaterials.2008.11.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Hu Y, et al. Cytosolic delivery mediated via electrostatic surface binding of protein, virus, or siRNA cargos to pH-responsive core-shell gel particles. Biomacromolecules. 2009;10:756–765. doi: 10.1021/bm801199z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Shen H, et al. Enhanced and prolonged cross-presentation following endosomal escape of exogenous antigens encapsulated in biodegradable nanoparticles. Immunology. 2006;117:78–88. doi: 10.1111/j.1365-2567.2005.02268.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Jennings GT, Bachmann MF. The coming of age of virus-like particle vaccines. Biol Chem. 2008;389:521–536. doi: 10.1515/bc.2008.064. [DOI] [PubMed] [Google Scholar]
  • 95.Heit A, et al. Antigen co-encapsulated with adjuvants efficiently drive protective T cell immunity. Eur J Immunol. 2007;37:2063–2074. doi: 10.1002/eji.200737169. [DOI] [PubMed] [Google Scholar]
  • 96.Schlosser E, et al. TLR ligands and antigen need to be coencapsulated into the same biodegradable microsphere for the generation of potent cytotoxic T lymphocyte responses. Vaccine. 2008;26:1626–1637. doi: 10.1016/j.vaccine.2008.01.030. [DOI] [PubMed] [Google Scholar]
  • 97.Susanne AK, et al. Innate signaling regulates cross-priming at the level of DC licensing and not antigen presentation. European Journal of Immunology. 40:103–112. doi: 10.1002/eji.200939559. [DOI] [PubMed] [Google Scholar]
  • 98.Le Bon A, Tough DF. Type I interferon as a stimulus for cross-priming. Cytokine Growth Factor Rev. 2008;19:33–40. doi: 10.1016/j.cytogfr.2007.10.007. [DOI] [PubMed] [Google Scholar]
  • 99.Gattinoni L, et al. Adoptive immunotherapy for cancer: building on success. Nat Rev Immunol. 2006;6:383–393. doi: 10.1038/nri1842. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES