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










Base de datos
Intervalo de año de publicación
1.
Cancers (Basel) ; 16(3)2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-38339227

RESUMEN

As part of a symposium, current and former directors of Immune Monitoring cores and investigative oncologists presented insights into the past, present and future of immune assessment. Dr. Gnjatic presented a classification of immune monitoring technologies ranging from universally applicable to experimental protocols, while emphasizing the need for assay harmonization. Dr. Obeng discussed physiologic differences among CD8 T cells that align with anti-tumor responses. Dr. Lyerly presented the Soldano Ferrone lecture, commemorating the passionate tumor immunologist who inspired many, and covered a timeline of monitoring technology development and its importance to immuno-oncology. Dr. Sonabend presented recent achievements in glioblastoma treatment, accentuating the range of monitoring techniques that allowed him to refine patient selection for clinical trials. Dr. Guevara-Patiño focused on hypoxia within the tumor environment and stressed that T cell viability is not to be confused with functionality. Dr. Butterfield accentuated monitoring of dendritic cell metabolic (dys)function as a determinant for tumor vaccine success. Lectures were interspersed with select abstract presentations. To summarize the concepts, Dr. Maecker from Stanford led an informative forum discussion, pointing towards the future of immune monitoring. Immune monitoring continues to be a guiding light towards effective immunotherapeutic strategies.

2.
J Leukoc Biol ; 115(2): 306-321, 2024 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-37949818

RESUMEN

The role of natural killer group 2D (NKG2D) in peripheral T cells as a costimulatory receptor is well established. However, its contribution to T cell thymic education and functional imprint is unknown. Here, we report significant changes in development, receptor signaling, transcriptional program, and function in T cells from mice lacking NKG2D signaling. In C57BL/6 (B6) and OT-I mice, we found that NKG2D deficiency results in Vß chain usage changes and stagnation of the double-positive stage in thymic T cell development. We found that the expression of CD5 and CD45 in thymocytes from NKG2D deficient mice were reduced, indicating a direct influence of NKG2D on the strength of T cell receptor (TCR) signaling during the developmental stage of T cells. Depicting the functional consequences of NKG2D, peripheral OT-I NKG2D-deficient cells were unresponsive to ovalbumin peptide stimulation. Paradoxically, while αCD3/CD28 agonist antibodies led to phenotypic T cell activation, their ability to produce cytokines remained severely compromised. We found that OT-I NKG2D-deficient cells activate STAT5 in response to interleukin-15 but were unable to phosphorylate ERK or S6 upon TCR engagement, underpinning a defect in TCR signaling. Finally, we showed that NKG2D is expressed in mouse and human thymic T cells at the double-negative stage, suggesting an evolutionarily conserved function during T cell development. The data presented in this study indicate that NKG2D impacts thymic T cell development at a fundamental level by reducing the TCR threshold and affecting the functional imprint of the thymic progeny. In summary, understanding the impact of NKG2D on thymic T cell development and TCR signaling contributes to our knowledge of immune system regulation, immune dysregulation, and the design of immunotherapies.


Asunto(s)
Subfamilia K de Receptores Similares a Lectina de Células NK , Timo , Animales , Ratones , Humanos , Ratones Endogámicos C57BL , Timocitos , Receptores de Antígenos de Linfocitos T
3.
Mol Ther Oncolytics ; 31: 100751, 2023 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-38075241

RESUMEN

CD33 and CD123 are expressed on the surface of human acute myeloid leukemia blasts and other noncancerous tissues such as hematopoietic stem cells. On-target off-tumor toxicities may limit chimeric antigen receptor T cell therapies that target both CD33 and CD123. To overcome this limitation, we developed bispecific human CD33/CD123 chimeric antigen receptor (CAR) T cells with an "AND" logic gate. We produced novel CD33 and CD123 scFvs from monoclonal antibodies that bound CD33 and CD123 and activated T cells. Screening of CD33 and CD123 CAR T cells for cytotoxicity, cytokine production, and proliferation was performed, and we selected scFvs for CD33/CD123 bispecific CARs. The bispecific CARs split 4-1BB co-stimulation on one scFv and CD3ζ on the other. In vitro testing of cytokine secretion and cytotoxicity resulted in selecting bispecific CAR 1 construct for in vivo analysis. The CD33/CD123 bispecific CAR T cells were able to control acute myeloid leukemia (AML) in a xenograft AML mouse model similar to monospecific CD33 and CD123 CAR T cells while showing no on-target off-tumor effects. Based on our findings, human CD33/CD123 bispecific CAR T cells are a promising cell-based approach to prevent AML and support clinical investigation.

4.
J Immunother Cancer ; 11(7)2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37433716

RESUMEN

BACKGROUND: Immunotherapies are becoming front-line treatments for many advanced cancers, and combinations of two or more therapies are beginning to be investigated. Based on their individual antitumor capabilities, we sought to determine whether combination oncolytic virus (OV) and radiation therapy (RT) may improve cancer outcomes. METHODS: To investigate the activity of this combination therapy, we used in vitro mouse and human cancer cell lines as well as a mouse model of skin cancer. After initial results, we further included immune checkpoint blockade, whose addition constituted a triple combination immunotherapy. RESULTS: Our findings demonstrate that OV and RT reduce tumor growth via conversion of immunologically 'cold' tumors to 'hot', via a CD8+ T cell-dependent and IL-1α-dependent mechanism that is associated with increased PD-1/PD-L1 expression, and the triple combination of OV, RT, and PD-1 checkpoint inhibition impedes tumor growth and prolongs survival. Further, we describe the response of a PD-1-refractory patient with cutaneous squamous cell carcinoma who received the triple combination of OV, RT, and immune checkpoint inhibitor (ICI), and went on to experience unexpected, prolonged control and survival. He remains off-treatment and is without evidence of progression for >44 months since study entry. CONCLUSIONS: Effective systemic antitumor immune response is rarely elicited by a single therapy. In a skin cancer mouse model, we demonstrate improved outcomes with combination OV, RT, and ICI treatment, which is associated with mechanisms involving augmented CD8+ T cell infiltration and IL-1α expression. We report tumor reduction and prolonged survival of a patient with skin cancer treated with combination OV, RT, and ICI. Overall, our data provide strong rationale for combining OV, RT, and ICI for treatment of patients with ICI-refractory skin and potentially other cancers.


Asunto(s)
Carcinoma de Células Escamosas , Inhibidores de Puntos de Control Inmunológico , Viroterapia Oncolítica , Neoplasias Cutáneas , Animales , Humanos , Masculino , Ratones , Carcinoma de Células Escamosas/terapia , Modelos Animales de Enfermedad , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunoterapia , Neoplasias Cutáneas/terapia , Línea Celular Tumoral , Terapia Combinada
5.
Cancer Res Commun ; 3(5): 896-907, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37377902

RESUMEN

Head and neck squamous cell carcinoma (HNSCC) has one of the most hypoxic and immunosuppressive tumor microenvironments (TME) among solid tumors. However, there is no proven therapeutic strategy to remodel the TME to be less hypoxic and proinflammatory. In this study, we classified tumors according to a Hypoxia-Immune signature, characterized the immune cells in each subgroup, and analyzed the signaling pathways to identify a potential therapeutic target that can remodel the TME. We confirmed that hypoxic tumors had significantly higher numbers of immunosuppressive cells, as evidenced by a lower ratio of CD8+ T cells to FOXP3+ regulatory T cells, compared with nonhypoxic tumors. Patients with hypoxic tumors had worse outcomes after treatment with pembrolizumab or nivolumab, anti-programmed cell death-1 inhibitors. Our expression analysis also indicated that hypoxic tumors predominantly increased the expression of the EGFR and TGFß pathway genes. Cetuximab, an anti-EGFR inhibitor, decreased the expression of hypoxia signature genes, suggesting that it may alleviate the effects of hypoxia and remodel the TME to become more proinflammatory. Our study provides a rationale for treatment strategies combining EGFR-targeted agents and immunotherapy in the management of hypoxic HNSCC. Significance: While the hypoxic and immunosuppressive TME of HNSCC has been well described, comprehensive evaluation of the immune cell components and signaling pathways contributing to immunotherapy resistance has been poorly characterized. We further identified additional molecular determinants and potential therapeutic targets of the hypoxic TME to fully leverage currently available targeted therapies that can be administered with immunotherapy.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Humanos , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Cetuximab/farmacología , Carcinoma de Células Escamosas/tratamiento farmacológico , Receptores ErbB/genética , Linfocitos T CD8-positivos/metabolismo , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Hipoxia/genética , Microambiente Tumoral/genética
6.
BioDrugs ; 37(4): 505-520, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37256534

RESUMEN

Antibody-drug conjugates (ADCs) have transformed the treatment landscape in oncology and become an essential therapeutic modality. In urothelial carcinoma (UC), the two ADCs that have been especially successful in clinical practice are enfortumab vedotin and sacituzumab govitecan. These drugs are currently approved as monotherapy for later lines of treatment in locally advanced or metastatic UC and have had a significant impact for patients with limited treatment options. Combinational trials, as well as additional ADCs, are currently being investigated in the treatment of UC for subsequent lines of therapy as overall survival rates remain dismal.


Asunto(s)
Carcinoma de Células Transicionales , Inmunoconjugados , Neoplasias de la Vejiga Urinaria , Humanos , Carcinoma de Células Transicionales/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Inmunoconjugados/uso terapéutico , Penicilinas
7.
Expert Opin Emerg Drugs ; 28(1): 17-26, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36882977

RESUMEN

INTRODUCTION: The treatment landscape for advanced-stage, unresectable or metastatic urothelial carcinoma (mUC) has shifted dramatically over a short period of time, with new therapeutic agents available for clinical use. However, despite these recent advances in the field, mUC continues to be a disease with significant morbidity and mortality and remains generally incurable. While platinum-based therapy remains the backbone of therapy, many patients are ineligible for chemotherapy or have failed initial chemotherapy treatment. In post-platinum treated patients, immunotherapy and antibody drug conjugates have provided incremental advances, but agents with better therapeutic index guided by precision medicine are needed. AREAS COVERED: This article covers the available monoclonal antibody therapies in mUC excluding immunotherapy and antibody drug conjugates. Included are a review of data utilizing monoclonal antibodies targeting VEG-F, HER-2, FGFR, and KIR-2 in the setting of mUC. A literature search from 6/2022- 9/2022 was performed utilizing PubMed with key terms including urothelial carcinoma, monoclonal antibody, VEG-F, HER-2, FGFR. EXPERT OPINION: Often used in combination with immunotherapy or other therapeutic agents, monoclonal antibody therapies have exhibited efficacy in mUC in early trials. Upcoming clinical trials will further explore their full clinical utility in treating mUC patients.


Asunto(s)
Carcinoma de Células Transicionales , Inmunoconjugados , Neoplasias de la Vejiga Urinaria , Humanos , Carcinoma de Células Transicionales/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Anticuerpos Monoclonales/efectos adversos , Inmunoterapia , Inmunoconjugados/uso terapéutico
8.
Int J Mol Sci ; 24(4)2023 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-36835342

RESUMEN

After exposure to an antigen, CD8 T cells reach a decision point about their fate: to become either short-lived effector cells (SLECs) or memory progenitor effector cells (MPECs). SLECs are specialized in providing an immediate effector function but have a shorter lifespan and lower proliferative capacity compared to MPECs. Upon encountering the cognate antigen during an infection, CD8 T cells rapidly expand and then contract to a level that is maintained for the memory phase after the peak of the response. Studies have shown that the contraction phase is mediated by TGFß and selectively targets SLECs, while sparing MPECs. The aim of this study is to investigate how the CD8 T cell precursor stage determines TGFß sensitivity. Our results demonstrate that MPECs and SLECs have differential responses to TGFß, with SLECs being more sensitive to TGFß than MPECs. This difference in sensitivity is associated with the levels of TGFßRI and RGS3, and the SLEC-related transcriptional activator T-bet binding to the TGFßRI promoter may provide a molecular basis for increased TGFß sensitivity in SLECs.


Asunto(s)
Linfocitos T CD8-positivos , Memoria Inmunológica , Subgrupos de Linfocitos T , Factor de Crecimiento Transformador beta , Animales , Ratones , Antígenos/metabolismo , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular , Ratones Endogámicos C57BL , Subgrupos de Linfocitos T/inmunología , Factor de Crecimiento Transformador beta/inmunología
9.
Mol Immunol ; 155: 1-6, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36634520

RESUMEN

CD8 T cells play a critical role in immunity against intracellular pathogens and cancer. A primary objective of T cell-based vaccine strategies is the induction of durable and effective immune responses. Achieving this goal involves more than simply boosting the numbers of responding T cells. Of particular interest is the induction of CD8 T cells with polycytokine capability, specifically with the ability of CD8 T cells to co-produce IFNγ, TNFα and IL-2. The presence of these polycytokine-producing CD8 T cells correlates strongly with protection against foreign pathogens and cancer. Therefore, approaches capable of inducing such polyfunctional responses are needed. NKG2D engagement on CD8 T cells has been shown to result in increased effector response. However, the manner in which NKG2D engagement results in improved CD8 T cell effector response is unclear. Here we demonstrate in vitro and in vivo that NKG2D engagement by its natural ligand, Rae-1ε, shifts the balance from single cytokine to polycytokine (IL-2, IFNγ, and TFNα) production. These data define a previously unrecognized process in which NKG2D costimulation on CD8 T cells results in improved effector responses.


Asunto(s)
Citocinas , Neoplasias , Humanos , Subfamilia K de Receptores Similares a Lectina de Células NK , Interleucina-2 , Linfocitos T CD8-positivos
10.
Immunol Invest ; 51(8): 2215-2225, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35797428

RESUMEN

CAR T-cell therapy has transformed the treatment of hematological malignancies of the B cell lineage. However, the quest to fulfil the same promise for solid tumors is still in its infancy. This review summarizes some of the challenges that the field is trying to overcome for effective treatment of human carcinomas, including tumor heterogeneity, the paucity of truly tumor-specific targets, immunosuppression and metabolic restrictions at solid tumor beds, and defective T-cell trafficking. All these barriers are being currently investigated and, in some cases, targeted, by multiple independent groups. With clinical interventions against multiple human malignancies and different platforms under accelerated clinical development, the next few years will see an array of cellular therapies, including CAR T-cells, progressively becoming routine interventions to eliminate currently incurable diseases, as it happened with some hematological malignancies.


Asunto(s)
Neoplasias Hematológicas , Neoplasias , Receptores Quiméricos de Antígenos , Humanos , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/metabolismo , Neoplasias/terapia , Neoplasias/metabolismo , Inmunoterapia Adoptiva , Linfocitos T/metabolismo , Neoplasias Hematológicas/terapia , Neoplasias Hematológicas/metabolismo , Microambiente Tumoral
11.
J Immunother Cancer ; 10(6)2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35676062

RESUMEN

BACKGROUND: T cell receptor (TCR) signaling profile is a fundamental property that underpins both adaptive and innate immunity in the host. Despite its potential clinical relevance, the TCR repertoire in peripheral blood has not been thoroughly explored for its value as an immunotherapy efficacy biomarker in head and neck squamous cell carcinoma (HNSCC). The purpose of the present study is to characterize and compare the TCR repertoire in peripheral blood mononuclear cells (PBMC) from patients with HNSCC treated with the combination of cetuximab and nivolumab. METHODS: We used the immunoSEQ assay to sequence the TCR beta (TCR-B) chain repertoire from serially obtained PBMC at baseline and during the treatments from a total of 41 patients who received the combination (NCT03370276). Key TCR repertoire metrics, including diversity and clonality, were calculated and compared between patients with different therapy responses and clinical characteristics (eg, human papillomavirus (HPV) status and smoking history). Patient survival outcomes were compared according to patient groups stratified by the TCR-B clonotyping. To confirm the observed patterns in TCR spectrum, samples from patients who achieved complete response (CR) and partial response (PR) were further profiled with the immunoSEQ deep resolution assay. RESULTS: Our data indicated that the patients who achieved CR and PR had an increased TCR sequence diversity in their baseline samples, this tendency being more pronounced in HPV-negative patients or those with a smoking history. Notably, the CR/PR group had the lowest proportion of patients with oligoclonal TCR clones (2 out of 8 patients), followed by the stable disease group (9 out of 20 patients) and lastly the progressive disease group (7 out of 10 patients). An overall trend toward favorable patient survival was also observed in the polyclonal group. Finally, we reported the shared TCR clones across patients within the same response group, as well as the shared clones by aligning immunoSEQ reads with TCR data retrieved from The Cancer Genome Atlas- head and neck squamous cell carcinoma (TCGA-HNSC) cohort. CONCLUSIONS: Our data suggest that, despite the great clinical heterogeneity of HNSCC and the limited responders in the present cohort, the peripheral TCR repertoires from pretreatment PBMC may be developed as biomarkers for the benefit of immunotherapy in HNSCC.


Asunto(s)
Neoplasias de Cabeza y Cuello , Infecciones por Papillomavirus , Biomarcadores , Cetuximab , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Humanos , Leucocitos Mononucleares , Nivolumab/farmacología , Nivolumab/uso terapéutico , Infecciones por Papillomavirus/tratamiento farmacológico , Receptores de Antígenos de Linfocitos T , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Linfocitos T
12.
Diagnostics (Basel) ; 12(4)2022 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-35454006

RESUMEN

Through a multitude of studies, the gut microbiota has been recognized as a significant influencer of both homeostasis and pathophysiology. Certain microbial taxa can even affect treatments such as cancer immunotherapies, including the immune checkpoint blockade. These taxa can impact such processes both individually as well as collectively through mechanisms from quorum sensing to metabolite production. Due to this overarching presence of the gut microbiota in many physiological processes distal to the GI tract, we hypothesized that mice bearing tumors at extraintestinal sites would display a distinct intestinal microbial signature from non-tumor-bearing mice, and that such a signature would involve taxa that collectively shift with tumor presence. Microbial OTUs were determined from 16S rRNA genes isolated from the fecal samples of C57BL/6 mice challenged with either B16-F10 melanoma cells or PBS control and analyzed using QIIME. Relative proportions of bacteria were determined for each mouse and, using machine-learning approaches, significantly altered taxa and co-occurrence patterns between tumor- and non-tumor-bearing mice were found. Mice with a tumor had elevated proportions of Ruminococcaceae, Peptococcaceae.g_rc4.4, and Christensenellaceae, as well as significant information gains and ReliefF weights for Bacteroidales.f__S24.7, Ruminococcaceae, Clostridiales, and Erysipelotrichaceae. Bacteroidales.f__S24.7, Ruminococcaceae, and Clostridiales were also implicated through shifting co-occurrences and PCA values. Using these seven taxa as a melanoma signature, a neural network reached an 80% tumor detection accuracy in a 10-fold stratified random sampling validation. These results indicated gut microbial proportions as a biosensor for tumor detection, and that shifting co-occurrences could be used to reveal relevant taxa.

13.
Cell Stress Chaperones ; 26(5): 845-857, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34542825

RESUMEN

Developing immunosuppressive therapies for autoimmune diseases comes with a caveat that immunosuppression may promote the risk of developing other conditions or diseases. We have previously shown that biolistic delivery of an expression construct encoding inducible HSP70 (HSP70i) with one amino acid modification in the dendritic cell (DC) activating moiety 435-445 (HSP70iQ435A) to mouse skin resulted in significant immunosuppressive activity of autoimmune vitiligo, associated with fewer tissue infiltrating T cells. To prepare HSP70iQ435A as a potential therapeutic for autoimmune vitiligo, in this study we evaluated whether and how biolistic delivery of HSP70iQ435A in mice affects anti-tumor responses. We found that HSP70iQ435A in fact supports anti-tumor responses in melanoma-challenged C57BL/6 mice. Biolistic delivery of the HSP70iQ435A-encoding construct to mice elicited significant anti-HSP70 titers, and anti-HSP70 IgG and IgM antibodies recognize surface-expressed and cytoplasmic HSP70i in human and mouse melanoma cells. A peptide scan revealed that the anti-HSP70 antibodies recognize a specific C-terminal motif within the HSP70i protein. The antibodies elicited surface CD107A expression among mouse NK cells, representative of antibody-mediated cellular cytotoxicity (ADCC), supporting the concept, that HSP70iQ435A-encoding DNA elicits a humoral response to the stress protein expressed selectively on the surface of melanoma cells. Thus, besides limiting autoimmunity and inflammation, HSP70iQ435A elicits humoral responses that limit tumor growth and may be used in conjunction with immune checkpoint inhibitors to not only control tumor but to also limit adverse events following tumor immunotherapy.


Asunto(s)
Autoinmunidad , Proteínas HSP70 de Choque Térmico/genética , Melanoma/genética , Melanoma/inmunología , Mutación/genética , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/inmunología , Animales , Anticuerpos/metabolismo , Autoinmunidad/genética , Degranulación de la Célula , Línea Celular Tumoral , ADN de Neoplasias/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/fisiología , Masculino , Melanoma/patología , Ratones Endogámicos C57BL , Modelos Biológicos , Neoplasias Cutáneas/patología
14.
Front Immunol ; 12: 714137, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34177971

RESUMEN

[This corrects the article DOI: 10.3389/fimmu.2021.624131.].

15.
Front Immunol ; 12: 624131, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33717132

RESUMEN

Vitiligo is an acquired multifactorial disease that affects melanocytes and results in skin depigmentation. In this review, we examine the role of cells stress and self-reactive T cells responses. Given the canonical and non-canonical functions of NKG2D, such as authenticating stressed target and enhance TCR signaling, we examine how melanocyte stress leads to the expression of ligands that are recognized by the activating receptor NKG2D, and how its signaling results in the turning of T cells against self (melanocyte suicide by proxy). We also discuss how this initiation phase is followed by T cell perpetuation, as NKG2D signaling results in self-sustained long-lasting T cells, with improved cytolytic properties.


Asunto(s)
Autoinmunidad , Linfocitos T CD8-positivos/metabolismo , Melanocitos/metabolismo , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Pigmentación de la Piel , Piel/metabolismo , Vitíligo/metabolismo , Animales , Linfocitos T CD8-positivos/inmunología , Microambiente Celular , Citotoxicidad Inmunológica , Humanos , Melanocitos/inmunología , Melanocitos/patología , Estrés Oxidativo , Transducción de Señal , Piel/inmunología , Piel/patología , Vitíligo/genética , Vitíligo/inmunología , Vitíligo/patología
16.
J Invest Dermatol ; 140(6): 1131-1133, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32446332

RESUMEN

In the study by Jacquemin et al., the authors reported that ligands for NKG2D are upregulated in vitiligo perilesional skin and especially in patients with active disease. The reasons for the elevated expression of NKG2D ligands are unknown. This study, however, provides a framework for understanding vitiligo: Skin resident CD8 T cells recognize and kill melanocytes through NKG2D signaling. This event results in the increased production and release of cyto/chemokines and the development of long-lasting CD8 T cells, which in turn causes the recruitment of new T cells, thus perpetuating and disseminating the disease.


Asunto(s)
Subfamilia K de Receptores Similares a Lectina de Células NK , Vitíligo , Linfocitos T CD8-positivos , Humanos , Melanocitos , Piel
17.
J Immunother Cancer ; 7(1): 48, 2019 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-30777125

RESUMEN

BACKGROUND: The development of memory responses is an evolutionary function of the adaptive immune system. We propose that for the immune system to populate the memory compartment with the best-suited CD8 T cells it utilizes a process of certification or molecular accreditation mediated through Natural Killer Group 2D (NKG2D). This process of certification assures that the memory compartment is filled with CD8 T cells that have demonstrated their ability to kill their cognate targets through a two-step process that utilizes T cell receptor (TCR) and NKG2D signaling. METHODS: One week after immunization with peptide-pulsed dendritic cells, NKG2D signaling was transiently blocked in vivo with a single injection of neutralizing antibodies. Under such conditions, we determined the importance of NKG2D signaling during the effector phase for memory formation without compromising NKG2D signaling at the memory phase. Both open (polyclonal) and closed (monoclonal) CD8 T cell repertoires were studied. RESULTS: We show that signaling through NKG2D mediated this certification. Temporary blockade of NKG2D signaling during the effector phase resulted in the formation of highly defective memory CD8 T cells characterized by altered expression of the ribosomal protein S6 and epigenetic modifiers, suggesting modifications in the T cell translational machinery and epigenetic programming. Finally, these uncertified memory cells were not protective against a B16 tumor challenge. CONCLUSION: Signaling through NKG2D during the effector phase (certification) favors the development of functional memory CD8 T cells, a previously undescribed role for NKG2D. Temporary blockade of NKG2D signaling during the effector phase results in the formation of highly defective memory CD8 T cells potentially by affecting the expression of the ribosomal protein S6 and epigenetic modifiers, suggesting alterations in T cell translational machinery and epigenetic programming.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica , Subfamilia K de Receptores Similares a Lectina de Células NK/inmunología , Animales , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones Endogámicos C57BL , Ratones Transgénicos
18.
J Invest Dermatol ; 138(12): 2531-2539, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30031029

RESUMEN

Human HSP70iQ435A carries a single amino-acid modification within the dendritic cell activating region and tolerizes dendritic cells in vitro. The underlying DNA was used to prevent and treat disease in vitiligo mouse models through reduced dendritic cell activation and diminished skin T-cell infiltration, suggesting the same may be useful for patients. Physiologic differences between mouse and human skin then called for studies in large animals with human-like skin. We established the efficiency of DNA jet injection into swine skin before subcloning HSP70iQ435A into clinically suitable vector pUMVC3. Vitiligo lesions in Sinclair swine were treated with plasmid DNA to measure changes in depigmentation, T-cell infiltration, expression of HSP70i in skin, serum HSP70i, and anti-HSP70i serum titers. Remarkable repigmentation following HSP70iQ435A-encoding DNA treatment persisted throughout the 6-month follow-up period. Repigmentation was accompanied by an initial influx of T cells accompanied by increased CD4/CD8 ratios, waning by week 15. Melanocytes spanned the border of repigmenting skin, suggesting that melanocyte repopulation precedes skin melanization. Serum titer fluctuations were not treatment-associated. Importantly, treatment did not interfere with melanoma immunosurveillance. These data encourage clinical testing of HSP70iQ435A.


Asunto(s)
Células Dendríticas/inmunología , Proteínas HSP70 de Choque Térmico/metabolismo , Inmunoterapia/métodos , Melanoma/inmunología , Pigmentación de la Piel/genética , Piel/patología , Linfocitos T/inmunología , Vitíligo/inmunología , Animales , Diferenciación Celular , Movimiento Celular , Células Cultivadas , ADN/genética , Modelos Animales de Enfermedad , Terapia Genética , Proteínas HSP70 de Choque Térmico/genética , Humanos , Tolerancia Inmunológica , Vigilancia Inmunológica , Inyecciones a Chorro , Activación de Linfocitos , Melanoma/genética , Melanoma/terapia , Mutación/genética , Neoplasias Experimentales , Piel/metabolismo , Porcinos , Vitíligo/genética , Vitíligo/terapia
19.
Cell Mol Immunol ; 15(5): 470-479, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29400704

RESUMEN

Natural killer group 2 member D (NKG2D) is a type II transmembrane receptor. NKG2D is present on NK cells in both mice and humans, whereas it is constitutively expressed on CD8+ T cells in humans but only expressed upon T-cell activation in mice. NKG2D is a promiscuous receptor that recognizes stress-induced surface ligands. In NK cells, NKG2D signaling is sufficient to unleash the killing response; in CD8+ T cells, this requires concurrent activation of the T-cell receptor (TCR). In this case, the function of NKG2D is to authenticate the recognition of a stressed target and enhance TCR signaling. CD28 has been established as an archetype provider of costimulation during T-cell priming. It has become apparent, however, that signals from other costimulatory receptors, such as NKG2D, are required for optimal T-cell function outside the priming phase. This review will focus on the similarities and differences between NKG2D and CD28; less well-described characteristics of NKG2D, such as the potential role of NKG2D in CD8+ T-cell memory formation, cancer immunity and autoimmunity; and the opportunities for targeting NKG2D in immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Inmunoterapia , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Animales , Autoinmunidad , Humanos , Memoria Inmunológica , Neoplasias/inmunología
20.
Cell Rep ; 17(4): 957-965, 2016 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-27760326

RESUMEN

In light of increased cancer prevalence and cancer-specific deaths in patients with infections, we investigated whether infections alter anti-tumor immune responses. We report that acute influenza infection of the lung promotes distal melanoma growth in the dermis and leads to accelerated cancer-specific host death. Furthermore, we show that during influenza infection, anti-melanoma CD8+ T cells are shunted from the tumor to the infection site, where they express high levels of the inhibitory receptor programmed cell death protein 1 (PD-1). Immunotherapy to block PD-1 reverses this loss of anti-tumor CD8+ T cells from the tumor and decreases infection-induced tumor growth. Our findings show that acute non-oncogenic infection can promote cancer growth, raising concerns regarding acute viral illness sequelae. They also suggest an unexpected role for PD-1 blockade in cancer immunotherapy and provide insight into the immune response when faced with concomitant challenges.


Asunto(s)
Melanoma/inmunología , Melanoma/patología , Oncogenes , Infecciones por Orthomyxoviridae/patología , Enfermedad Aguda , Animales , Linfocitos T CD8-positivos/inmunología , Proliferación Celular , Pulmón/patología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Receptor de Muerte Celular Programada 1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...