Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 11: 584680, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33193401

RESUMEN

Therapeutic vaccines can elicit tumor-specific cytotoxic T lymphocytes (CTLs), but durable reductions in tumor burden require vaccines that stimulate high-avidity CTLs. Recent advances in immunotherapy responses have led to renewed interest in vaccine approaches, including dendritic cell vaccine strategies. However, dendritic cell requirements for vaccines that generate potent anti-tumor T-cell responses are unclear. Here we use mathematical modeling to show that, counterintuitively, increasing levels of immature dendritic cells may lead to selective expansion of high-avidity CTLs. This finding is in contrast with traditional dendritic cell vaccine approaches that have sought to harness ex vivo generated mature dendritic cells. We show that the injection of vaccine antigens in the context of increased numbers of immature dendritic cells results in a decreased overall peptide:MHC complex load that favors high-avidity CTL activation and expansion. Overall, our results provide a firm basis for further development of this approach, both alone and in combination with other immunotherapies such as checkpoint blockade.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Linfocitos T Citotóxicos/inmunología , Antígenos/inmunología , Humanos , Inmunoterapia/métodos , Complejo Mayor de Histocompatibilidad/inmunología , Neoplasias/inmunología
2.
Methods Enzymol ; 632: 503-519, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32000912

RESUMEN

The interaction strength between CD8+ T cells' TCR and cognate peptide-MHC (pMHC) impacts on the CD8+ T cell response against pathogens and tumors (Martinez-Usatorre, Donda, Zehn, & Romero, 2018; Zehn, Lee, & Bevan, 2009). CD8+ T cell responses against tumors are characterized by the presence of low affinity CD8+ T cells specific for nonmutated tumor associated self-antigens (TAA) and potentially high affinity tumor specific CD8+ T cells recognizing mutated self-antigens (Gros et al., 2016; Kvistborg et al., 2012; McMahan & Slansky, 2007). High affinity T cells display enhanced survival, expansion capacity and tumor control (Martinez-Usatorre et al., 2018; Schmid et al., 2010). In fact, recent clinical trials using neoantigen tumor vaccines showed prolonged progression free survival in melanoma patients (Ott et al., 2017; Sahin et al., 2017), while only modest clinical efficacy was obtained with TAA vaccines (Romero et al., 2016). However, the highly individual nature of neoantigens constitutes a major technical and economical hurdle for routine clinical application. Thus, the characterization of TAA-specific CD8+ T cell responses may reveal new strategies to enhance their anti-tumor properties. In parallel, the identification of high affinity antigens and CD8+ T cells may be essential to design effective tumor vaccines and adoptive cell transfer therapies. Therefore, in this chapter, we describe how to generate tumor cell lines with stable expression of affinity-ranged antigens and methods to assess T-cell affinity.


Asunto(s)
Antígenos de Neoplasias/inmunología , Línea Celular Tumoral/inmunología , Antígenos de Neoplasias/genética , Autoantígenos/genética , Autoantígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral/metabolismo , Proliferación Celular , Células Clonales/inmunología , Células Clonales/metabolismo , Células HEK293 , Humanos , Neoplasias/genética , Neoplasias/inmunología , Retroviridae/genética , Retroviridae/inmunología , Transducción Genética
3.
J Theor Biol ; 486: 110067, 2020 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-31704311

RESUMEN

Therapeutic cancer vaccines often do not substantially reduce tumour burden, despite stimulating anti-tumour cytotoxic T lymphocytes (CTLs). Recent experiments have shown that the majority of vaccine-elicited CTLs may be of low-avidity. Moreover, low-avidity CTLs, which are abundant, do not kill cancer cells and potentially inhibit the ability of high-avidity T cells to kill cancer cells. By modelling CTL selection using a system of ordinary differential equations, we show that the efficacy of the peptide vaccine may be improved by controlling its delivery and dosage to preferentially elicit high-avidity CTLs. Our simulations predict that weekly, reduced doses of a vaccine may result in a greater than 90% reduction in cancer concentration. By contrast, a standard vaccine protocol such as a high-dose injection given every 2 weeks induces only a 65% reduction. Our model demonstrates a proof-of-concept approach to targeting immune responses for CTL selection, thereby offering a technique to potentially improve existing therapies.


Asunto(s)
Vacunas contra el Cáncer , Neoplasias , Humanos , Neoplasias/terapia , Péptidos , Linfocitos T Citotóxicos , Vacunas de Subunidad
4.
Ther Adv Vaccines Immunother ; 6(2): 31-47, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29998219

RESUMEN

The interplay between tumours and the immune system has long been known to involve complex interactions between tumour cells, immune cells and the tumour microenvironment. The progress of checkpoint inhibitors in the clinic in the last decade has highlighted again the role of the immune system in the fight against cancer. Numerous efforts have been undertaken to develop ways of stimulating the cellular immune response to eradicate tumours. These interventions include the identification of appropriate tumour antigens as targets for therapy. In this review, we summarize progress in selection of target tumour antigen. Targeting self antigens has the problem of thymic deletion of high-affinity T-cell responses leaving a diminished repertoire of low-affinity T cells that fail to kill tumour cells. Thymic regulation appears to be less stringent for differentiation of cancer-testis antigens, as many tumour rejection antigens fall into this category. More recently, targeting neo-epitopes or post-translational modifications such as a phosphorylation or stress-induced citrullination has shown great promise in preclinical studies. Of particular interest is that the responses can be mediated by both CD4 and CD8 T cells. Previous vaccines have targeted CD8 T-cell responses but more recently, the central role of CD4 T cells in orchestrating inflammation within tumours and also differentiating into potent killer cells has been recognized. The design of vaccines to induce such immune responses is discussed herein. Liposomally encoded ribonucleic acid (RNA), targeted deoxyribonucleic acid (DNA) or long peptides linked to toll-like receptor (TLR) adjuvants are the most promising new vaccine approaches. These exciting new approaches suggest that the 'Holy Grail' of a simple nontoxic cancer vaccine may be on the horizon. A major hurdle in tumour therapy is also to overcome the suppressive tumour environment. We address current progress in combination therapies and suggest that these are likely to show the most promise for the future.

5.
Math Biosci ; 287: 12-23, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27765528

RESUMEN

The expression level of tissue-restricted autoantigens (TSA) in the thymus is crucial for the negative selection of autoreactive T cells during central tolerance. The autoimmune regulator factor (AIRE) plays an important role in the positive regulation of these TSA in medullary thymic epithelial cells and, consequently, in the negative selection of high-avidity autoreactive T cells. Recent studies, however, revealed that thymic islet cell autoantigen (ICA69) expression level in non-obese diabetic (NOD) mice, prone to developing type 1 diabetes (T1D), is reduced due to an increase in the binding affinity of AIRE to the Ica1-promoter region, which regulates ICA69 protein synthesis. This seemed to suggest that AIRE acts as a transcriptional repressor of Ica1 gene in the thymus, causing down regulation in the expression level of ICA69. To investigate this hypothesis and the apparent dual role of AIRE in negative selection, we develop a series of mathematical models of increasing complexity describing the temporal dynamics of self-reactive T cells, AIRE-mRNA and AIRE-(in)dependent thymic TSA-associated genes. The goal is to understand how changing the binding affinity of AIRE to Ica1-promoter affects both T-cell tolerance and the dual role of the transcription factor. Using stability analysis and numerical computations, we show that the model possesses a bistable switch, consisting of healthy and autoimmune states, in the expression level of Ica1 gene with respect to AIRE binding affinity, and that it can capture the experimentally observed dual role of AIRE. We also show that the model must contain a positive feedback loop exerted by T cells on AIRE expression (e.g., via lymphotoxin released by T cells) to produce bistability. Our results suggest that the expression-level of AIRE-mRNA in the healthy state is lower than that of the autoimmune state, and that negative selection is very sensitive to parameter perturbations in T-cell avidity.


Asunto(s)
Autoantígenos/metabolismo , Modelos Teóricos , Timo/inmunología , Timo/metabolismo , Factores de Transcripción/fisiología , Animales , Ratones , Proteína AIRE
6.
Oncoimmunology ; 5(1): e1104448, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26942093

RESUMEN

Despite its negative regulatory role on tumor-specific T cells, Programmed cell death 1 (PD-1) is also a marker of activated tumor-infiltrating T cells. In cancer, PD-1 blockade partially reverses T cell dysfunction allowing the amplification of tumor reactive T cells. Here, we investigated the role of PD-1 signaling on effector/memory human T cells specific for shared melanoma antigens, derived from blood. We documented for the first time the existence of melanoma-specific T cell clones unable to express PD-1. This stable feature was due to the persistent methylation of the PDCD1 promoter. These PD-1neg clones were of lower avidity than their PD-1pos counterparts, suggesting that high-affinity-specific T cell clones unable to express PD-1 are not or rarely present in peripheral blood, as they are probably eliminated by negative selection, due to their high reactivity. We also documented the existence of such PD-1neg T cell clones in melanoma tumor-infiltrating lymphocytes (TIL), which also exhibited a lower functional avidity than PD-1pos TIL clones. This clearly shows that PD-1 expression identifies antigen-specific T cell clonotypes of high functional avidity. Finally, we demonstrated that PD-1 blockade during the in vitro selection process of Melan-A-specific T cells favored the amplification of higher avidity T cell clonotypes. This preferential amplification of high-avidity memory T cells upon PD-1 blockade resonates with the expansion of reactive T cells, including neo-antigen-specific T cells observed in anti-PD-1-treated patients. This feature should also be a useful biomarker of clinical efficiency, while providing new insights for adoptive transfer treatments.

7.
Proc Natl Acad Sci U S A ; 112(7): 2151-6, 2015 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-25646416

RESUMEN

Vaccines eliciting immunity against influenza A viruses (IAVs) are currently antibody-based with hemagglutinin-directed antibody titer the only universally accepted immune correlate of protection. To investigate the disconnection between observed CD8 T-cell responses and immunity to IAV, we used a Poisson liquid chromatography data-independent acquisition MS method to physically detect PR8/34 (H1N1), X31 (H3N2), and Victoria/75 (H3N2) epitopes bound to HLA-A*02:01 on human epithelial cells following in vitro infection. Among 32 PR8 peptides (8-10mers) with predicted IC50 < 60 nM, 9 were present, whereas 23 were absent. At 18 h postinfection, epitope copies per cell varied from a low of 0.5 for M13-11 to a high of >500 for M1(58-66) with PA, HA, PB1, PB2, and NA epitopes also detected. However, aside from M1(58-66), natural CD8 memory responses against conserved presented epitopes were either absent or only weakly observed by blood Elispot. Moreover, the functional avidities of the immunodominant M1(58-66)/HLA-A*02:01-specific T cells were so poor as to be unable to effectively recognize infected human epithelium. Analysis of T-cell responses to primary PR8 infection in HLA-A*02:01 transgenic B6 mice underscores the poor avidity of T cells recognizing M1(58-66). By maintaining high levels of surface expression of this epitope on epithelial and dendritic cells, the virus exploits the combination of immunodominance and functional inadequacy to evade HLA-A*02:01-restricted T-cell immunity. A rational approach to CD8 vaccines must characterize processing and presentation of pathogen-derived epitopes as well as resultant immune responses. Correspondingly, vaccines may be directed against "stealth" epitopes, overriding viral chicanery.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Epítopos/análisis , Virus de la Influenza A/inmunología , Pulmón/virología , Cromatografía Liquida , Células Epiteliales/inmunología , Células Epiteliales/virología , Epítopos/inmunología , Humanos , Pulmón/inmunología
8.
Virology ; 468-470: 479-489, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25261870

RESUMEN

The lung-derived dendritic cell (LDC) recruitment following intranasal (i.n.) vaccination of different poxviral vector-based vaccines/adjuvants were evaluated to decipher how these factors influenced CD8(+) T cell avidity. Compared to the standard i.n. recombinant fowlpox virus (FPV)-HIV vaccination, the FPV-HIV IL-13Rα2 or IL-4Rα antagonist adjuvanted vaccines that induced higher avidity CD8(+) T cells, also recruited significantly elevated MHCII(+) CD11c(+) CD11b(+) CD103(-) CD64(-) MAR-1(-) conventional DC (cDCs) to the lung mucosae (hierarchy: IL-4R antagonist>IL-13Rα2>unadjuvanted). In contrast, elevated CD11b(-) CD103(+) LDCs were detected in animals that received recombinant HIV vaccinia virus (rVV) or Modified Vaccinia Ankara virus (MVA) vector-based vaccines. Adoptive transfer studies indicated that CD11b(-) CD103(+) LDCs significantly dampened HIV-specific CD8(+) T cell avidity compared to CD11b(+) CD103(-) LDCs. Collectively; our observations revealed that rFPV vector prime and transient inhibition of IL-4/IL-13 at the vaccination site favoured the recruitment of unique LDCs, associated with the induction of high quality immunity.


Asunto(s)
Vacunas contra el SIDA/inmunología , Adyuvantes Inmunológicos/farmacología , Células Presentadoras de Antígenos/fisiología , Linfocitos T CD8-positivos/fisiología , Vacunas contra el SIDA/administración & dosificación , Traslado Adoptivo , Animales , Antígenos CD , Antígeno CD11b , Femenino , Cadenas alfa de Integrinas , Interleucina-13/genética , Interleucina-13/metabolismo , Pulmón/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Receptores Tipo II de Interleucina-4/inmunología , Mucosa Respiratoria/citología , Mucosa Respiratoria/inmunología
9.
Vaccine ; 32(43): 5703-14, 2014 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-25151041

RESUMEN

We have established that the efficacy of a heterologous poxvirus vectored HIV vaccine, fowlpox virus (FPV)-HIV gag/pol prime followed by attenuated vaccinia virus (VV)-HIV gag/pol booster immunisation, is strongly influenced by the cytokine milieu at the priming vaccination site, with endogenous IL-13 detrimental to the quality of the HIV specific CD8+ T cell response induced. We have now developed a novel HIV vaccine that co-expresses a C-terminal deletion mutant of the mouse IL-4, deleted for the essential tyrosine (Y119) required for signalling. In our vaccine system, the mutant IL-4C118 can bind to IL-4 type I and II receptors with high affinity, and transiently prevent the signalling of both IL-4 and IL-13 at the vaccination site. When this IL-4C118 adjuvanted vaccine was used in an intranasal rFPV/intramuscular rVV prime-boost immunisation strategy, greatly enhanced mucosal/systemic HIV specific CD8+ T cells with higher functional avidity, expressing IFN-γ, TNF-α and IL-2 and greater protective efficacy were detected. Surprisingly, the IL-4C118 adjuvanted vaccines also induced robust long-lived HIV gag-specific serum antibody responses, specifically IgG1 and IgG2a. The p55-gag IgG2a responses induced were of a higher magnitude relative to the IL-13Rα2 adjuvant vaccine. More interestingly, our recently tested IL-13Rα2 adjuvanted vaccine which only inhibited IL-13 activity, even though induced excellent high avidity HIV-specific CD8+ T cells, had a detrimental impact on the induction of gag-specific IgG2a antibody immunity. Our observations suggest that (i) IL-4 cell-signalling in the absence of IL-13 retarded gag-specific antibody isotype class switching, or (ii) IL-13Rα2 signalling was involved in inducing good gag-specific B cell immunity. Thus, we believe our novel IL-4R antagonist adjuvant strategy offers great promise not only for HIV-1 vaccines, but also against a range of chronic infections where sustained high quality mucosal and systemic T and B cell immunity are required for protection.


Asunto(s)
Vacunas contra el SIDA/inmunología , Linfocitos B/inmunología , Linfocitos T CD8-positivos/inmunología , Infecciones por VIH/prevención & control , Receptores de Interleucina-4/antagonistas & inhibidores , Adyuvantes Inmunológicos/farmacología , Animales , Femenino , Anticuerpos Anti-VIH/sangre , Anticuerpos Anti-VIH/inmunología , Cambio de Clase de Inmunoglobulina , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Memoria Inmunológica , Interferón gamma/inmunología , Interleucina-13/inmunología , Interleucina-2/inmunología , Interleucina-4/inmunología , Ratones Endogámicos BALB C , Precursores de Proteínas/inmunología , Factor de Necrosis Tumoral alfa/inmunología
10.
Cytokine Growth Factor Rev ; 25(4): 437-42, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25159217

RESUMEN

The roles of interleukin (IL)-4 and IL-13 during both innate and adaptive Th2 mediated immunity have received considerable scrutiny, however, mechanisms by which these cytokines influence the cellular interactions involved in negatively modulating the development of effective Th1 immunity are poorly characterized. In this article we discuss the recent advances in IL-4/IL-13 biology, mainly (i) role of these cytokines in allergic inflammation, atopic dermatitis, cancer, transplant rejection, bacterial/viral infections, and specifically the therapeutic potential of IL-13Rα2, (ii) insights into how "alarmin" stimulation activate IL-4/IL-13 at the lung mucosae, (iii) how these two cytokines modulate antigen-specific CD8(+) T cell quality/avidity in a vaccine route dependent manner and (iv) finally discuss the potential of using transient inhibition of IL-4 and/or IL-13 at the vaccination site as a platform vaccine technology to induce strong sustained high quality CD8(+) T cell immunity for protection against many chronic mucosal pathogens such as HIV-1.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Subunidad alfa2 del Receptor de Interleucina-13/inmunología , Interleucina-13/inmunología , Interleucina-4/inmunología , Receptores de Interleucina-4/inmunología , Vacunas contra el SIDA/inmunología , Síndrome de Inmunodeficiencia Adquirida/inmunología , Síndrome de Inmunodeficiencia Adquirida/prevención & control , Adyuvantes Inmunológicos/uso terapéutico , Humanos , Hipersensibilidad/inmunología , Interleucina-13/antagonistas & inhibidores , Interleucina-4/antagonistas & inhibidores , Neoplasias/inmunología , Células TH1/inmunología , Células Th2/inmunología
11.
Immunol Lett ; 161(2): 204-6, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24462961

RESUMEN

The ICI 2013 Mucosal Vaccine Workshop presentations covered a wide range of topics, these mainly fell into three categories: (i) Understanding the interactions of host and microbes, specifically commensal pathogens and improving the antigen uptake via the (microfold cells) M cells to induce effective IgA antibody immunity at the gut mucosa; (ii) effective plant-based vaccines and (iii) development of prophylactic and therapeutic mucosal-based vaccine strategies for virus infections such as human immunodeficiency virus (HIV), influenza and human papillomavirus (HPV) associated head and neck cancers. How to improve the efficacy of oral vaccines, novel intranasal mucosal adjuvants and a unique intra-cheek delivery method were also discussed. Presenters emphasized the differences associated with systemic and mucosal vaccination, specifically, how mucosal vaccines unlike systemic delivery can induce effective immunity at the first line of defence. Collectively, the workshop provided insights into recent developments in the mucosal vaccine research field, highlighting the complexities associated with designing safe and effective mucosal vaccines.


Asunto(s)
Inmunidad Mucosa , Membrana Mucosa/inmunología , Adyuvantes Inmunológicos , Animales , Antígenos/inmunología , Tracto Gastrointestinal/inmunología , Neoplasias de Cabeza y Cuello/inmunología , Neoplasias de Cabeza y Cuello/terapia , Humanos , Vacunas/administración & dosificación , Vacunas/inmunología , Virosis/inmunología , Virosis/prevención & control
12.
Vaccines (Basel) ; 2(3): 537-48, 2014 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-26344744

RESUMEN

While T cells recognise the complex of peptide and major histocompatibility complex (MHC) at the cell surface, changes in the dose and/or structure of the peptide component can have profound effects on T cell activation and function. In addition, the repertoire of T cells capable of responding to any given peptide is variable, but broader than a single clone. Consequently, peptide parameters that affect the interaction between T cells and peptide/MHC have been shown to select particular T cell clones for expansion and this impacts on clearance of disease. T cells with high functional avidity are selected on low doses of peptide, while low avidity T cells are favoured in high peptide concentrations. Altering the structure of the peptide ligand can also influence the selection and function of peptide-specific T cell clones. In this review, we will explore the evidence that the choice of peptide dose or the structure of the peptide are critical parameters in an effective vaccine designed to activate T cells.

13.
Front Immunol ; 4: 363, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24265631

RESUMEN

Adoptive transfer of T cells gene-engineered with antigen-specific T cell receptors (TCRs) has proven its feasibility and therapeutic potential in the treatment of malignant tumors. To ensure further clinical development of TCR gene therapy, it is necessary to target immunogenic epitopes that are related to oncogenesis and selectively expressed by tumor tissue, and implement strategies that result in optimal T cell fitness. In addition, in particular for the treatment of solid tumors, it is equally necessary to include strategies that counteract the immune-suppressive nature of the tumor micro-environment. Here, we will provide an overview of the current status of TCR gene therapy, and redefine the following three challenges of improvement: "choice of target antigen"; "fitness of T cells"; and "sensitization of tumor milieu." We will categorize and discuss potential strategies to address each of these challenges, and argue that advancement of clinical TCR gene therapy critically depends on developments toward each of the three challenges.

14.
Vaccine ; 31(41): 4548-55, 2013 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-23933364

RESUMEN

We have shown that mucosal HIV-1 recombinant pox viral vaccination can induce high, avidity HIV-specific CD8(+) T cells with reduced interleukin (IL)-4 and IL-13 expression compared to, systemic vaccine delivery. In the current study how these cytokines act to regulate anti-viral CD8(+) T, cell avidity following HIV-1 recombinant pox viral prime-boost vaccination was investigated. Out of a panel of T cell avidity markers tested, only CD8 expression levels were found to be enhanced on, KdGag197-205 (HIV)-specific CD8(+) T cells obtained from IL-13(-/-), IL-4(-/-) and signal transducer and, activator of transcription of 6 (STAT6)(-/-) mice compared to wild-type (WT) controls following, vaccination. Elevated CD8 expression levels in this instance also correlated with polyfunctionality, (interferon (IFN)-γ, tumour necorsis factor (TNF)-α and IL-2 production) and the avidity of HIVspecific CD8(+) T cells. Furthermore, mucosal vaccination and vaccination with the novel adjuvanted IL-13 inhibitor (i.e. IL-13Rα2) vaccines significantly enhanced CD8 expression levels on HIV-specific CD8(+), T cells, which correlated with avidity. Using anti-CD8 antibodies that blocked CD8 availability on CD8(+), T cells, it was established that CD8 played an important role in increasing HIV-specific CD8(+) T cell avidity and polyfunctionality in IL-4(-/-), IL-13(-/-) and STAT6(-/-) mice compared to WT controls, following vaccination. Collectively, our data demonstrate that IL-4 and IL-13 dampen CD8 expression levels on anti-viral CD8(+) T cells, which can down-regulate anti-viral CD8(+) T cell avidity and, polyfunctionality following HIV-1 recombinant pox viral vaccination. These findings can be exploited to, design more efficacious vaccines not only against HIV-1, but many chronic infections where high, avidity CD8(+) T cells help protection.


Asunto(s)
Vacunas contra el SIDA/inmunología , Antígenos CD8/biosíntesis , Linfocitos T CD8-positivos/inmunología , VIH-1/inmunología , Interleucina-13/inmunología , Interleucina-4/inmunología , Vacunas contra el SIDA/administración & dosificación , Administración a través de la Mucosa , Animales , Regulación hacia Abajo , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA