548
Views
5
CrossRef citations to date
0
Altmetric
Commentary & View

Cell-based immunotherapy of prion diseases by adoptive transfer of antigen-loaded dendritic cells or antigen-primed CD4+ T lymphocytes

&
Pages 66-71 | Received 27 Apr 2010, Accepted 04 Jun 2010, Published online: 01 Apr 2010

Abstract

Prion diseases are neurodegenerative conditions caused by the transconformation of a normal host glycoprotein, the cellular prion protein (PrPc) into a neurotoxic, self-aggregating conformer (PrPSc). TSEs are ineluctably fatal and no treatment is yet available. In principle, prion diseases could be attacked from different angles including: blocking conversion of PrPc into PrPSc, accelerating the clearance of amyloid deposits in peripheral tissues and brain, stopping prion progression in secondary lymphoid organs, reducing brain inflammation and promoting neuronal healing. There are many indications that adaptive and innate immunity might mediate those effects but so far, the achievements of immunointervention have not matched all expectations. Difficulties arise from the impossibility to diagnose TSE before substantial brain damage, poor accessibility of the CNS to immunological agents, deep immune tolerance to self-PrP, and short term effects of many immune interventions contrasting with the slow progression of TSEs. Here, we discuss two approaches, inspired from cancer immunotherapy, which might overcome some of those obstacles. One is vaccination with antigen-pulsed or antigen-transduced dendritic cells to bypass self-tolerance. The other one is the adoptive transfer of PrP-sensitized CD4+ T cells which can promote humoral, cell-mediated or regulatory responses, coordinate adaptive and innate immunity and have long lasting effects.

The Present Situation

The demonstration, ten years ago, that antibodies could block the propagation of prion infection in vitro, in infected cell linesCitation1,Citation2 and in vivo in scrapie-bearing miceCitation3Citation5 generated great enthusiasm. Parallel results obtained against Alzheimer disease (AD)Citation6 or Parkinson diseaseCitation7 further supported the idea that vaccinal strategies which had been historically so successful against classical infectious diseases could be similarly applied to neurodegenerative conditions. Ten years later, in spite of numerous encouraging reports, there is some questioning regarding the possibility to propose efficient and safe immunotherapy against neurodegenerative proteinopathies, notably against transmissible spongiform encephalopathies (TSEs). The obstacles have been well identified and extensively reviewed.Citation8,Citation9 Self-tolerance to PrPc is the main concern. Due to the strong tolerogenicity of PrP, the T-cell and B-cell repertoires which escape tolerance and are available for vaccination appear to be considerably impaired, both quantitatively and qualitatively. Immune selection operates evidently against the lymphocytes that are the most self-reactive, which means those which recognize PrP epitopes with high specificity and affinity and which would be the most effective therapeutically. This explains why it is difficult to raise robust vaccinal responses against PrP and to achieve solid and durable protection. In most trials of active immunization, the attack rate is unchanged and life extension remains modest. As already mentioned, passive transfer of antibodies generated in PrP-deficient mice was the starting point of immunotherapy in TSEs. Efforts have been pursued since then, but have encountered limited success due to the difficulty to make antibodies cross the meningial barrier and penetrate into the brain at a stage when prions have already invaded the nervous tissue. Therefore, the treatment is effective only if started concomitantly with peripheral infection or very shortly after. Antibodies used in those assays were directed against PrPc and selected for their epitope specificityCitation10 or their pharmacological qualities.Citation11 In order to overcome the problem of brain accessibility, antibodies were also administered through intra-ventricular infusion with an osmotic pump, resulting in onset delay and disease attenuation with some prion strains even when treatment was initiated a few weeks after inoculation.Citation12 Antibodies specific for the 37/67 kDa laminin receptor which acts as a cell surface receptor for prions were also used with definite success.Citation13 To overcome the problem of blood brain barrier crossing, some groups have engineered single chain Fv antibodies (scFv) which penetrate more easily into the brain owing to their lower molecular weight.Citation14,Citation15 The latest development in this field has been the insertion of scFv antibody genes into specific viral vectors such as adeno-associated viruses in order to achieve antibody delivery within the CNS.Citation16Citation18 Here again, scrapie is slowed down, even after direct prion inoculation into the brain, which constitutes a definite progress, but ultimately all mice succumb to the neurological lesions.

In order to translate immunotherapy into clinic, strategies must be therefore substantially improved. Improvement may come from different directions. A first one should be to strengthen the response of the available anti-PrP repertoire. Ingenious procedures of immunization have been developed including the use of potent adjuvantsCitation19 and of highly immunogenic bacterial and viral vectors encoding the Prnp gene sequence,Citation20Citation22 but there is probably still room for progress. Second, one should consider other protective pathways, beside antibodies, notably when the disease is fully established in the brain. For instance it might be more pertinent to skew the immune response toward a cell-mediated profile which would associate activated CD4+ T cells with recruited macrophages and microglial cells rather than aiming at getting a humoral response which hardly reaches the CNS. Third, it may be necessary to supply the failing immune system with lymphocytes enriched for more avid anti-PrP T-cell receptors (TCR) and secreting chemokines and cytokines of interest. This could be done by selecting and expanding lymphocytes in vitro before reinfusing them into the host or even by engineering the expression of new antigen receptors. Here, we describe two cell-based strategies which have been developed with success in cancer immunotherapy and which respond to the criteria suggested above. One is the use of antigen-pulsed dendritic cells (DCs) for vaccination, a strategy which should potentiate the immune response generated by the available immune repertoire, with the possibility to reorient responses to a desired mode. The other is the adoptive transfer of PrP-primed T helper cells which could serve as a study model to understand what type of response would be most effective at a given stage of disease evolution. It could also be a clinical alternative to adoptive antibody therapy by providing the host immune system with relatively long-lived, good quality memory effector cells. The two strategies will be described and discussed in the light of recent results obtained in our laboratory.

Vaccination with Antigen Loaded DCs

The process of antigen presentation to T lymphocytes is absolutely central to the genesis of immune responses. DCs, macrophages and B lymphocytes are the main professional antigen presenting cells (APCs). Among them, DCs have the unique capacity to activate naive T cells, thereby initiating primary immune responses.Citation23,Citation24 Optimal T-lymphocyte activation depends on a series of signals. Signal 1 is antigen-specific; it is delivered through the interaction between the antigen-MHC complex on the APC and the TCR. Signal 2 is a costimulatory signal delivered by APCs and aimed at amplifying signal 1 which is insufficient by itself to activate T lymphocytes and, if not complemented by signal 2, leads to an aborted response. Because DCs express high quantities of MHC class I and class II products together with a wide range of costimulatory molecules such as CD86, CD80 and CD40, they are the most efficient APCs to provide signals 1 and 2 to T lymphocytes.Citation25 In addition, DCs secrete a panel of cytokines (signal 3) which skew the differentiation of CD4+ T cells toward distinct lineages.Citation26 Naive T helper cells (Th0) may differentiate into T helper type 1 (Th1) T cells which produce proinflammatory cytokines, mediate cellular immune responses, cooperate with CD8+ T cells for cytotoxic responses and mobilize effectors of innate immunity. Alternatively, Th0 cells may differentiate into Th2 T cells which cooperate with B cells for antibody production. They may also differentiate into Th17 T cells which induce strong inflammatory responses or into regulatory T cells (Treg) whose function is to prevent deleterious autoimmunity. The choice of lineage depends essentially on the presenting DC, which integrates, via a wide range of receptors, an ensemble of physical, chemical and biological messages including the presence and the origin of a pathogen or the distress of surrounding tissues.Citation27Citation29 DC precursors migrate from bone marrow to peripheral tissues where they capture, process and present at their cell surface antigenic peptides associated to MHC class I or II molecules. DCs mature and subsequently migrate to the T cell zones of secondary lymphoid organs where they come into contact with antigen-specific T lymphocytes and initiate their activation and lineage commitment.Citation30

DCs play a major role in vaccination. In classical vaccination designs, DCs are randomly targeted in vivo by the antigen usually injected with an adjuvant.Citation31 However, in more recent protocols, DCs can be specifically targeted in vitro. DCs are differentiated and expanded from bone-marrow, peripheral blood or cord blood precursors.Citation32Citation34 They are subsequently matured and loaded with the antigen of interest and reinfused into the original donor. Specific targeting avoids the random delivery of antigen to irrelevant DC or to APC populations which might anergize rather than activate T cells or skew them toward an undesirable type of response, for instance pro-inflammatory Th17 T cells instead of Th2 T cells. By targeting DCs in vitro, the antigen under the right formulation is delivered only to the relevant APCs appropriately matured.Citation28 Vaccination with targeted DCs has been so far mainly developed against cancer. In a majority of instances, the objective was to raise CD8+ cytotoxic T cells killing tumor cells expressing tumor specific antigens in a MHC class-I context.Citation35 Several clinical studies have shown encouraging results against diverse types of cancers including melanoma, renal carcinomaCitation36,Citation37 and colon or lung cancer.Citation38 More recently, some laboratories have succeeded in using DC vaccination in order to generate MHC class-II restricted CD4+ T-cell responses against tumors. Those studies underlined the necessity for the CD4+ T cells to cooperate with CD8+ T cells for an efficient eradication of tumor cells, both in mice and humans.Citation39Citation41 But more recently, it was shown that CD4+ T cells can eliminate melanoma cells on their own, in the absence of CD8+ T or of natural killer (NK) cells.Citation42

Those encouraging results have prompted us to transpose DC vaccination to prion diseases. Two MHC class-II restricted 30-mer peptides of PrP (PrP98–127 and PrP158–187) had been previously identified as potent stimulators of CD4+ T cells when directly injected with adjuvant into PrP-deficient (Prnp/) mice, but not into wt mice naturally tolerant to PrP.Citation43 We first asked whether DCs pulsed with these peptides would overcome tolerance in wt mice. The answer was positive. Mice twice injected with peptide-pulsed DCs develop CD4+ T cells which proliferate and produce cytokines (IL-4 and IFNγ) in an antigen-dependant way.Citation44 Of note, consistent with previous observations made in Prnp/ mice, DCs loaded with PrP158-187 elicit a Th1 and Th2 response with production of IFNγ and of IL-4 whereas the response elicited by DCs loaded with PrP98–127 is exclusively centered on IL-4, suggesting that the choice of the peptide may determine the type of immune response. Antibodies recognizing epitopes expressed on native PrPc are detected in parallel. A detailed analysis of the interactions between the three partners—DCs, T cells and B cells—shows that DCs make direct and independent contacts with the T and the B cell partners. T cells recognize peptide/MHC class-II complexes resulting from the processing of the loaded peptides whereas B cells recognize conformational epitopes of PrPc expressed on the DC cell surface. There is no overlap between the T and B epitopes and both peptide and membrane PrPc are necessary to achieve full cooperation.

Next, we inoculated i.p., a scrapie agent (strain 139A), into mice which had been twice challenged with PrP98–127-pulsed DC and were subsequently boosted every month. Two out of ten mice never became sick and showed no PrPSc in their brain. The eight other mice developed scrapie, but with a longer incubation and longer survival than controls.Citation44 The total disease duration was extended by 20% whereas it was only around 10% in most reported studies. Interestingly, the duration of the prolongation was correlated with the intensity of the antibody response measured 45 days after infection. The two mice which were definitively protected displayed the highest antibody titers.

The advantage of using DC as a vaccine support was also demonstrated in a recent study performed in the laboratory by Rosset and colleagues.Citation45 There, instead of being pulsed with antigen, DCs were transduced with a recombinant adenovirus. Mice which had received several boosts of antigenic DCs were subsequently infected with 139A. The best protection was achieved with DCs transduced with a recombinant adenovirus encoding the sequence of human PrP. The conferred protection was considerably higher than when similar recombinant adenoviruses were directly injected in vivo. Both peptide-pulsed and virally-transduced DCs have their advantages and drawbacks. Loading DCs with pure and well-defined synthetic peptides is simple and safe, but it requires the identification of the PrP peptide for a given MHC class II context. Viral transduction of DCs with a full length PrP gene requires an additional step of virus production and might be therefore less safe or less easy to perform, but the peptide sampling and insertion onto MHC class-II products is done by the DC.

Thus, the use of in vitro matured and targeted DCs as a vaccine vector presents definite advantages over direct in vivo delivery of antigen. DCs evoke good T-and B-cell responses including antibodies against native epitopes of PrPc which have the capacity to prevent PrP conversion and hence to block prion expansion. Incidentally, this result shows that B-cell precursors of potential therapeutic interest are not totally absent from the repertoire which has gone through tolerance selection. Even though the B cells which escaped tolerance are probably rare and of mediocre avidity, they may have a chance to expand by antigen stimulation and to improve antibody affinity by somatic mutations of their immunoglobulin genes.

Adoptive T-Cell Transfer

CD4+ T lymphocytes play a key role in adaptive immune responses. They respectively orchestrate cell-mediated, cytotoxic, humoral, inflammatory or suppressive responses through their ability to differentiate into a wide range of functionally different and even opposite lineages, such as Th1, Th2, Th17 or Treg.Citation46Citation48 As already mentioned, the decision of lineage commitment comes in the first place from the DC which tailors the response that will be best adapted to the situation.Citation30 Quite importantly, the decision can be changed either by modifying the conditions under which DCs mature or by reprogramming CD4+ T helper cells.Citation49

CD4+ T cells have been so far neglected in the context of imunotherapy against TSE. One reason is that the main focus has been put on B cells and antibodies. The other is that CD4+ T cells have been seen more as foes than as friends following the complications of cell-mediated meningoencephalitis generated by the vaccination of AD patient with Aβ peptide.Citation50 But the shortcomings of passive antibody transfer which is effective only against the transmitted forms of TSEs and at the early phase of lymphoinvasion, underline the necessity to develop alternative strategies. Adoptive T-cell therapy could be one of them. CD4+ T cells might exert anti-prion effects in different ways. As Th2 T cells, they may help B cells to differentiate into antibody-producing plasmocytes, induce immunoglobulin switch and contribute to the affinity maturation of antibodies. Thus, supplying the host immune system with a cohort of efficient Th2 helper cells might improve the quantity and the quality of secreted therapeutic antibodies. CD4+ T cells with a Th1 or a Treg profile could be also helpful at more advanced stages of disease, and notably against sporadic and familial forms of TSE which start developing in the brain. Via the secretion of pro-inflammatory cytokines and of chemokines, proinflammatory T cells might attract macrophages into the brain and might activate microglial cells which could clear amyloid deposits, degrade clusters of infectious PrPSc and facilitate neuronal repair. Conversely, Treg lymphocytes might soothe brain inflammation by releasing TGFβ or IL-10. The crucial issue here is to identify precisely which type of CD4+ T cells is most effective at a given stage of disease evolution while avoiding putative autoimmune complications.

To begin investigating those questions, we have developed a very basic study model in which the CD4+ T cells come from PrP-deficient mice, not tolerant to PrP and therefore able to generate full-fledged anti-PrP responses and recipients are histocompatible mice expressing PrP and susceptible to scrapie.Citation51 We have first asked how long CD4+ T cells sensitized against PrP can survive and remain functional after engraftment into recipients expressing PrP. The results were highly encouraging as T cells appeared to resist peripheral tolerance; they could still proliferate and secrete cytokines, three months after transfer, provided that they were regularly recalled with antigen. We then asked whether such lymphocytes would protect infected mice. This was done under various conditions, in mice made partially or totally lymphopenic, with cell transfer performed before or just at the time of infection. In all situations, T cells primed and iteratively boosted attenuated disease progression. The attack rate was not reduced as with DC vaccination, but there was a substantial life prolongation. An important point to be underscored is that circulating antibodies against native PrPc were low, actually close to background, in all experimental settings. This means that resident B cells did not readily respond to the help brought by the transferred T cells and that protection may not be necessarily mediated by antibodies.Citation51

Many important issues remain to be investigated. So far the transferred T cells have been directly collected from immunized Prnp/ mice without polarization toward a specific profile. Those T cells are probably a mixture of Th1 and Th2. It will be essential to examine the consequences of transferring pure lineages of Th1 or Th2 T cells as well as Treg and Th17 T cells. Another major issue is to find out the therapeutic window of adoptive T-cell transfer. The time limit for antibody therapy is apparently around 30 days after infection with a mouse adapted prion strain.Citation4 It will be important to see whether T cells can still be beneficial when injected beyond this point, notably at the clinical onset or in mice which have been infected by intracranial route. One should also find out whether the risk of adverse autoimmune reactions does really exist or is a simple inference from what has been reported in AD.

To increase the power of our study model, we have recently generated a TCR transgenic mouse which expresses a high frequency of PrP-specific CD4+ T cells. A first analysis of the mouse phenotype shows that anti-PrP TCRs come out of the thymus and populate the periphery only in Prnp/ mice (manuscript in preparation). Central negative selection contributes therefore significantly to the shaping of the CD4+ PrP-specific T-cell repertoire. The T cells carrying the transgenic TCR are highly responsive to PrP and can be expanded in cell culture without prior in vivo priming. They represent an ideal source of lymphocytes for generating activated effector T cells with a polarized profile and for examining the consequences of their adoptive transfer into healthy or infected recipients. Adoptive transfer of T cells from immunized Prnp/ donors into PrP-positive recipients is clearly not transposable to clinical situations, but it is a useful model for answering important methodological questions.

The protocols for TSE patients should be modeled on those currently developed in cancer immunotherapy.Citation52,Citation53 Assuming that a substantial number of PrP-specific T helper lymphocytes with TCRs of reasonable affinity survive central and peripheral tolerance, they could be submitted to in vitro antigen-driven selection, polarization and expansion before being reinjected into the patient. If it appears that the number of good affinity T helper cells is not sufficient, one should consider the possibility of transducing T cells either with physiological MHC-restricted TCR genes conferring high avidityCitation54 or alternatively, with non MHC-restricted chimeric antigen receptors (CAR) which express at the cell membrane an antigen receptor constituted by the variable domains of an anti-PrPc antibody.Citation55 The use of CARs would present the advantage of being totally independent of the MHC class-II context of the patient, and therefore not to necessitate the identification of fitting peptides, as would be the case for physiological α/β TCRs.

Perspectives

Neither DC vaccination nor adoptive T-cell therapy can be translated into clinic at the present stage. The attack rate is modestly reduced with DC vaccination and still not with adoptive T-cell transfer. Life extension is in the order of 20% only, which is obviously insufficient. However one can easily foresee how both strategies might be substantially improved. The most urgent point is to identify the type of adaptive immune response: humoral, cell-mediated, inflammatory, inflammatory plus innate or regulatory, which will be best adapted to the state of disease advancement, to the clinical status of the patient, and most probably too, to the prion strain. On the one hand, our results show that the level of antibodies generated by DC vaccination correlates with protection. On the other hand, protection in mice receiving adoptively transferred T cells is not accompanied by the production of antibodies. Thus the question of humoral versus cellmediated immunity is totally open. To identify which mode of response is most efficient might be achieved in a basic study model such as the one that we are currently developing between Prnp/ and wt mice. In a second stage, mice humanized for PrP and for the lymphoid system56 should be used to probe human prions. Once the type of beneficial immune response is clearly defined, either DCs or T cells can be matured, differentiated or reprogrammed so as to skew the immune responses toward the most helpful profile while avoiding harmful autoimmunity.

Beside engineering good affinity TCRs or CARs on the patient T cells, it might be advisable to transduce DCs or CD4+ T helper cells with genes encoding for cytokines, chemokines and chemokine receptors. Such mediators would facilitate the migration of cells to the sites of interest: gut lymphoid system, draining lymph nodes and brain. For DCs, they would potentiate their impact upon T and B cells; for CD4+ T cells, they would enhance their capacity to cross the brain barrier and recruit agents of innate immunity. The experience accumulating in cancer immunotherapy will be most useful for developing rational and effective immunointerventions against TSE. One may as well anticipate that the lessons drawn from prion diseases will benefit other neurodegenerative conditions such as Alzheimer and Parkinson diseases.

Abbreviations

PrPc=

cellular prion protein

PrPSc=

scrapie prion protein

TSE=

transmissible spongiform encephalopathies

CNS=

central nervous system

AD=

Alzheimer disease

TCR=

T-cell receptor

scFv=

single chain antibodies

DCs=

dendritic cells

APC=

professional antigen presenting cell

MHC=

major histocompatibility complex

Th cell=

T helper cell

NK cell=

natural killer cell

IFN=

interferon

IL=

interleukin

T reg=

regulatory T cell

CAR=

chimeric antigen receptors

Acknowledgements

We thank Pauline Gourdain, Saci Iken and Martine Rosset for their inputs into the studies referred in this “Perspective.” The studies were funded by European Union Project No. FOOD-CT-2006-023144, by Groupement d’Intérêt Scientifique “Maladies à Prions,” and by INSERM and Université Pierre et Marie Curie Paris 6. V.B. was supported by a post-doctoral fellowship from INSERM.

References

  • Enari M, Flechsig E, Weissmann C. Scrapie prion protein accumulation by scrapie-infected neuroblastoma cells abrogated by exposure to a prion protein antibody. Proc Natl Acad Sci USA 2001; 98:9295 - 9299
  • Peretz D, Williamson RA, Kaneko K, Vergara J, Leclerc E, Schmitt-Ulms G, et al. Antibodies inhibit prion propagation and clear cell cultures of prion infectivity. Nature 2001; 412:739 - 743
  • Heppner FL, Musahl C, Arrighi I, Klein MA, Rulicke T, Oesch B, et al. Prevention of scrapie pathogenesis by transgenic expression of anti-prion protein antibodies. Science 2001; 294:178 - 182
  • White AR, Enever P, Tayebi M, Mushens R, Linehan J, Brandner S, et al. Monoclonal antibodies inhibit prion replication and delay the development of prion disease. Nature 2003; 422:80 - 83
  • Sigurdsson EM, Sy MS, Li R, Scholtzova H, Kascsak RJ, Kascsak R, et al. Anti-prion antibodies for prophylaxis following prion exposure in mice. Neurosci Lett 2003; 336:185 - 187
  • Schenk D, Barbour R, Dunn W, Gordon G, Grajeda H, Guido T, et al. Immunization with amyloid-beta attenuates Alzheimer-disease-like pathology in the PDAPP mouse. Nature 1999; 400:173 - 177
  • Masliah E, Rockenstein E, Adame A, Alford M, Crews L, Hashimoto M, et al. Effects of alpha-synuclein immunization in a mouse model of Parkinson’s disease. Neuron 2005; 46:857 - 868
  • Aguzzi A, Sigurdson CJ. Antiprion immunotherapy: to suppress or to stimulate?. Nat Rev Immunol 2004; 4:725 - 736
  • Muller-Schiffmann A, Korth C. Vaccine approaches to prevent and treat prion infection: progress and challenges. BioDrugs 2008; 22:45 - 52
  • Sadowski MJ, Pankiewicz J, Prelli F, Scholtzova H, Spinner DS, Kascsak RB, et al. Anti-PrP Mab 6D11 suppresses PrP(Sc) replication in prion infected myeloid precursor line FDC-P1/22L and in the lymphoreticular system in vivo. Neurobiol Dis 2009; 34:267 - 278
  • Feraudet-Tarisse C, Andreoletti O, Morel N, Simon S, Lacroux C, Mathey J, et al. Immunotherapeutic effect of anti-PrP monoclonal antibodies in transmissible spongiform encephalopathy mouse models: pharmacokinetic and pharmacodynamic analysis. J Gen Virol 91:1635 - 1645
  • Song CH, Furuoka H, Kim CL, Ogino M, Suzuki A, Hasebe R, et al. Effect of intraventricular infusion of anti-prion protein monoclonal antibodies on disease progression in prion-infected mice. J Gen Virol 2008; 89:1533 - 1544
  • Zuber C, Mitteregger G, Pace C, Zerr I, Kretzschmar HA, Weiss S. Anti-LRP/LR antibody W3 hampers peripheral PrPSc propagation in scrapie infected mice. Prion 2007; 1:207 - 212
  • Luginbuhl B, Kanyo Z, Jones RM, Fletterick RJ, Prusiner SB, Cohen FE, et al. Directed evolution of an anti-prion protein scFv fragment to an affinity of 1 pM and its structural interpretation. J Mol Biol 2006; 363:75 - 97
  • Padiolleau-Lefevre S, Alexandrenne C, Dkhissi F, Clement G, Essono S, Blache C, et al. Expression and detection strategies for an scFv fragment retaining the same high affinity than Fab and whole antibody: Implications for therapeutic use in prion diseases. Mol Immunol 2007; 44:1888 - 1896
  • Wuertzer CA, Sullivan MA, Qiu X, Federoff HJ. CNS delivery of vectored prion-specific single-chain antibodies delays disease onset. Mol Ther 2008; 16:481 - 486
  • Zuber C, Mitteregger G, Schuhmann N, Rey C, Knackmuss S, Rupprecht W, et al. Delivery of singlechain antibodies (scFvs) directed against the 37/67 kDa laminin receptor into mice via recombinant adeno-associated viral vectors for prion disease gene therapy. J Gen Virol 2008; 89:2055 - 2061
  • Zuber C, Knackmuss S, Rey C, Reusch U, Rottgen P, Frohlich T, et al. Single chain Fv antibodies directed against the 37 kDa/67 kDa laminin receptor as therapeutic tools in prion diseases. Mol Immunol 2008; 45:144 - 1451
  • Rosset MB, Ballerini C, Gregoire S, Metharom P, Carnaud C, Aucouturier P. Breaking immune tolerance to the prion protein using prion protein peptides plus oligodeoxynucleotide-CpG in mice. J Immunol 2004; 172:5168 - 5174
  • Fernandez-Borges N, Brun A, Whitton JL, Parra B, Diaz-San Segundo F, Salguero FJ, et al. DNA vaccination can break immunological tolerance to PrP in wild-type mice and attenuates prion disease after intracerebral challenge. J Virol 2006; 80:9970 - 9976
  • Goni F, Prelli F, Schreiber F, Scholtzova H, Chung E, Kascsak R, et al. High titers of mucosal and systemic anti-PrP antibodies abrogate oral prion infection in mucosal-vaccinated mice. Neuroscience 2008; 153:679 - 686
  • Nikles D, Bach P, Boller K, Merten CA, Montrasio F, Heppner FL, et al. Circumventing tolerance to the prion protein (PrP): vaccination with PrP-displaying retrovirus particles induces humoral immune responses against the native form of cellular PrP. J Virol 2005; 79:4033 - 4042
  • Steinman RM, Nussenzweig MC. Dendritic cells: features and functions. Immunol Rev 1980; 53:127 - 147
  • Steinman RM, Witmer MD. Lymphoid dendritic cells are potent stimulators of the primary mixed leukocyte reaction in mice. Proc Natl Acad Sci USA 1978; 75:5132 - 5136
  • Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature 1998; 392:245 - 252
  • Mosmann TR, Sad S. The expanding universe of T-cell subsets: Th1, Th2 and more. Immunol Today 1996; 17:138 - 146
  • Matzinger P. Tolerance, danger and the extended family. Annu Rev Immunol 1994; 12:991 - 1045
  • Palucka AK, Ueno H, Fay JW, Banchereau J. Taming cancer by inducing immunity via dendritic cells. Immunol Rev 2007; 220:129 - 150
  • Pulendran B, Palucka K, Banchereau J. Sensing pathogens and tuning immune responses. Science 2001; 293:253 - 256
  • Banchereau J, Briere F, Caux C, Davoust J, Lebecque S, Liu YJ, et al. Annu Rev Immunol 2000; 18:767 - 811
  • Tritto E, Mosca F, De Gregorio E. Mechanism of action of licensed vaccine adjuvants. Vaccine 2009; 27:3331 - 4
  • Caux C, Dezutter-Dambuyant C, Schmitt D, Banchereau J. GM-CSF and TNF-alpha cooperate in the generation of dendritic Langerhans cells. Nature 1992; 360:258 - 261
  • Romani N, Gruner S, Brang D, Kampgen E, Lenz A, Trockenbacher B, et al. Proliferating dendritic cell progenitors in human blood. J Exp Med 1994; 180:83 - 93
  • Sallusto F, Lanzavecchia A. Efficient presentation of soluble antigen by cultured human dendritic cells is maintained by granulocyte/macrophage colonystimulating factor plus interleukin 4 and downregulated by tumor necrosis factor alpha. J Exp Med 1994; 179:1109 - 1118
  • Rosenberg SA, Packard BS, Aebersold PM, Solomon D, Topalian SL, Toy ST, et al. Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma. A preliminary report. N Engl J Med 1988; 319:1676 - 1680
  • Nestle FO, Alijagic S, Gilliet M, Sun Y, Grabbe S, Dummer R, et al. Vaccination of melanoma patients with peptide- or tumor lysate-pulsed dendritic cells. Nat Med 1998; 4:328 - 332
  • Zitvogel L, Angevin E, Tursz T. Dendritic cell-based immunotherapy of cancer. Ann Oncol 2000; 11:199 - 205
  • Fong L, Hou Y, Rivas A, Benike C, Yuen A, Fisher GA, et al. Altered peptide ligand vaccination with Flt3 ligand expanded dendritic cells for tumor immunotherapy. Proc Natl Acad Sci USA 2001; 98:8809 - 8814
  • Czerniecki BJ, Koski GK, Koldovsky U, Xu S, Cohen PA, Mick R, et al. Targeting HER-2/neu in early breast cancer development using dendritic cells with staged interleukin-12 burst secretion. Cancer Res 2007; 67:1842 - 1852
  • Fujisawa Y, Nabekura T, Nakao T, Nakamura Y, Takahashi T, Kawachi Y, et al. The induction of tumor-specific CD4+ T cells via major histocompatibility complex class II is required to gain optimal anti-tumor immunity against B16 melanoma cell line in tumor immunotherapy using dendritic cells. Exp Dermatol 2009; 18:396 - 403
  • Wang RF. Enhancing antitumor immune responses: intracellular peptide delivery and identification of MHC class II-restricted tumor antigens. Immunol Rev 2002; 188:65 - 80
  • Xie Y, Akpinarli A, Maris C, Hipkiss EL, Lane M, Kwon EK, et al. Naive tumor-specific CD4(+) T cells differentiated in vivo eradicate established melanoma. J Exp Med 207:651 - 667
  • Gregoire S, Logre C, Metharom P, Loing E, Chomilier J, Rosset MB, et al. Identification of two immunogenic domains of the prion protein—PrP— which activate class II-restricted T cells and elicit antibody responses against the native molecule. J Leukoc Biol 2004; 76:125 - 134
  • Bachy V, Ballerini C, Gourdain P, Prignon A, Iken S, Antoine N, et al. Mouse vaccination with dendritic cells loaded with prion protein peptides overcomes tolerance and delays scrapie. J Gen Virol 91:809 - 820
  • Rosset MB, Sacquin A, Lecollinet S, Chaigneau T, Adam M, Crespeau F, et al. Dendritic cell-mediatedimmunization with xenogenic PrP and adenoviral vectors breaks tolerance and prolongs mice survival against experimental scrapie. PLoS One 2009; 4:4917
  • Mosmann TR, Coffman RL. TH1 and TH2 cells: different patterns of lymphokine secretion lead to different functional properties. Annu Rev Immunol 1989; 7:145 - 173
  • Stockinger B, Veldhoen M, Martin B. Th17 T cells: linking innate and adaptive immunity. Semin Immunol 2007; 19:353 - 361
  • Wing K, Sakaguchi S. Regulatory T cells exert checks and balances on self tolerance and autoimmunity. Nat Immunol 11:7 - 13
  • Bluestone JA, Mackay CR, O’Shea JJ, Stockinger B. The functional plasticity of T cell subsets. Nat Rev Immunol 2009; 9:811 - 816
  • Orgogozo JM, Gilman S, Dartigues JF, Laurent B, Puel M, Kirby LC, et al. Subacute meningoencephalitis in a subset of patients with AD after Abeta42 immunization. Neurology 2003; 61:46 - 54
  • Gourdain P, Gregoire S, Iken S, Bachy V, Dorban G, Chaigneau T, et al. Adoptive transfer of T lymphocytes sensitized against the prion protein attenuates prion invasion in scrapie-infected mice. J Immunol 2009; 183:6619 - 6628
  • Kammertoens T, Blankenstein T. Making and circumventing tolerance to cancer. Eur J Immunol 2009; 39:2345 - 2353
  • Schumacher TN, Restifo NP. Adoptive T cell therapy of cancer. Curr Opin Immunol 2009; 21:187 - 189
  • Johnson LA, Morgan RA, Dudley ME, Cassard L, Yang JC, Hughes MS, et al. Gene therapy with human and mouse T-cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen. Blood 2009; 114:535 - 546
  • Sadelain M, Brentjens R, Riviere I. The promise and potential pitfalls of chimeric antigen receptors. Curr Opin Immunol 2009; 21:215 - 223
  • Manz MG, Di Santo JP. Renaissance for mouse models of human hematopoiesis and immunobiology. Nat Immunol 2009; 10:1039 - 1042

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.