Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Physiology (Bethesda) ; 37(1): 46-52, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34486395

RESUMEN

The hypoxia-hypoxia-inducible factor (HIF)-1α-A2-adenosinergic pathway protects tissues from inflammatory damage during antipathogen immune responses. The elimination of this physiological tissue-protecting mechanism by supplemental oxygenation may contribute to the high mortality of oxygen-ventilated COVID-19 patients by exacerbating inflammatory lung damage. Restoration of this pathway with hypoxia-adenosinergic drugs may improve outcomes in these patients.


Asunto(s)
COVID-19 , Neoplasias , Humanos , Hipoxia , Receptor de Adenosina A2A , SARS-CoV-2
2.
Adv Funct Mater ; 31(37)2021 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-37745940

RESUMEN

Solid tumors are protected from antitumor immune responses due to their hypoxic microenvironments. Weakening hypoxia-driven immunosuppression by hyperoxic breathing of 60% oxygen has shown to be effective in unleashing antitumor immune cells against solid tumors. However, efficacy of systemic oxygenation is limited against solid tumors outside of lungs and has been associated with unwanted side effects. As a result, it is essential to develop targeted oxygenation alternatives to weaken tumor hypoxia as novel approaches to restore immune responses against cancer. Herein, we report on injectable oxygen-generating cryogels (O2-cryogels) to reverse tumor-induced hypoxia. These macroporous biomaterials were designed to locally deliver oxygen, inhibit the expression of hypoxia-inducible genes in hypoxic melanoma cells, and reduce the accumulation of immunosuppressive extracellular adenosine. Our data show that O2-cryogels enhance T cell-mediated secretion of cytotoxic proteins, restoring the killing ability of tumor-specific CTLs, both in vitro and in vivo. In summary, O2-cryogels provide a unique and safe platform to supply oxygen as a co-adjuvant in hypoxic tumors and have the potential to improve cancer immunotherapies.

3.
J Immunol ; 201(2): 782-791, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29802128

RESUMEN

Tumor hypoxia-driven accumulation of extracellular adenosine was shown to facilitate tumor evasion by engaging the immunosuppressive, intracellular cAMP-elevating A2 adenosine receptors (A2R) on tumor-reactive effector T cells, but there remains a need for careful evaluation of the limiting factors and properties of A2R blockade-enabled antitumor immunity. In studies of A2AR and/or A2BR gene-deficient mice, we found that A2AR deletion-but not A2BR deletion-liberates endogenous CD8+ T cell antitumor immunity against weakly immunogenic MCA205 sarcomas. Studies of adoptively transferred A2AR-/-, A2BR-/-, or A2AR-/-/A2BR-/- tumor-reactive T cells confirmed that immunosuppression in the tumor microenvironment was mediated by A2AR on CD8+ T cells. Treatment with A2AR antagonist mimicked A2AR gene deletion in adoptive T cell immunotherapy. This therapeutic benefit of targeting A2AR was independent of the anatomical location of tumor growth. The enhanced antitumor reactivity also led to the eradication of established intracranial tumors, which was associated with mouse survival and the maintenance of long-lasting, tumor-specific immunological memory. The blockade of the A2AR on adoptively transferred T cells by synthetic A2AR antagonist led to higher levels of IFN-γ secretion by tumor-infiltrating CD8+ T cells. These data clarify the mechanism of hypoxia-driven immunosuppression in the tumor microenvironment by A2AR on tumor-reactive CD8+ T cells and show that selective A2AR antagonists can be effective in improving the outcomes of T cell-based immunotherapies. Demonstration of the T cell dose dependency of tumor rejection points to a major limitation of current cancer immunotherapies, in which the presence of sufficient numbers of tumor-reactive T cells in a patient is not known.


Asunto(s)
Neoplasias Encefálicas/inmunología , Linfocitos T CD8-positivos/inmunología , Hipoxia/inmunología , Inmunoterapia Adoptiva/métodos , Receptor de Adenosina A2A/metabolismo , Sarcoma/inmunología , Adenosina/metabolismo , Antagonistas del Receptor de Adenosina A2/farmacología , Animales , Linfocitos T CD8-positivos/trasplante , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Tolerancia Inmunológica , Inmunidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Adenosina A2A/genética , Escape del Tumor , Microambiente Tumoral
4.
Adv Exp Med Biol ; 1136: 113-121, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31201720

RESUMEN

Long-term studies of anti-pathogen and anti-tumor immunity have provided complementary genetic and pharmacological evidence for the immunosuppressive and immunomodulatory effects of Hypoxia-HIF-1α and adenosine-mediated suppression via the A2A adenosine receptor signaling pathway (Hypoxia-A2A-adenosinergic). This pathway is life saving when it protects inflamed tissues of vital organs from collateral damage by overactive anti-pathogen immune cells or enables the differentiation of cells of adaptive immunity. However, the Hypoxia-A2A-adenosinergic immunosuppression can also prevent tumor rejection by inhibiting the anti-tumor effects of T and NK cells. In addition, this suppressive pathway has been shown to mask tumors due to the hypoxia-HIF-α-mediated loss of MHC Class I molecules on tumor cells. It is suggested that it will be impossible to realize the full anti-tumor capacities of current cancer immunotherapies without simultaneous administration of anti-Hypoxia-A2A-Adenosinergic drugs that inactivate this tumor-protecting mechanism in hypoxic and adenosine-rich tumors.Here, we overview the supporting evidence for the conceptually novel immunotherapeutic motivation to breathe supplemental oxygen (40-60%) or to repurpose already available oxygenation agents in combination with current immunotherapies. Preclinical studies provide strong support for oxygen immunotherapy to enable much stronger tumor regression by weakening immunosuppression by A2A adenosine receptors and by the Hypoxia➔HIF-1α axis. The results of these studies emphasize the value of systemic oxygenation as clinically feasible, promising, and as a valuable tool for mechanistic investigations of tumor biology and cancer immunology. Perhaps the most effective and feasible among individual members of this novel class of anti-tumor drugs are oxygenation agents.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Terapia de Inmunosupresión , Neoplasias/patología , Hipoxia Tumoral , Hipoxia de la Célula , Línea Celular Tumoral , Humanos , Tolerancia Inmunológica , Receptor de Adenosina A2A/fisiología , Transducción de Señal
5.
J Biol Chem ; 292(4): 1211-1217, 2017 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-27974461

RESUMEN

T follicular helper (TFH) cells have been shown to be critically required for the germinal center (GC) reaction where B cells undergo class switch recombination and clonal selection to generate high affinity neutralizing antibodies. However, detailed knowledge of the physiological cues within the GC microenvironment that regulate T cell help is limited. The cAMP-elevating, Gs protein-coupled A2a adenosine receptor (A2aR) is an evolutionarily conserved receptor that limits and redirects cellular immunity. However, the role of A2aR in humoral immunity and B cell differentiation is unknown. We hypothesized that the hypoxic microenvironment within the GC facilitates an extracellular adenosine-rich milieu, which serves to limit TFH frequency and function, and also promotes immunosuppressive T follicular regulatory cells (TFR). In support of this hypothesis, we found that following immunization, mice lacking A2aR (A2aRKO) exhibited a significant expansion of T follicular cells, as well as increases in TFH to TFR ratio, GC T cell frequency, GC B cell frequency, and class switching of GC B cells to IgG1. Transfer of CD4 T cells from A2aRKO or wild type donors into T cell-deficient hosts revealed that these increases were largely T cell-intrinsic. Finally, injection of A2aR agonist, CGS21680, following immunization suppressed T follicular differentiation, GC B cell frequency, and class switching of GC B cells to IgG1. Taken together, these observations point to a previously unappreciated role of GS protein-coupled A2aR in regulating humoral immunity, which may be pharmacologically targeted during vaccination or pathological states in which GC-derived autoantibodies contribute to the pathology.


Asunto(s)
Autoanticuerpos/inmunología , Centro Germinal/inmunología , Inmunidad Humoral , Cambio de Clase de Inmunoglobulina/inmunología , Inmunoglobulina G/inmunología , Receptores Purinérgicos P1/inmunología , Linfocitos T Reguladores/inmunología , Adenosina/análogos & derivados , Adenosina/farmacología , Animales , Autoanticuerpos/genética , Linfocitos B/inmunología , Cambio de Clase de Inmunoglobulina/efectos de los fármacos , Inmunoglobulina G/genética , Ratones , Ratones Noqueados , Fenetilaminas/farmacología , Receptores Purinérgicos P1/genética
6.
J Immunol ; 197(10): 4014-4020, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27798169

RESUMEN

Germinal centers (GCs) are anatomic sites where B cells undergo secondary diversification to produce high-affinity, class-switched Abs. We hypothesized that proliferating B cells in GCs create a hypoxic microenvironment that governs their further differentiation. Using molecular markers, we found GCs to be predominantly hypoxic. Compared to normoxia (21% O2), hypoxic culture conditions (1% O2) in vitro accelerated class switching and plasma cell formation and enhanced expression of GL-7 on B and CD4+ T cells. Reversal of GC hypoxia in vivo by breathing 60% O2 during immunization resulted in reduced frequencies of GC B cells, T follicular helper cells, and plasmacytes, as well as lower expression of ICOS on T follicular helper cells. Importantly, this reversal of GC hypoxia decreased Ag-specific serum IgG1 and reduced the frequency of IgG1+ B cells within the Ag-specific GC. Taken together, these observations reveal a critical role for hypoxia in GC B cell differentiation.


Asunto(s)
Hipoxia de la Célula , Centro Germinal/inmunología , Centro Germinal/metabolismo , Cambio de Clase de Inmunoglobulina , Recombinación Genética , Animales , Linfocitos B/inmunología , Linfocitos B/fisiología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/fisiología , Diferenciación Celular , Centro Germinal/citología , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Inmunoglobulinas/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Oxígeno/metabolismo , Células Plasmáticas/inmunología , Células Plasmáticas/fisiología
7.
Eur J Immunol ; 43(3): 655-66, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23208786

RESUMEN

Hypoxia-adenosinergic suppression and redirection of the immune response has been implicated in the regulation of antipathogen and antitumor immunity, with hypoxia-inducible factor 1α (HIF-1α) playing a major role. In this study, we investigated the role of isoform I.1, a quantitatively minor alternative isoform of HIF-1α, in antibacterial immunity and sepsis survival. By using the cecal ligation and puncture model of bacterial peritonitis, we studied the function of I.1 isoform in T cells using mice with total I.1 isoform deficiency and mice with T-cell-targeted I.1 knockdown. We found that genetic deletion of the I.1 isoform resulted in enhanced resistance to septic lethality, significantly reduced bacterial load in peripheral blood, increased M1 macrophage polarization, augmented levels of proinflammatory cytokines in serum, and significantly decreased levels of the anti-inflammatory cytokine IL-10. Our data suggest a previously unrecognized immunosuppressive role for the I.1 isoform in T cells during bacterial sepsis. We interpret these data as indicative that the activation-inducible isoform I.1 hinders the contribution of T cells to the antibacterial response by affecting M1/M2 macrophage polarization and microbicidal function.


Asunto(s)
Infecciones Bacterianas/genética , Infecciones Bacterianas/inmunología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Peritonitis/genética , Peritonitis/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Infecciones Bacterianas/mortalidad , Modelos Animales de Enfermedad , Subunidad alfa del Factor 1 Inducible por Hipoxia/deficiencia , Activación de Linfocitos/inmunología , Macrófagos/inmunología , Masculino , Ratones , Ratones Noqueados , Especificidad de Órganos/genética , Peritonitis/mortalidad , Isoformas de Proteínas , Receptores de Antígenos de Linfocitos T/metabolismo , Sepsis/genética , Sepsis/inmunología , Sepsis/mortalidad , Activación Transcripcional
8.
Bioorg Med Chem ; 21(23): 7453-64, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24126093

RESUMEN

Molecular modeling techniques were applied to the design, synthesis and optimization of a new series of xanthine based adenosine A(2A) receptor antagonists. The optimized lead compound was converted to a PEG derivative and a functional in vitro bioassay used to confirm efficacy. Additionally, the PEGylated version showed enhanced aqueous solubility and was inert to photoisomerization, a known limitation of existing antagonists of this class.


Asunto(s)
Diseño de Fármacos , Antagonistas de Receptores Purinérgicos P1/química , Antagonistas de Receptores Purinérgicos P1/farmacología , Receptor de Adenosina A2A/metabolismo , Xantina/química , Xantina/farmacología , Línea Celular , Cristalografía por Rayos X , Humanos , Hipoxia/terapia , Inmunoterapia , Modelos Moleculares , Polietilenglicoles/química , Polietilenglicoles/farmacología , Receptor de Adenosina A2A/química
9.
J Immunol ; 186(4): 2444-53, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21242513

RESUMEN

Antimicrobial treatment strategies must improve to reduce the high mortality rates in septic patients. In noninfectious models of acute inflammation, activation of A2B adenosine receptors (A2BR) in extracellular adenosine-rich microenvironments causes immunosuppression. We examined A2BR in antibacterial responses in the cecal ligation and puncture (CLP) model of sepsis. Antagonism of A2BR significantly increased survival, enhanced bacterial phagocytosis, and decreased IL-6 and MIP-2 (a CXC chemokine) levels after CLP in outbred (ICR/CD-1) mice. During the CLP-induced septic response in A2BR knockout mice, hemodynamic parameters were improved compared with wild-type mice in addition to better survival and decreased plasma IL-6 levels. A2BR deficiency resulted in a dramatic 4-log reduction in peritoneal bacteria. The mechanism of these improvements was due to enhanced macrophage phagocytic activity without augmenting neutrophil phagocytosis of bacteria. Following ex vivo LPS stimulation, septic macrophages from A2BR knockout mice had increased IL-6 and TNF-α secretion compared with wild-type mice. A therapeutic intervention with A2BR blockade was studied by using a plasma biomarker to direct therapy to those mice predicted to die. Pharmacological blockade of A2BR even 32 h after the onset of sepsis increased survival by 65% in those mice predicted to die. Thus, even the late treatment with an A2BR antagonist significantly improved survival of mice (ICR/CD-1) that were otherwise determined to die according to plasma IL-6 levels. Our findings of enhanced bacterial clearance and host survival suggest that antagonism of A2BRs offers a therapeutic target to improve macrophage function in a late treatment protocol that improves sepsis survival.


Asunto(s)
Inmunosupresores/antagonistas & inhibidores , Inmunosupresores/metabolismo , Macrófagos/inmunología , Fagocitosis/inmunología , Receptor de Adenosina A2B/metabolismo , Sepsis/inmunología , Regulación hacia Arriba/inmunología , Animales , Antígenos CD1/biosíntesis , Ciego , Femenino , Interleucina-6/antagonistas & inhibidores , Interleucina-6/metabolismo , Ligadura , Macrófagos/microbiología , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Noqueados , Ratones Transgénicos , Fagocitosis/genética , Punciones , Receptor de Adenosina A2B/deficiencia , Receptor de Adenosina A2B/genética , Sepsis/genética , Sepsis/mortalidad , Tasa de Supervivencia , Regulación hacia Arriba/genética
10.
J Clin Invest ; 130(11): 5629-5637, 2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-32870821

RESUMEN

Hypoxia/HIF-1α- and extracellular adenosine/A2 adenosine receptor-mediated immunosuppression protects tissues from collateral damage by antipathogen immune cells. However, this mechanism also protects cancerous tissues by inhibiting antitumor immune cells in hypoxic and extracellular adenosine-rich tumors that are the most resistant to current therapies. Here, we explain a potentially novel, antiimmunosuppressive reasoning to justify strategies using respiratory hyperoxia and oxygenation agents in cancer treatment. Earlier attempts to use oxygenation of tumors as a monotherapy or to improve radiotherapy have failed because oxygenation protocols were not combined with immunotherapies of cancer. In contrast, the proposal for therapeutic use of antihypoxic oxygenation described here was motivated by the need to prevent the hypoxia/HIF-1α-driven accumulation of extracellular adenosine to (a) unleash antitumor immune cells from inhibition by intracellular cAMP and (b) prevent immunosuppressive transcription of cAMP response element- and hypoxia response element-containing immunosuppressive gene products (e.g., TGF-ß). Use of oxygenation agents together with inhibitors of the A2A adenosine receptor may be required to enable the most effective cancer immunotherapy. The emerging outcomes of clinical trials of cancer patients refractory to all other treatments provide support for the molecular and immunological mechanism-based approach to cancer immunotherapy described here.


Asunto(s)
Antineoplásicos/uso terapéutico , Hiperoxia , Inmunoterapia , Neoplasias , Oxígeno/uso terapéutico , Animales , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias/terapia
11.
Curr Opin Pharmacol ; 53: 84-90, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32841869

RESUMEN

The promising results of the first in-human clinical study using A2AR antagonists for treatment of renal cell carcinoma highlight two decades of research into the hypoxia-A2-adenosinergic pathway. Importantly, clinical responses have been observed in patients who previously progressed on anti-PD-1/PDL-1 therapy, emphasizing the clinical importance of targeting A2AR signaling in cancer immunotherapies. Recently, it has been shown that systemic oxygenation weakens all known stages of the hypoxia-A2-adenosinergic axis. Therefore, we advocate the clinical use of systemic oxygenation and oxygenation agents in combination with A2AR blockade to further improve cancer immunotherapies. This approach is expected to completely eliminate the upstream (hypoxia-HIF-1α) and downstream (adenosine-A2AR) stages of the immunosuppressive hypoxia-adenosinergic signaling axis. This might be a necessary strategy to maximize the therapeutic benefits of A2AR antagonists and increase susceptibility of tumors to cancer treatments.


Asunto(s)
Antagonistas del Receptor de Adenosina A2/uso terapéutico , Inmunoterapia , Neoplasias/terapia , Oxígeno/uso terapéutico , Receptor de Adenosina A2A/inmunología , Hipoxia Tumoral , 5'-Nucleotidasa/inmunología , Adenosina/inmunología , Animales , Antígenos CD/inmunología , Apirasa/inmunología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/inmunología , Tolerancia Inmunológica , Neoplasias/inmunología
12.
Front Immunol ; 11: 570041, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33117358

RESUMEN

The blockade of immunological negative regulators offered a novel therapeutic approach that revolutionized the immunotherapy of cancer. Still, a significant portion of patients fail to respond to anti-PD-1/PD-L1 and/or anti-CTLA-4 therapy or experience significant adverse effects. We propose that one of the major reasons that many patients do not respond to this form of therapy is due to the powerful physiological suppression mediated by hypoxia-adenosinergic signaling. Indeed, both inflamed and cancerous tissues are hypoxic and rich in extracellular adenosine, in part due to stabilization of the transcription factor hypoxia-inducible factor 1 alpha (HIF-1α). Adenosine signals through adenosine A2A receptors (A2AR) to suppress anti-tumor and anti-pathogen immune responses. Several classes of anti-hypoxia-A2AR therapeutics have been offered to refractory cancer patients, with A2AR blockers, inhibitors of adenosine-generating enzymes such as CD39 and CD73, and hypoxia-targeting drugs now reaching the clinical stage. Clinical results have confirmed preclinical observations that blockade of the hypoxia-adenosine-A2AR axis synergizes with inhibitors of immune checkpoints to induce tumor rejection. Thus, A2AR blockers provide a new hope for the majority of patients who are nonresponsive to current immunotherapeutic approaches including checkpoint blockade. Here, we discuss the discoveries that firmly implicate the A2AR as a critical and non-redundant biochemical negative regulator of the immune response and highlight the importance of targeting the hypoxia-adenosine-A2AR axis to manipulate anti-pathogen and anti-tumor immune responses.


Asunto(s)
Hipoxia/metabolismo , Terapia de Inmunosupresión , Inmunoterapia , Neoplasias/terapia , Receptor de Adenosina A2A/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Adenosina/metabolismo , Animales , Biomarcadores , Humanos , Terapia de Inmunosupresión/métodos , Terapia Molecular Dirigida , Neoplasias/etiología , Neoplasias/metabolismo , Transducción de Señal/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Resultado del Tratamiento
14.
PLoS One ; 12(11): e0187314, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29155844

RESUMEN

Human cancers are known to downregulate Major Histocompatibility Complex (MHC) class I expression thereby escaping recognition and rejection by anti-tumor T cells. Here we report that oxygen tension in the tumor microenvironment (TME) serves as an extrinsic cue that regulates antigen presentation by MHC class I molecules. In support of this view, hypoxia is shown to negatively regulate MHC expression in a HIF-dependent manner as evidenced by (i) lower MHC expression in the hypoxic TME in vivo and in hypoxic 3-dimensional (3D) but not 2-dimensional (2D) tumor cell cultures in vitro; (ii) decreased MHC in human renal cell carcinomas with constitutive expression of HIF due to genetic loss of von Hippel-Lindau (VHL) function as compared with isogenically paired cells with restored VHL function, and iii) increased MHC in tumor cells with siRNA-mediated knockdown of HIF. In addition, hypoxia downregulated antigen presenting proteins like TAP 1/2 and LMP7 that are known to have a dominant role in surface display of peptide-MHC complexes. Corroborating oxygen-dependent regulation of MHC antigen presentation, hyperoxia (60% oxygen) transcriptionally upregulated MHC expression and increased levels of TAP2, LMP2 and 7. In conclusion, this study reveals a novel mechanism by which intra-tumoral hypoxia and HIF can potentiate immune escape. It also suggests the use of hyperoxia to improve tumor cell-based cancer vaccines and for mining novel immune epitopes. Furthermore, this study highlights the advantage of 3D cell cultures in reproducing hypoxia-dependent changes observed in the TME.


Asunto(s)
Hipoxia de la Célula/inmunología , Genes MHC Clase I/inmunología , Factor 1 Inducible por Hipoxia/genética , Neoplasias Renales/inmunología , Transportador de Casetes de Unión a ATP, Subfamilia B, Miembro 2/genética , Transportador de Casetes de Unión a ATP, Subfamilia B, Miembro 2/inmunología , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Factor 1 Inducible por Hipoxia/inmunología , Neoplasias Renales/genética , Neoplasias Renales/patología , Oxígeno/metabolismo , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/inmunología , Linfocitos T/inmunología , Activación Transcripcional/genética , Activación Transcripcional/inmunología , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/inmunología
15.
Cell Rep ; 21(13): 3672-3680, 2017 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-29281817

RESUMEN

Rapidly evolving pathogens such as HIV or influenza can quickly mutate their antigenic profiles, reducing the efficacy of conventional vaccines. Despite this challenge, functionally required epitopes are highly conserved among heterologous viral strains and represent a key vulnerability that could be targeted during vaccine development. As the antigenicity of these conserved epitopes is frequently subdominant, there is a critical need for innovative vaccination strategies designed to target these neutralizing epitopes. Here, we immunized mice with antigens containing discrete immunodominant and subdominant moieties and show that treatment with soluble heterologous antigen bearing only the immunodominant epitope selectively suppresses these germinal center (GC) B cells. By exploiting this intrinsic tolerance mechanism, we promote the expansion of subdominant B cells in the GC and the subsequent long-lived components of the humoral response. We propose that this strategy may be applied to elicit preferential expansion of subdominant B cells that recognize weakly immunogenic epitopes on microbial pathogens.


Asunto(s)
Linfocitos B/metabolismo , Centro Germinal/metabolismo , Epítopos Inmunodominantes/metabolismo , Animales , Formación de Anticuerpos , Recuento de Células , Células Clonales , Ratones Endogámicos C57BL , Nitrofenoles/química , Ovalbúmina/inmunología , Fenilacetatos/química , Células Plasmáticas/metabolismo , Solubilidad
16.
Int J Med Chem ; 2017: 4852537, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29201461

RESUMEN

Antagonism of the adenosine A2A receptor on T cells blocks the hypoxia-adenosinergic pathway to promote tumor rejection. Using an in vivo immunoassay based on the Concanavalin A mouse model, a series of A2A antagonists were studied and identified preladenant as a potent lead compound for development. Molecular modeling was employed to assist drug design and subsequent synthesis of analogs and those of tozadenant, including fluorinated polyethylene glycol PEGylated derivatives. The efficacy of the analogs was evaluated using two in vitro functional bioassays, and compound 29, a fluorinated triethylene glycol derivative of preladenant, was confirmed as a potential immunotherapeutic agent.

17.
Curr Opin Pharmacol ; 29: 90-6, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27429212

RESUMEN

Hypoxic and adenosine rich tumor microenvironments represent an important barrier that must be overcome to enable T and NK cells to reject tumors. The A2A adenosine receptor (A2AR) on activated immune cells was identified as a critical and non-redundant mediator of physiological immunosuppression. Observations showing that tumor-protecting A2AR also suppress and redirect the anti-tumor immune response pointed to the importance of inhibiting this pathway to improve cancer immunotherapy. We advocated (i) blocking immunosuppressive adenosine-A2AR-cAMP-mediated intracellular signaling by A2AR antagonists and (ii) weakening hypoxia-HIF-1α-mediated accumulation of extracellular adenosine by oxygenation agents that also inhibits CD39/CD73 adenosine-generating enzymes. In view of commencing clinical trials of synthetic A2AR antagonists in combination with cancer immunotherapies, we discuss their promise and exclusion criteria.


Asunto(s)
Antagonistas del Receptor de Adenosina A2/farmacología , Inmunoterapia/métodos , Neoplasias/tratamiento farmacológico , Adenosina/metabolismo , Animales , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/inmunología , Tolerancia Inmunológica/inmunología , Células Asesinas Naturales/inmunología , Neoplasias/inmunología , Neoplasias/patología , Transducción de Señal/inmunología , Linfocitos T/inmunología , Microambiente Tumoral/inmunología
19.
Oncoimmunology ; 4(12): e1052934, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26587328

RESUMEN

Oxygenation of tumors weakens the tumor-protecting immunosuppressive signaling by A2A adenosine receptors in hypoxic and extracellular adenosine-rich microenvironments. This, in turn, unleashes the otherwise inhibited tumor-reactive T and natural killer (NK) cells. Oxygenation of tumors thus emerges as a novel checkpoint inhibitor of potential therapeutic value, but only in combination with cancer immunotherapies.

20.
Sci Transl Med ; 7(277): 277ra30, 2015 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-25739764

RESUMEN

Antitumor T cells either avoid or are inhibited in hypoxic and extracellular adenosine-rich tumor microenvironments (TMEs) by A2A adenosine receptors. This may limit further advances in cancer immunotherapy. There is a need for readily available and safe treatments that weaken the hypoxia-A2-adenosinergic immunosuppression in the TME. Recently, we reported that respiratory hyperoxia decreases intratumoral hypoxia and concentrations of extracellular adenosine. We show that it also reverses the hypoxia-adenosinergic immunosuppression in the TME. This, in turn, stimulates (i) enhanced intratumoral infiltration and reduced inhibition of endogenously developed or adoptively transfered tumor-reactive CD8 T cells, (ii) increased proinflammatory cytokines and decreased immunosuppressive molecules, such as transforming growth factor-ß (TGF-ß), (iii) weakened immunosuppression by regulatory T cells, and (iv) improved lung tumor regression and long-term survival in mice. Respiratory hyperoxia also promoted the regression of spontaneous metastasis from orthotopically grown breast tumors. These effects are entirely T cell- and natural killer cell-dependent, thereby justifying the testing of supplemental oxygen as an immunological coadjuvant to combine with existing immunotherapies for cancer.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Oxígeno/uso terapéutico , Adenosina/metabolismo , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Hiperoxia/complicaciones , Hiperoxia/patología , Hipoxia/complicaciones , Hipoxia/inmunología , Hipoxia/patología , Terapia de Inmunosupresión , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Metástasis de la Neoplasia , Neoplasias/patología , Oxígeno/farmacología , Inducción de Remisión , Respiración/efectos de los fármacos , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Microambiente Tumoral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA