Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
J Adv Nurs ; 2024 Feb 08.
Article in English | MEDLINE | ID: mdl-38332481

ABSTRACT

AIMS: To explore the lived experiences of mature female students undertaking a Bachelor of Nursing (Adult) programme in the UK, to gain insight into the challenges and barriers faced by students and investigate the factors that support students who have considered leaving, to stay and continue with their studies. BACKGROUND: There is a global shortage of nurses and challenges exist in ensuring that enough nurses are available to provide care in the complex and rapidly changing care environments. Initiatives introduced to increase the number of Registered Nurses (RN), include increasing the number of students enrolled on pre-registration nursing programmes. However, the success of this intervention is contingent on the number of students who go on to complete their course. DESIGN: This qualitative study employed Interpretative Phenomenological Analysis (IPA), which provided a methodological framework and analytical approach to enable an exploration of participants' individual and shared lived experiences. METHODS: Eight female, mature students at the end of their second year of a Bachelor of Nursing (Adult) programme at a Higher Education Institution in South Wales participated in semi-structured, face-to-face interviews, which were analysed idiographically before group-level analysis was undertaken. FINDINGS: The analysis revealed three superordinate themes: 'Ambition to become a Registered Nurse'; 'Jugging Roles' and 'Particular Support Needs for a Particular Student'. CONCLUSION: Each student had a unique history, their past and present social and psychological experiences were multifaceted and complex. These differences resulted in varying degrees of resilience and motivations to continue their studies. These findings are important for ensuring that services develop and provide effective support to maximize retention and, ultimately, increase the number of students entering the RN workforce. PATIENT OF PUBLIC CONTRIBUTION: No patient or public contribution. IMPACT STATEMENT: This research expands on current literature regarding the needs of mature female students, a growing student nurse demographic. Every student had a dynamic set of circumstances and demonstrated that the identification of 'at-risk' students, purely based on demographics or information on a Curriculum Vitae, is problematic and potentially futile. This knowledge could be used to tailor University support systems and inform curriculum development and support systems for maximizing student retention. These findings are important for ensuring that services continue to develop and provide effective support to maximize retention and completion and, ultimately, increase the number of students entering the Nursing and Midwifery Council register.

2.
Immunother Adv ; 4(1): ltad030, 2024.
Article in English | MEDLINE | ID: mdl-38223410

ABSTRACT

The concept of a therapeutic cancer vaccine to activate anti-tumour immunity pre-dates innovations in checkpoint blockade immunotherapies. However, vaccination strategies have yet to show the hoped-for successes in patients, and unanswered questions regarding the underlying immunological mechanisms behind cancer vaccines have hampered translation to clinical practice. Recent advances in our understanding of the potential of tumour mutational burden and neo-antigen-reactive T cells for response to immunotherapy have re-ignited enthusiasm for cancer vaccination strategies, coupled with the development of novel mRNA-based vaccines following successes in prevention of COVID-19. Here we summarise current developments in cancer vaccines and discuss how advances in our comprehension of the cellular interplay in immunotherapy-responsive tumours may inform better design of therapeutic cancer vaccines, with a focus on the role of dendritic cells as the orchestrators of anti-tumour immunity. The increasing number of clinical trials and research being funnelled into cancer vaccines has demonstrated the 'proof-of-principle', supporting the hypothesis that therapeutic vaccines have potential as an immuno-oncology agent. For efficacious and safe cancer vaccines to be developed, better understanding of the underpinning immunological mechanisms is paramount.

4.
Mucosal Immunol ; 16(5): 753-763, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37385586

ABSTRACT

Macrophages play essential roles in tissue homeostasis, defense, and repair. Their functions are highly tissue-specific, and when damage and inflammation stimulate repopulation by circulating monocytes, the incoming monocytes rapidly acquire the same, tissue-specific functions as the previous, resident macrophages. Several environmental factors are thought to guide the functional differentiation of recruited monocytes, including metabolic pressures imposed by the fuel sources available in each tissue. Here we discuss whether such a model of metabolic determinism can be applied to macrophage differentiation across barrier sites, from the lung to the skin. We suggest an alternative model, in which metabolic phenotype is a consequence of macrophage longevity rather than an early driver of tissue-specific adaption.


Subject(s)
Macrophages , Monocytes , Humans , Macrophages/metabolism , Monocytes/metabolism , Homeostasis , Inflammation/metabolism , Lung/metabolism
5.
Nat Commun ; 14(1): 2880, 2023 05 19.
Article in English | MEDLINE | ID: mdl-37208336

ABSTRACT

Regulation of cutaneous immunity is severely compromised in inflammatory skin disease. To investigate the molecular crosstalk underpinning tolerance versus inflammation in atopic dermatitis, we utilise a human in vivo allergen challenge study, exposing atopic dermatitis patients to house dust mite. Here we analyse transcriptional programmes at the population and single cell levels in parallel with immunophenotyping of cutaneous immunocytes revealed a distinct dichotomy in atopic dermatitis patient responsiveness to house dust mite challenge. Our study shows that reactivity to house dust mite was associated with high basal levels of TNF-expressing cutaneous Th17 T cells, and documents the presence of hub structures where Langerhans cells and T cells co-localised. Mechanistically, we identify expression of metallothioneins and transcriptional programmes encoding antioxidant defences across all skin cell types, that appear to protect against allergen-induced inflammation. Furthermore, single nucleotide polymorphisms in the MTIX gene are associated with patients who did not react to house dust mite, opening up possibilities for therapeutic interventions modulating metallothionein expression in atopic dermatitis.


Subject(s)
Dermatitis, Atopic , Animals , Humans , Dermatitis, Atopic/genetics , Allergens , Inflammation/genetics , Skin , Pyroglyphidae
6.
Sci Adv ; 9(15): eadd1992, 2023 04 14.
Article in English | MEDLINE | ID: mdl-37043573

ABSTRACT

While skin is a site of active immune surveillance, primary melanomas often escape detection. Here, we have developed an in silico model to determine the local cross-talk between melanomas and Langerhans cells (LCs), the primary antigen-presenting cells at the site of melanoma development. The model predicts that melanomas fail to activate LC migration to lymph nodes until tumors reach a critical size, which is determined by a positive TNF-α feedback loop within melanomas, in line with our observations of murine tumors. In silico drug screening, supported by subsequent experimental testing, shows that treatment of primary tumors with MAPK pathway inhibitors may further prevent LC migration. In addition, our in silico model predicts treatment combinations that bypass LC dysfunction. In conclusion, our combined approach of in silico and in vivo studies suggests a molecular mechanism that explains how early melanomas develop under the radar of immune surveillance by LC.


Subject(s)
Melanoma , Skin , Mice , Animals , Cell Movement , Skin/metabolism , Langerhans Cells/metabolism , Melanoma/metabolism
7.
Cell Rep ; 39(7): 110819, 2022 05 17.
Article in English | MEDLINE | ID: mdl-35584681

ABSTRACT

T cell pathology in the skin leads to monocyte influx, but we have little understanding of the fate of recruited cells within the diseased niche, or the long-term impact on cutaneous immune homeostasis. By combining a murine model of acute graft-versus-host disease (aGVHD) with analysis of patient samples, we demonstrate that pathology initiates dermis-specific macrophage differentiation and show that aGVHD-primed macrophages continue to dominate the dermal compartment at the relative expense of quiescent MHCIIint cells. Exposure of the altered dermal niche to topical haptens after disease resolution results in hyper-activation of regulatory T cells (Treg), but local breakdown in tolerance. Disease-imprinted macrophages express increased IL-1ß and are predicted to elicit altered TNF superfamily interactions with cutaneous Treg, and we demonstrate the direct loss of T cell regulation within the resolved skin. Thus, T cell pathology leaves an immunological scar in the skin marked by failure to re-set immune homeostasis.


Subject(s)
Graft vs Host Disease , Skin , Animals , Humans , Immune Tolerance , Macrophages/metabolism , Mice , Monocytes/metabolism , Skin/metabolism , T-Lymphocytes, Regulatory
8.
NPJ Digit Med ; 5(1): 18, 2022 Feb 14.
Article in English | MEDLINE | ID: mdl-35165389

ABSTRACT

The COVID-19 pandemic has pushed healthcare systems globally to a breaking point. The urgent need for effective and affordable COVID-19 treatments calls for repurposing combinations of approved drugs. The challenge is to identify which combinations are likely to be most effective and at what stages of the disease. Here, we present the first disease-stage executable signalling network model of SARS-CoV-2-host interactions used to predict effective repurposed drug combinations for treating early- and late stage severe disease. Using our executable model, we performed in silico screening of 9870 pairs of 140 potential targets and have identified nine new drug combinations. Camostat and Apilimod were predicted to be the most promising combination in effectively supressing viral replication in the early stages of severe disease and were validated experimentally in human Caco-2 cells. Our study further demonstrates the power of executable mechanistic modelling to enable rapid pre-clinical evaluation of combination therapies tailored to disease progression. It also presents a novel resource and expandable model system that can respond to further needs in the pandemic.

10.
Immunology ; 163(1): 105-111, 2021 05.
Article in English | MEDLINE | ID: mdl-33502012

ABSTRACT

Whether resident and recruited myeloid cells may impair or aid healing of acute skin wounds remains a debated question. To begin to address this, we examined the importance of CD11c+ myeloid cells in the early activation of skin wound repair. We find that an absence of CD11c+ cells delays wound closure and epidermal proliferation, likely due to defects in the activation of the IL-23-IL-22 axis that is required for wound healing.


Subject(s)
CD11 Antigens/deficiency , Dendritic Cells/immunology , Skin/immunology , Wound Healing , Wounds and Injuries/immunology , Animals , CD11 Antigens/genetics , Dendritic Cells/metabolism , Disease Models, Animal , Kinetics , Langerhans Cells/immunology , Langerhans Cells/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Skin/metabolism , Skin/pathology , Wounds and Injuries/genetics , Wounds and Injuries/metabolism , Wounds and Injuries/pathology
12.
J Clin Invest ; 130(4): 1896-1911, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31917684

ABSTRACT

Acute graft-versus-host disease (GVHD) is initially triggered by alloreactive T cells, which damage peripheral tissues and lymphoid organs. Subsequent transition to chronic GVHD involves the emergence of autoimmunity, although the underlying mechanisms driving this process are unclear. Here, we tested the hypothesis that acute GVHD blocks peripheral tolerance of autoreactive T cells by impairing lymph node (LN) display of peripheral tissue-restricted antigens (PTAs). At the initiation of GVHD, LN fibroblastic reticular cells (FRCs) rapidly reduced expression of genes regulated by DEAF1, an autoimmune regulator-like transcription factor required for intranodal expression of PTAs. Subsequently, GVHD led to the selective elimination of the FRC population, and blocked the repair pathways required for its regeneration. We used a transgenic mouse model to show that the loss of presentation of an intestinal PTA by FRCs during GVHD resulted in the activation of autoaggressive T cells and gut injury. Finally, we show that FRCs normally expressed a unique PTA gene signature that was highly enriched for genes expressed in the target organs affected by chronic GVHD. In conclusion, acute GVHD damages and prevents repair of the FRC network, thus disabling an essential platform for purging autoreactive T cells from the repertoire.


Subject(s)
Autoantigens/immunology , Autoimmunity , Graft vs Host Disease/immunology , Intestinal Diseases/immunology , Lymph Nodes/immunology , T-Lymphocytes/immunology , Animals , Autoantigens/genetics , Graft vs Host Disease/genetics , Graft vs Host Disease/pathology , Intestinal Diseases/genetics , Intestinal Diseases/pathology , Lymph Nodes/pathology , Mice , Mice, Knockout , T-Lymphocytes/pathology
13.
Sci Immunol ; 4(38)2019 08 23.
Article in English | MEDLINE | ID: mdl-31444235

ABSTRACT

A dense population of embryo-derived Langerhans cells (eLCs) is maintained within the sealed epidermis without contribution from circulating cells. When this network is perturbed by transient exposure to ultraviolet light, short-term LCs are temporarily reconstituted from an initial wave of monocytes but thought to be superseded by more permanent repopulation with undefined LC precursors. However, the extent to which this process is relevant to immunopathological processes that damage LC population integrity is not known. Using a model of allogeneic hematopoietic stem cell transplantation, where alloreactive T cells directly target eLCs, we have asked whether and how the original LC network is ultimately restored. We find that donor monocytes, but not dendritic cells, are the precursors of long-term LCs in this context. Destruction of eLCs leads to recruitment of a wave of monocytes that engraft in the epidermis and undergo a sequential pathway of differentiation via transcriptionally distinct EpCAM+ precursors. Monocyte-derived LCs acquire the capacity of self-renewal, and proliferation in the epidermis matched that of steady-state eLCs. However, we identified a bottleneck in the differentiation and survival of epidermal monocytes, which, together with the slow rate of renewal of mature LCs, limits repair of the network. Furthermore, replenishment of the LC network leads to constitutive entry of cells into the epidermal compartment. Thus, immune injury triggers functional adaptation of mechanisms used to maintain tissue-resident macrophages at other sites, but this process is highly inefficient in the skin.


Subject(s)
Langerhans Cells/immunology , Monocytes/immunology , Animals , Cells, Cultured , Humans , Langerhans Cells/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
14.
Sci Immunol ; 4(35)2019 05 31.
Article in English | MEDLINE | ID: mdl-31152091

ABSTRACT

CTLA-4 is a critical negative regulator of the immune system and a major target for immunotherapy. However, precisely how it functions in vivo to maintain immune homeostasis is not clear. As a highly endocytic molecule, CTLA-4 can capture costimulatory ligands from opposing cells by a process of transendocytosis (TE). By restricting costimulatory ligand expression in this manner, CTLA-4 controls the CD28-dependent activation of T cells. Regulatory T cells (Tregs) constitutively express CTLA-4 at high levels and, in its absence, show defects in TE and suppressive function. Activated conventional T cells (Tconv) are also capable of CTLA-4-dependent TE; however, the relative use of this mechanism by Tregs and Tconv in vivo remains unclear. Here, we set out to characterize both the perpetrators and cellular targets of CTLA-4 TE in vivo. We found that Tregs showed constitutive cell surface recruitment of CTLA-4 ex vivo and performed TE rapidly after TCR stimulation. Tregs outperformed activated Tconv at TE in vivo, and expression of ICOS marked Tregs with this capability. Using TCR transgenic Tregs that recognize a protein expressed in the pancreas, we showed that the presentation of tissue-derived self-antigen could trigger Tregs to capture costimulatory ligands in vivo. Last, we identified migratory dendritic cells (DCs) as the major target for Treg-based CTLA-4-dependent regulation in the steady state. These data support a model in which CTLA-4 expressed on Tregs dynamically regulates the phenotype of DCs trafficking to lymph nodes from peripheral tissues in an antigen-dependent manner.


Subject(s)
CTLA-4 Antigen/metabolism , Cell Movement/immunology , Dendritic Cells/immunology , T-Lymphocytes, Regulatory/immunology , Transcytosis/immunology , Animals , Antigen Presentation/immunology , Autoantigens/immunology , B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , CTLA-4 Antigen/genetics , Female , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Phenotype , Receptors, Antigen, T-Cell/metabolism
16.
Front Immunol ; 9: 963, 2018.
Article in English | MEDLINE | ID: mdl-29770141

ABSTRACT

The skin is the most common target organ affected by graft-versus-host disease (GVHD), with severity and response to therapy representing important predictors of patient survival. Although many of the initiating events in GVHD pathogenesis have been defined, less is known about why treatment resistance occurs or why there is often a permanent failure to restore tissue homeostasis. Emerging data suggest that the unique immune microenvironment in the skin is responsible for defining location- and context-specific mechanisms of injury that are distinct from those involved in other target organs. In this review, we address recent advances in our understanding of GVHD biology in the skin and outline the new research themes that will ultimately enable design of precision therapies.


Subject(s)
Graft vs Host Disease/immunology , Graft vs Host Disease/physiopathology , Skin Diseases/immunology , Animals , B-Lymphocytes/immunology , Cellular Microenvironment/immunology , Cytokines/immunology , Hematopoietic Stem Cell Transplantation , Humans , Mice , Microbiota/immunology , Skin Diseases/etiology , T-Lymphocytes/immunology , Transplantation, Homologous
17.
Mol Ther ; 26(6): 1471-1481, 2018 06 06.
Article in English | MEDLINE | ID: mdl-29628306

ABSTRACT

Ongoing clinical trials explore T cell receptor (TCR) gene therapy as a treatment option for cancer, but responses in solid tumors are hampered by the immunosuppressive microenvironment. The production of TCR gene-engineered T cells requires full T cell activation in vitro, and it is currently unknown whether in vivo interactions with conventional dendritic cells (cDCs) regulate the accumulation and function of engineered T cells in tumors. Using the B16 melanoma model and the inducible depletion of CD11c+ cells in CD11c.diphtheria toxin receptor (DTR) mice, we analyzed the interaction between tumor-resident cDCs and engineered T cells expressing the melanoma-specific TRP-2 TCR. We found that depletion of CD11c+ cells triggered the recruitment of cross-presenting cDC1 into the tumor and enhanced the accumulation of TCR-engineered T cells. We show that the recruited tumor cDCs present melanoma tumor antigen, leading to enhanced activation of TCR-engineered T cells. In addition, detailed analysis of the tumor myeloid compartment revealed that the depletion of a population of DT-sensitive macrophages can contribute to the accumulation of tumor-infiltrating T cells. Together, these data suggest that the relative frequency of tumor-resident cDCs and macrophages may impact the therapeutic efficacy of TCR gene therapy in solid tumors.


Subject(s)
Dendritic Cells/metabolism , Macrophages/metabolism , Receptors, Antigen, T-Cell/metabolism , Animals , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , CD11c Antigen/immunology , CD11c Antigen/metabolism , Heparin-binding EGF-like Growth Factor/immunology , Heparin-binding EGF-like Growth Factor/metabolism , Humans , Immunotherapy, Adoptive/methods , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, T-Cell/immunology
18.
JCI Insight ; 3(5)2018 03 08.
Article in English | MEDLINE | ID: mdl-29515032

ABSTRACT

Graft-versus-host disease (GVHD) is a life-threatening complication of allogeneic stem cell transplantation induced by the influx of donor-derived effector T cells (TE) into peripheral tissues. Current treatment strategies rely on targeting systemic T cells; however, the precise location and nature of instructions that program TE to become pathogenic and trigger injury are unknown. We therefore used weighted gene coexpression network analysis to construct an unbiased spatial map of TE differentiation during the evolution of GVHD and identified wide variation in effector programs in mice and humans according to location. Idiosyncrasy of effector programming in affected organs did not result from variation in T cell receptor repertoire or the selection of optimally activated TE. Instead, TE were reprogrammed by tissue-autonomous mechanisms in target organs for site-specific proinflammatory functions that were highly divergent from those primed in lymph nodes. In the skin, we combined the correlation-based network with a module-based differential expression analysis and showed that Langerhans cells provided in situ instructions for a Notch-dependent T cell gene cluster critical for triggering local injury. Thus, the principal determinant of TE pathogenicity in GVHD is the final destination, highlighting the need for target organ-specific approaches to block immunopathology while avoiding global immune suppression.


Subject(s)
Cellular Reprogramming/immunology , Graft vs Host Disease/immunology , Langerhans Cells/immunology , Skin/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Surface/genetics , Antigens, Surface/metabolism , Bone Marrow Transplantation/adverse effects , Cells, Cultured , Cellular Reprogramming/genetics , Disease Models, Animal , Female , Gene Expression Regulation/immunology , Graft vs Host Disease/pathology , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Langerhans Cells/metabolism , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Male , Mannose-Binding Lectins/genetics , Mannose-Binding Lectins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multigene Family/genetics , Multigene Family/immunology , Primary Cell Culture , Receptors, Notch/metabolism , Skin/cytology , Skin/pathology , T-Lymphocytes, Cytotoxic/metabolism , Transplantation Chimera , Transplantation, Homologous/adverse effects
19.
J Clin Invest ; 128(5): 2010-2024, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29485974

ABSTRACT

A key predictor for the success of gene-modified T cell therapies for cancer is the persistence of transferred cells in the patient. The propensity of less differentiated memory T cells to expand and survive efficiently has therefore made them attractive candidates for clinical application. We hypothesized that redirecting T cells to specialized niches in the BM that support memory differentiation would confer increased therapeutic efficacy. We show that overexpression of chemokine receptor CXCR4 in CD8+ T cells (TCXCR4) enhanced their migration toward vascular-associated CXCL12+ cells in the BM and increased their local engraftment. Increased access of TCXCR4 to the BM microenvironment induced IL-15-dependent homeostatic expansion and promoted the differentiation of memory precursor-like cells with low expression of programmed death-1, resistance to apoptosis, and a heightened capacity to generate polyfunctional cytokine-producing effector cells. Following transfer to lymphoma-bearing mice, TCXCR4 showed a greater capacity for effector expansion and better tumor protection, the latter being independent of changes in trafficking to the tumor bed or local out-competition of regulatory T cells. Thus, redirected homing of T cells to the BM confers increased memory differentiation and antitumor immunity, suggesting an innovative solution to increase the persistence and functions of therapeutic T cells.


Subject(s)
Bone Marrow/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Movement/immunology , Immunologic Memory , Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Bone Marrow/pathology , CD8-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Chemokine CXCL12/genetics , Chemokine CXCL12/immunology , Humans , Interleukin-15/genetics , Interleukin-15/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/therapy , Receptors, CXCR4/genetics , Receptors, CXCR4/immunology , T-Lymphocytes, Regulatory/pathology
20.
Mol Ther ; 25(2): 504-511, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28153097

ABSTRACT

Recombinant lentiviral vectors (LVs) are highly effective vaccination vehicles that elicit protective T cell immunity in disease models. Dendritic cells (DCs) acquire antigen at sites of vaccination and migrate to draining lymph nodes, where they prime vaccine-specific T cells. The potency with which LVs activate CD8+ T cell immunity has been attributed to the transduction of DCs at the immunization site and durable presentation of LV-encoded antigens. However, it is not known how LV-encoded antigens continue to be presented to T cells once directly transduced DCs have turned over. Here, we report that LV-encoded antigen is efficiently cross-presented by DCs in vitro. We have further exploited the temporal depletion of DCs in the murine CD11c.DTR (diphtheria toxin receptor) model to demonstrate that repopulating DCs that were absent at the time of immunization cross-present LV-encoded antigen to T cells in vivo. Indirect presentation of antigen from transduced cells by DCs is sufficient to prime functional effector T cells that control tumor growth. These data suggest that DCs cross-present immunogenic antigen from LV-transduced cells, thereby facilitating prolonged activation of T cells in the absence of circulating LV particles. These are findings that may impact on the future design of LV vaccination strategies.


Subject(s)
Antigen Presentation , Antigens/immunology , Cross-Priming/immunology , Dendritic Cells/immunology , Immunity, Cellular , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Antigens/genetics , Biomarkers , CD11c Antigen/metabolism , Cancer Vaccines/immunology , Coculture Techniques , Dendritic Cells/metabolism , Genetic Vectors/genetics , Heparin-binding EGF-like Growth Factor/immunology , Lentivirus/genetics , Lymph Nodes/immunology , Lymph Nodes/metabolism , Mice , Models, Animal , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Transduction, Genetic , Vaccines/genetics , Vaccines/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...