Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Analyst ; 146(4): 1430-1443, 2021 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-33410834

RESUMEN

Selective turn-on luminescence properties are shown by a non-luminescent metalloreceptor upon the addition of phosphate anions in CH3CN and hydrazine in CH3CN/H2O (6/4, v/v). The non-luminescent metalloreceptors [RuII(phen)2(TpH)]2PF6- (RtpH) and [RuII(Phen)2(TpI)]2PF6- (RtpI) {phen = 1,10-phenanthroline; TpH = 2-(2-(4-(pyridin-2-yl)-1H-1,2,3-triazol-1-yl)ethyl)isoindoline-1,3-dione; and TpI = 2-(2-(5-iodo-4-(pyridin-2-yl)-1H-1,2,3-triazol-1-yl)ethyl)isoindoline-1,3-dione} were synthesized and characterized. Both metalloreceptors have excellent sensing properties for phosphate anions (H2PO4- and H2P2O72-) over other anions in CH3CN. The limit of detection (LOD) values were calculated to be 79 nM and 48 nM for H2PO4- upon addition to RtpH and RtpI, respectively. Noncovalent interactions play a key role in the sensing of phosphate anions, among which the halogen-anion interaction showed superior recognition properties over the hydrogen-anion interaction. Comparative electrochemical experiments, 1H NMR titration, 31P NMR titration, and lifetime studies also show that RtpI has better sensing properties, as evidenced by its more drastic emission response to H2PO4- anions compared with RtpH. Moreover, the metalloreceptors showed a remarkable fluorescence increase (at ∼584 nm) upon the addition of hydrazine, without the interference of other amines in CH3CN/H2O (6/4, v/v). Interestingly, fluorescence enhancement was observed within live HeLa cells upon hydrazine addition, which is caused by the efficient photoinduced electron transfer process.


Asunto(s)
Fosfatos , Rutenio , Aniones , Células HeLa , Humanos , Hidrazinas , Ligandos , Ftalimidas , Piridinas , Triazoles
2.
Cell Mol Life Sci ; 77(9): 1745-1770, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-31690961

RESUMEN

Tumor vascularization occurs through several distinct biological processes, which not only vary between tumor type and anatomic location, but also occur simultaneously within the same cancer tissue. These processes are orchestrated by a range of secreted factors and signaling pathways and can involve participation of non-endothelial cells, such as progenitors or cancer stem cells. Anti-angiogenic therapies using either antibodies or tyrosine kinase inhibitors have been approved to treat several types of cancer. However, the benefit of treatment has so far been modest, some patients not responding at all and others acquiring resistance. It is becoming increasingly clear that blocking tumors from accessing the circulation is not an easy task to accomplish. Tumor vessel functionality and gene expression often differ vastly when comparing different cancer subtypes, and vessel phenotype can be markedly heterogeneous within a single tumor. Here, we summarize the current understanding of cellular and molecular mechanisms involved in tumor angiogenesis and discuss challenges and opportunities associated with vascular targeting.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Neoplasias/patología , Neovascularización Patológica/patología , Animales , Humanos , Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Transducción de Señal
3.
Semin Cancer Biol ; 45: 23-35, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28257957

RESUMEN

Cell therapy is an advanced form of cancer immunotherapy that has had remarkable clinical progress in the past decade in the search for cure of cancer. Most success has been achieved for chimeric antigen receptor (CAR) T-cells where CAR T-cells targeting CD19 show very high complete response rates for patients with refractory acute B-cell acute lymphoblastic leukemia (ALL) and are close to approval for this indication. CD19 CAR T-cells are also effective against B-cell chronic lymphoblastic leukemia (CLL) and B-cell lymphomas. Although encouraging, CAR T-cells have not yet proven clinically effective for solid tumors. This is mainly due to the lack of specific and homogenously expressed targets to direct the T-cells against and a hostile immunosuppressive tumor microenvironment in solid tumors. Cancer vaccines based on dendritic cells (DC) are also making progress although clinical efficacy is still lacking. The likelihood of success is however increasing now when individual tumors can be sequences and patient-specific neoepitopes identified. Neoepitopes and/or neoantigens can then be included in patient-based DC vaccines. This review discusses recent advancements of DC vaccines and CAR T-cells with emphasis on the cancer-immunity cycle, and current efforts to design novel cell therapies.


Asunto(s)
Antineoplásicos/uso terapéutico , Inmunidad/efectos de los fármacos , Neoplasias/inmunología , Neoplasias/terapia , Animales , Antígenos de Neoplasias/inmunología , Antineoplásicos/farmacología , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Tratamiento Basado en Trasplante de Células y Tejidos , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Diseño de Fármacos , Ingeniería Genética , Terapia Genética , Humanos , Inmunidad/genética , Inmunomodulación/efectos de los fármacos , Inmunomodulación/genética , Inmunoterapia/efectos adversos , Inmunoterapia/métodos , Neoplasias/genética , Neoplasias/metabolismo , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
5.
J Immunol ; 193(5): 2287-96, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25049358

RESUMEN

Helicobacter pylori neutrophil-activating protein (HP-NAP) is a major virulence factor involved in H. pylori infection. Both HP-NAP protein and oncolytic viruses encoding HP-NAP have been suggested as immunotherapeutic anticancer agents and adjuvants for vaccination but with little known about its mode of action to activate adaptive immunity. Dendritic cells (DCs) are key players in bridging innate and adaptive immune responses, and in this study we aim to evaluate the effect of HP-NAP on DC maturation, migration, and induction of adaptive immune response. Maturation markers CD83, CD80, CD86, HLA-DR, CD40, and CCR7 were upregulated on human DCs after treatment with supernatants from HP-NAP adenovirus-infected cells. HP-NAP-activated DCs had a Th1 cytokine secretion profile, with high IL-12 and relatively low IL-10 secretion, and migrated toward CCL19. Ag-specific T cells were efficiently expanded by Ag-presenting HP-NAP-activated DCs, which is an important property of functionally mature DCs. Furthermore, intradermal injections of HP-NAP-encoding adenovirus in C57BL/6 mice enhanced resident DC migration to draining lymph nodes, which was verified by imaging lymph nodes by two-photon microscopy and by phenotyping migrating cells by flow cytometry. In conclusion, therapeutic effects of HP-NAP are mediated by maturation of DCs and subsequent activation of Ag-specific T cells in addition to provoking innate immunity.


Asunto(s)
Adenoviridae , Proteínas Bacterianas/inmunología , Movimiento Celular/inmunología , Células Dendríticas/inmunología , Vectores Genéticos , Helicobacter pylori/inmunología , Células TH1/inmunología , Animales , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/inmunología , Proteínas Bacterianas/genética , Línea Celular Tumoral , Movimiento Celular/genética , Células Dendríticas/citología , Helicobacter pylori/genética , Humanos , Inmunidad Innata/genética , Interleucina-10/genética , Interleucina-10/inmunología , Interleucina-12/genética , Interleucina-12/inmunología , Ratones , Células TH1/citología
6.
BMC Cancer ; 14: 30, 2014 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-24438073

RESUMEN

BACKGROUND: Adoptive transfer of T cells genetically engineered with a chimeric antigen receptor (CAR) has successfully been used to treat both chronic and acute lymphocytic leukemia as well as other hematological cancers. Experimental therapy with CAR-engineered T cells has also shown promising results on solid tumors. The prostate stem cell antigen (PSCA) is a protein expressed on the surface of prostate epithelial cells as well as in primary and metastatic prostate cancer cells and therefore a promising target for immunotherapy of prostate cancer. METHODS: We developed a third-generation CAR against PSCA including the CD28, OX-40 and CD3 ζ signaling domains. T cells were transduced with a lentivirus encoding the PSCA-CAR and evaluated for cytokine production (paired Student's t-test), proliferation (paired Student's t-test), CD107a expression (paired Student's t-test) and target cell killing in vitro and tumor growth and survival in vivo (Log-rank test comparing Kaplan-Meier survival curves). RESULTS: PSCA-CAR T cells exhibit specific interferon (IFN)-γ and interleukin (IL)-2 secretion and specific proliferation in response to PSCA-expressing target cells. Furthermore, the PSCA-CAR-engineered T cells efficiently kill PSCA-expressing tumor cells in vitro and systemic treatment with PSCA-CAR-engineered T cells significantly delays subcutaneous tumor growth and prolongs survival of mice. CONCLUSIONS: Our data confirms that PSCA-CAR T cells may be developed for treatment of prostate cancer.


Asunto(s)
Antígenos de Neoplasias/inmunología , Terapia Genética/métodos , Inmunoterapia Adoptiva/métodos , Proteínas de Neoplasias/inmunología , Neoplasias de la Próstata/terapia , Receptores de Antígenos de Linfocitos T/biosíntesis , Linfocitos T/trasplante , Animales , Degranulación de la Célula , Línea Celular Tumoral , Proliferación Celular , Técnicas de Cocultivo , Citocinas/metabolismo , Citotoxicidad Inmunológica , Proteínas Ligadas a GPI/inmunología , Humanos , Proteína 1 de la Membrana Asociada a los Lisosomas/metabolismo , Masculino , Ratones , Ratones Desnudos , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Receptores de Antígenos de Linfocitos T/genética , Proteínas Recombinantes de Fusión/biosíntesis , Linfocitos T/inmunología , Linfocitos T/metabolismo , Factores de Tiempo , Transducción Genética , Transfección , Carga Tumoral
7.
Mol Ther ; 21(11): 2008-18, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23817216

RESUMEN

Helicobacter pylori neutrophil-activating protein (HP-NAP) is a major virulence factor involved in H. pylori infection. HP-NAP can mediate antitumor effects by recruiting neutrophils and inducing Th1-type differentiation in the tumor microenvironment. It therefore holds strong potential as a therapeutic gene. Here, we armed a replication-selective, infection-enhanced adenovirus with secretory HP-NAP, Ad5PTDf35-[Δ24-sNAP], and evaluated its therapeutic efficacy against neuroendocrine tumors. We observed that it could specifically infect and eradicate a wide range of tumor cells lines from different origin in vitro. Insertion of secretory HP-NAP did not affect the stability or replicative capacity of the virus and infected tumor cells could efficiently secrete HP-NAP. Intratumoral administration of the virus in nude mice xenografted with neuroendocrine tumors improved median survival. Evidence of biological HP-NAP activity was observed 24 hours after treatment with neutrophil infiltration in tumors and an increase of proinflammatory cytokines such as tumor necrosis factor (TNF)-α and MIP2-α in the systemic circulation. Furthermore, evidence of Th1-type immune polarization was observed as a result of increase in IL-12/23 p40 cytokine concentrations 72 hours postvirus administration. Our observations suggest that HP-NAP can serve as a potent immunomodulator in promoting antitumor immune response in the tumor microenvironment and enhance the therapeutic effect of oncolytic adenovirus.


Asunto(s)
Adenoviridae/genética , Proteínas Bacterianas/uso terapéutico , Tumores Neuroendocrinos/terapia , Virus Oncolíticos/genética , Adenoviridae/metabolismo , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Citocinas/metabolismo , Femenino , Terapia Genética , Vectores Genéticos , Humanos , Ratones , Ratones Desnudos , Tumores Neuroendocrinos/inmunología , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Viroterapia Oncolítica , Virus Oncolíticos/metabolismo , Recombinación Genética , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Factores de Virulencia/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Spectrochim Acta A Mol Biomol Spectrosc ; 305: 123483, 2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-37804708

RESUMEN

BACKGROUND: Nucleoside polyphosphate (NPP) anions are important for enzymatic activity and should be monitored by scientists in industry and medicine. By elucidating enzyme kinetics and processes, it aids in the discovery of effective inhibitors and activators. Nucleoside polyphosphate (NPP) anions are used by kinases, GTPases, and glycosyltransferases (GTs). Phosphorylation of certain amino acid residues (Ser, Thr, and Tyr) on proteins requires the breakdown of ATP by protein kinases, which produces ADP. Protein kinases, breakdown of ATP, and NPP are the focus of oncology drug development because the aberrant control of kinase activity is a common cause of cancer. RESULTS: However, a discriminative turn-on fluorescent property is exhibited by non-fluorescent p-tertbutylcalix[4]arene modified 1,2,3-triazole containing bis-ruthenium polypyridyl complex (RL) upon the addition of phosphate anions such as (dihydrogen pyrophosphate (H2P2O72-) and dihydrogen phosphate (H2PO4-)) in CH3CN solvent and Adenosine Diphosphate (ADP) in CH3CN/HEPES (pH = 7.4) buffer (9/1, v/v). The probe RL shows a better-recognizing ability with pyrophosphate anion (H2P2O72-) than dihydrogen phosphate anion (H2PO4-). With H2P2O72- and H2PO4- anions, the RL detection limit was calculated to be as low as 83 nM and 198 nM, respectively. SIGNIFICANCE: The calix[4]arene macrocycle's excellent size and binding cone conformation make it a good host-guest interface for the pyrophosphate anion and ADP. The bis-ruthenium polypyridyl complex's connection to the p-tertbutyl calix[4]arene moiety creates the ADP selectivity turn-on sensor. When moving from mono-nuclear to bi-nuclear ruthenium complex anchored on p-tertbutyl calix[4]arene, the probe can differentiate ADP, ATP, and AMP. Furthermore, this platform is a great resource for creating devices to simultaneously assess phosphate anions in environmental samples.


Asunto(s)
Fosfatos , Rutenio , Fosfatos/química , Difosfatos , Rutenio/química , Nucleósidos , Aniones/química , Adenosina Difosfato , Adenosina Trifosfato , Proteínas Quinasas
9.
Front Immunol ; 14: 1275378, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37954592

RESUMEN

Tertiary lymphoid structures (TLS) are lymph node-like aggregates that can form in association with chronic inflammation or cancer. Mature TLS are organized into B and T cell zones, and are not encapsulated but include all cell types necessary for eliciting an adaptive immune response. TLS have been observed in various cancer types and are generally associated with a positive prognosis as well as increased sensitivity to cancer immunotherapy. However, a comprehensive understanding of the roles of TLS in eliciting anti-tumor immunity as well as the mechanisms involved in their formation and function is still lacking. Further studies in orthotopic, immunocompetent cancer models are necessary to evaluate the influence of TLS on cancer therapies, and to develop new treatments that promote their formation in cancer. Here, we review key insights obtained from functional murine studies, discuss appropriate models that can be used to study cancer-associated TLS, and suggest guidelines on how to identify TLS and distinguish them from other antigen-presenting niches.


Asunto(s)
Neoplasias , Estructuras Linfoides Terciarias , Animales , Ratones , Neoplasias/terapia , Neoplasias/patología , Pronóstico , Ganglios Linfáticos/metabolismo , Inflamación/patología
10.
Nat Commun ; 14(1): 4732, 2023 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-37563127

RESUMEN

Chimeric antigen receptor (CAR)-T cell therapy is rapidly advancing as cancer treatment, however, designing an optimal CAR remains challenging. A single-chain variable fragment (scFv) is generally used as CAR targeting moiety, wherein the complementarity-determining regions (CDRs) define its specificity. We report here that the CDR loops can cause CAR clustering, leading to antigen-independent tonic signalling and subsequent CAR-T cell dysfunction. We show via CARs incorporating scFvs with identical framework and varying CDR sequences that CARs may cluster on the T cell surface, which leads to antigen-independent CAR-T cell activation, characterized by increased cell size and interferon (IFN)-γ secretion. This results in CAR-T cell exhaustion, activation-induced cell death and reduced responsiveness to target-antigen-expressing tumour cells. CDR mutagenesis confirms that the CAR-clustering is mediated by CDR-loops. In summary, antigen-independent tonic signalling can be induced by CDR-mediated CAR clustering, which could not be predicted from the scFv sequences, but could be tested for by evaluating the activity of unstimulated CAR-T cells.


Asunto(s)
Regiones Determinantes de Complementariedad , Anticuerpos de Cadena Única , Regiones Determinantes de Complementariedad/genética , Regiones Determinantes de Complementariedad/metabolismo , Linfocitos T , Inmunoterapia Adoptiva/métodos , Transducción de Señal , Línea Celular Tumoral , Receptores de Antígenos de Linfocitos T/metabolismo
11.
Clin Cancer Res ; 29(20): 4139-4152, 2023 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-37540566

RESUMEN

PURPOSE: Although CD19 chimeric antigen receptor T cells (CAR-T) therapy has shown remarkable success in B-cell malignancies, a substantial fraction of patients do not obtain a long-term clinical response. This could be influenced by the quality of the individual CAR-T infusion product. To shed some light on this, clinical outcome was correlated to characteristics of CAR-T infusion products. PATIENTS AND METHODS: In this phase II study, patients with B-cell lymphoma (n = 23) or leukemia (n = 1) received one or two infusions of third-generation CD19-directed CAR-Ts (2 × 108/m2). The clinical trial was registered at clinicaltrials.gov: NCT03068416. We investigated the transcriptional profile of individual CD19 CAR-T infusion products using targeted single-cell RNA sequencing and multicolor flow cytometry. RESULTS: Two CAR-T infusions were not better than one in the settings used in this study. As for the CAR-T infusion products, we found that effector-like CD8+CAR-Ts with a high polyfunctionality, high cytotoxic and cytokine production profile, and low dysfunctional signature were associated with clinical response. An extended ex vivo expansion time during CAR-T manufacturing negatively influenced the proportion of effector CD8+CAR-Ts in the infusion product. CONCLUSIONS: We identified cell-intrinsic characteristics of effector CD8+CAR-Ts correlating with response that could be used as an indicator for clinical outcome. The results in the study also serve as a guide to CAR-T manufacturing practices.

12.
Cancer Cell ; 41(6): 1134-1151.e10, 2023 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-37172581

RESUMEN

Glioblastomas are aggressive brain tumors that are largely immunotherapy resistant. This is associated with immunosuppression and a dysfunctional tumor vasculature, which hinder T cell infiltration. LIGHT/TNFSF14 can induce high endothelial venules (HEVs) and tertiary lymphoid structures (TLS), suggesting that its therapeutic expression could promote T cell recruitment. Here, we use a brain endothelial cell-targeted adeno-associated viral (AAV) vector to express LIGHT in the glioma vasculature (AAV-LIGHT). We found that systemic AAV-LIGHT treatment induces tumor-associated HEVs and T cell-rich TLS, prolonging survival in αPD-1-resistant murine glioma. AAV-LIGHT treatment reduces T cell exhaustion and promotes TCF1+CD8+ stem-like T cells, which reside in TLS and intratumoral antigen-presenting niches. Tumor regression upon AAV-LIGHT therapy correlates with tumor-specific cytotoxic/memory T cell responses. Our work reveals that altering vascular phenotype through vessel-targeted expression of LIGHT promotes efficient anti-tumor T cell responses and prolongs survival in glioma. These findings have broader implications for treatment of other immunotherapy-resistant cancers.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Glioma , Ratones , Animales , Glioma/genética , Glioma/terapia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/irrigación sanguínea , Glioblastoma/genética , Fenotipo , Encéfalo , Microambiente Tumoral
14.
Nat Biomed Eng ; 6(7): 830-841, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35379957

RESUMEN

Chimeric antigen receptor T cells (CAR T cells) are effective against haematologic malignancies. However, in solid tumours, their potency is hampered by local immunosuppression and by the heterogeneous expression of the antigen that the CAR targets. Here we show that CAR T cells expressing a pluripotent pro-inflammatory neutrophil-activating protein (NAP) from Helicobacter pylori trigger endogenous bystander T-cell responses against solid cancers. In mice with subcutaneous murine pancreatic ductal adenocarcinomas, neuroblastomas or colon carcinomas, CAR(NAP) T cells led to slower tumour growth and higher survival rates than conventional mouse CAR T cells, regardless of target antigen, tumour type and host haplotype. In tumours with heterogeneous antigen expression, NAP secretion induced the formation of an immunologically 'hot' microenvironment that supported dendritic cell maturation and bystander responses, as indicated by epitope spreading and infiltration of cytotoxic CD8+ T cells targeting tumour-associated antigens other than the CAR-targeted antigen. CAR T cells armed with NAP neither increased off-tumour toxicity nor hampered the efficacy of CAR T cells, and hence may have advantageous translational potential.


Asunto(s)
Inmunoterapia Adoptiva , Neoplasias Pancreáticas , Animales , Antígenos de Neoplasias/metabolismo , Linfocitos T CD8-positivos , Ratones , Neoplasias Pancreáticas/metabolismo , Microambiente Tumoral , Factores de Virulencia/metabolismo
15.
Cancer Gene Ther ; 29(11): 1628-1635, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35596069

RESUMEN

There is a great demand for improved oncolytic viruses that selectively replicate within cancer cells while sparing normal cells. Here, we describe a novel oncolytic adenovirus, Ixovex-1, that obtains a cancer-selective replication phenotype by modulating the level of expression of the different, alternatively spliced E1B mRNA isoforms. Ixovex-1 is a recombinant adenovirus that carries a single point mutation in the E1B-93R 3' splice acceptor site that results in overexpression of the E1B-156R splice isoform. In this paper, we studied the characteristics of this novel oncolytic adenovirus by validating its in vitro behaviour in a panel of normal cells and cancer cells. We additionally studied its anti-tumour efficacy in vivo. Ixovex-1 significantly inhibited tumour growth and prolonged survival of mice in an immune-deficient lung carcinoma tumour implantation model. In complementation experiments, overexpression of E1B-156R was shown to increase the oncolytic index of both Ad5wt and ONYX-015. In contrast to prior viruses of similar type, Ixovex-1 includes a functional E3B region for better in vivo efficacy. Throughout this study, the Ixovex-1 virus has been proven to be superior in competency compared to a virus with multiple deletions.


Asunto(s)
Infecciones por Adenoviridae , Neoplasias , Viroterapia Oncolítica , Virus Oncolíticos , Ratones , Animales , Adenoviridae/genética , Adenoviridae/metabolismo , Proteínas E1B de Adenovirus/genética , Proteínas E1B de Adenovirus/metabolismo , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Virus Oncolíticos/metabolismo , Neoplasias/genética , Neoplasias/terapia , Replicación Viral/genética
16.
Neuro Oncol ; 24(3): 398-411, 2022 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-34347079

RESUMEN

BACKGROUND: Tumor vessels in glioma are molecularly and functionally abnormal, contributing to treatment resistance. Proteins differentially expressed in glioma vessels can change vessel phenotype and be targeted for therapy. ELTD1 (Adgrl4) is an orphan member of the adhesion G-protein-coupled receptor family upregulated in glioma vessels and has been suggested as a potential therapeutic target. However, the role of ELTD1 in regulating vessel function in glioblastoma is poorly understood. METHODS: ELTD1 expression in human gliomas and its association with patient survival was determined using tissue microarrays and public databases. The role of ELTD1 in regulating tumor vessel phenotype was analyzed using orthotopic glioma models and ELTD1-/- mice. Endothelial cells isolated from murine gliomas were transcriptionally profiled to determine differentially expressed genes and pathways. The consequence of ELTD1 deletion on glioma immunity was determined by treating tumor-bearing mice with PD-1-blocking antibodies. RESULTS: ELTD1 levels were upregulated in human glioma vessels, increased with tumor malignancy, and were associated with poor patient survival. Progression of orthotopic gliomas was not affected by ELTD1 deletion, however, tumor vascular function was improved in ELTD1-/- mice. Bioinformatic analysis of differentially expressed genes indicated increased inflammatory response and decreased proliferation in tumor endothelium in ELTD1-/- mice. Consistent with an enhanced inflammatory response, ELTD1 deletion improved T-cell infiltration in GL261-bearing mice after PD-1 checkpoint blockade. CONCLUSION: Our data demonstrate that ELTD1 participates in inducing vascular dysfunction in glioma, and suggest that targeting of ELTD1 may normalize the vessels and improve the response to immunotherapy.


Asunto(s)
Neoplasias Encefálicas , Glioma , Receptores Acoplados a Proteínas G/genética , Animales , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Células Endoteliales/metabolismo , Eliminación de Gen , Glioma/tratamiento farmacológico , Glioma/patología , Humanos , Ratones , Ratones Noqueados , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Linfocitos T/metabolismo
17.
Front Immunol ; 12: 724739, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34539661

RESUMEN

Glioblastoma is the most common and aggressive brain tumor, which is uniformly lethal due to its extreme invasiveness and the absence of curative therapies. Immune checkpoint inhibitors have not yet proven efficacious for glioblastoma patients, due in part to the low prevalence of tumor-reactive T cells within the tumor microenvironment. The priming of tumor antigen-directed T cells in the cervical lymph nodes is complicated by the shortage of dendritic cells and lack of appropriate lymphatic vessels within the brain parenchyma. However, recent data suggest that naive T cells may also be primed within brain tumor-associated tertiary lymphoid structures. Here, we review the current understanding of the formation of these structures within the central nervous system, and hypothesize that promotion of tertiary lymphoid structures could enhance priming of tumor antigen-targeted T cells and sensitize glioblastomas to cancer immunotherapy.


Asunto(s)
Neoplasias Encefálicas/inmunología , Sistema Nervioso Central/inmunología , Glioblastoma/inmunología , Linfocitos T/inmunología , Estructuras Linfoides Terciarias/inmunología , Animales , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Sistema Nervioso Central/patología , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Humanos , Inmunoterapia , Estructuras Linfoides Terciarias/patología , Microambiente Tumoral
18.
Mol Ther Oncolytics ; 21: 356-366, 2021 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-34141872

RESUMEN

Oncolytic viruses (OVs) represent promising therapeutic agents for cancer therapy by selective oncolysis and induction of anti-tumor immunity. OVs can be engineered to express tumor-associated antigens and immune-modulating agents to provoke stronger antitumor immunity. Here, we engineered vaccinia virus (VV) and Semliki Forest virus (SFV) to express neuroblastoma-associated antigen disialoganglioside (GD2) and the immune modulator Helicobacter pylori neutrophil-activating protein (NAP) and compared their therapeutic potency. Oncolytic VV did not exhibit any antitumor benefits, whereas SFV was able to delay subcutaneous neuroblastoma (NXS2) tumor growth. Additional expression of the GD2 mimotope (GD2m) by VV-GD2m or SFV-GD2m did not improve their anti-tumor capacity compared to the parent viruses. Further arming these OVs with NAP resulted in contrasting anti-tumor efficacy. VV (VV-GD2m-NAP) significantly improved therapeutic efficacy compared to VV-GD2m, which was also associated with a significantly elevated anti-GD2 antibody, whereas there was no additive antitumor efficacy for SFV-GD2m-NAP compared to SFV-GD2m, nor was the anti-GD2 antibody response improved. Instead, NAP induced higher neutralizing antibodies against SFV. These observations suggest that distinct immune stimulation profiles are elicited when the same immunostimulatory factor is expressed by different OVs. Therefore, careful consideration and detailed characterization are needed when engineering OVs with immune-modulators.

19.
Mol Ther Oncolytics ; 21: 37-46, 2021 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-33869741

RESUMEN

Oncolytic virotherapy holds promise of effective immunotherapy against otherwise nonresponsive cancers such as glioblastoma. Our previous findings have shown that although oncolytic Semliki Forest virus (SFV) is effective against various mouse glioblastoma models, its therapeutic potency is hampered by type I interferon (IFN-I)-mediated antiviral signaling. In this study, we constructed a novel IFN-I-resistant SFV construct, SFV-AM6, and evaluated its therapeutic potency in vitro, ex vivo, and in vivo in the IFN-I competent mouse GL261 glioma model. In vitro analysis shows that SFV-AM6 causes immunogenic apoptosis in GL261 cells despite high IFN-I signaling. MicroRNA-124 de-targeted SFV-AM6-124T selectively replicates in glioma cells, and it can infect orthotopic GL261 gliomas when administered intraperitoneally. The combination of SFV-AM6-124T and anti-programmed death 1 (PD1) immunotherapy resulted in increased immune cell infiltration in GL261 gliomas, including an increased tumor-reactive CD8+ fraction. Our results show that SFV-AM6-124T can overcome hurdles of innate anti-viral signaling. Combination therapy with SFV-AM6-124T and anti-PD1 promotes the inflammatory response and improves the immune microenvironment in the GL261 glioma model.

20.
Nat Commun ; 12(1): 4127, 2021 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-34226552

RESUMEN

Gliomas are brain tumors characterized by an immunosuppressive microenvironment. Immunostimulatory agonistic CD40 antibodies (αCD40) are in clinical development for solid tumors, but are yet to be evaluated for glioma. Here, we demonstrate that systemic delivery of αCD40 in preclinical glioma models induces the formation of tertiary lymphoid structures (TLS) in proximity of meningeal tissue. In treatment-naïve glioma patients, the presence of TLS correlates with increased T cell infiltration. However, systemic delivery of αCD40 induces hypofunctional T cells and impairs the response to immune checkpoint inhibitors in pre-clinical glioma models. This is associated with a systemic induction of suppressive CD11b+ B cells post-αCD40 treatment, which accumulate in the tumor microenvironment. Our work unveils the pleiotropic effects of αCD40 therapy in glioma and reveals that immunotherapies can modulate TLS formation in the brain, opening up for future opportunities to regulate the immune response.


Asunto(s)
Antígenos CD40/inmunología , Glioma/tratamiento farmacológico , Estructuras Linfoides Terciarias/inmunología , Animales , Antineoplásicos/farmacología , Linfocitos B/inmunología , Neoplasias Encefálicas/tratamiento farmacológico , Antígeno CD11b , Línea Celular Tumoral , Citocinas , Femenino , Expresión Génica , Glioma/patología , Humanos , Inmunoglobulina G/genética , Inmunoterapia , Masculino , Ratones , Ratones Endogámicos C57BL , Células Mieloides , Fenotipo , Linfocitos T , Microambiente Tumoral/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA