Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
J Neurooncol ; 151(1): 21-28, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33398531

RESUMEN

PURPOSE: CNS malignancies are currently the most common cause of disease related deaths in children. Although brainstem gliomas are invariably fatal cancers in children, clinical studies against this disease are limited. This review is to lead to a succinct collection of knowledge of known biological mechanisms of this disease and discuss available therapeutics. METHODS: A hallmark of brainstem gliomas are mutations in the histone H3.3 with the majority of cases expressing the mutation K27M on histone 3.3. Recent studies using whole genome sequencing have revealed other mutations associated with disease. Current standard clinical practice may merely involve radiation and/or chemotherapy with little hope for long term survival. Here we discuss the potential of new therapies. CONCLUSION: Despite the lack of treatment options using frequently practiced clinical techniques, immunotherapeutic strategies have recently been developed to target brainstem gliomas. To target brainstem gliomas, investigators are evaluating the use of broad non-targeted therapy with immune checkpoint inhibitors. Alternatively, others have begun to explore adoptive T cell strategies against these fatal malignancies.


Asunto(s)
Neoplasias del Tronco Encefálico , Glioma , Neoplasias del Tronco Encefálico/genética , Neoplasias del Tronco Encefálico/terapia , Glioma/genética , Glioma/terapia , Histonas/genética , Humanos , Mutación
2.
J Proteome Res ; 19(1): 424-431, 2020 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-31713431

RESUMEN

Parkinson's disease (PD) is characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta of the brain, as well as the degeneration of motor and nonmotor circuitries. The cause of neuronal death is currently unknown, although chronic neuroinflammation, aggregated α-synuclein, mitochondrial dysfunction, and oxidative stress have all been implicated. Gliosis has been shown to exacerbate neuroinflammation via secretion of proinflammatory cytokines, and there is a subsequent infiltration of T lymphocytes (T-cells), into the brain of PD patients. Using liquid chromatography-high-resolution mass spectrometry (LC-HRMS), we have observed metabolomic changes in stool samples, thought to be associated with the potential disease-modifying effect of immunotherapy administered to transgenic Parkinsonian (A53T) mice. Significant elevations (p < 0.05) in metabolites associated with immune response (taurine, histamine, and its methylated product, 3-methylhistamine) are identified as being higher in the mice undergoing immunotherapy. Furthermore, a reduction in triacylglycerol (TG) and diacylglycerol (DG) expressions in stool following immunotherapy suggests a regulation of lipid breakdown or biosynthesis with the vaccine. These "omics" markers (among others reported in this article) along with weight gain and increased life expectancy suggest that immunotherapy is positively modifying the disease state.


Asunto(s)
Heces/química , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/terapia , Animales , Peso Corporal , Cromatografía Líquida de Alta Presión/métodos , Modelos Animales de Enfermedad , Femenino , Inmunoterapia/métodos , Lipidómica , Lípidos/análisis , Espectrometría de Masas/métodos , Metabolómica , Ratones Transgénicos , Enfermedad de Parkinson/etiología
3.
Stem Cells ; 37(2): 166-175, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30353618

RESUMEN

Hematopoietic stem and progenitor cells (HSPCs) are the progenitor cells that can regenerate the entire blood compartment, including the immune system. Recent studies have unearthed considerable immune-modulating potential of these cells. They can migrate through chemotactic gradients, differentiate into functional immune cells, and crosstalk with immune cells during infections, autoimmune diseases, and cancers. Although the primary role of HSPCs during solid malignancies is considered immunosuppressive, recent studies have discovered immune-activating HSPCs and progeny. In this review, we will discuss the recent evidence that HSPCs act as immunomodulators during solid cancers and highlight the future directions of discovery. Stem Cells 2019;37:166-175.


Asunto(s)
Células Madre Hematopoyéticas/inmunología , Inmunidad/inmunología , Inmunoterapia/métodos , Neoplasias/inmunología , Células Madre/inmunología
4.
Mol Ther ; 27(4): 837-849, 2019 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-30448196

RESUMEN

With the presence of the blood-brain barrier (BBB), successful immunotherapeutic drug delivery to CNS malignancies remains a challenge. Immunomodulatory agents, such as cytokines, can reprogram the intratumoral microenvironment; however, systemic cytokine delivery has limited access to the CNS. To bypass the limitations of systemically administered cytokines, we investigated if RNA-modified T cells could deliver macromolecules directly to brain tumors. The abilities of T cells to cross the BBB and mediate direct cytotoxic killing of intracranial tumors make them an attractive tool as biological carriers. Using T cell mRNA electroporation, we demonstrated that activated T cells can be modified to secrete granulocyte macrophage colony-stimulating factor (GM-CSF) protein while retaining their inherent effector functions in vitro. GM-CSF RNA-modified T cells effectively delivered GM-CSF to intracranial tumors in vivo and significantly extended overall survival in an orthotopic treatment model. Importantly, GM-CSF RNA-modified T cells demonstrated superior anti-tumor efficacy as compared to unmodified T cells alone or in combination with systemic administration of recombinant GM-CSF. Anti-tumor effects were associated with increased IFN-γ secretion locally within the tumor microenvironment and systemic antigen-specific T cell expansion. These findings demonstrate that RNA-modified T cells may serve as a versatile platform for the effective delivery of biological agents to CNS tumors.


Asunto(s)
Neoplasias Encefálicas/terapia , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Inmunoterapia Adoptiva/métodos , ARN/genética , Linfocitos T/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Neoplasias Encefálicas/mortalidad , Línea Celular Tumoral , Supervivencia Celular/genética , Modelos Animales de Enfermedad , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Proteínas Fluorescentes Verdes/metabolismo , Interferón gamma/biosíntesis , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Transfección/métodos , Microambiente Tumoral/genética
5.
Neuro Oncol ; 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38990913

RESUMEN

Brain tumors, particularly glioblastoma (GBM), are devastating and challenging to treat, with a low 5-year survival rate of only 6.6%. Mouse models are established to understand tumorigenesis and develop new therapeutic strategies. Large-scale genomic studies have facilitated the identification of genetic alterations driving human brain tumor development and progression. Genetically engineered mouse models (GEMMs) with clinically relevant genetic alterations are widely used to investigate tumor origin. Additionally, syngeneic implantation models, utilizing cell lines derived from GEMMs or other sources, are popular for their consistent and relatively short latency period, addressing various brain cancer research questions. In recent years, the success of immunotherapy in specific cancer types has led to a surge in cancer immunology-related research which specifically necessitates the utilization of immunocompetent mouse models. In this review, we provide a comprehensive summary of GEMMs and syngeneic mouse models for adult brain tumors, emphasizing key features such as model origin, genetic alteration background, oncogenic mechanisms, and immune-related characteristics. Our review serves as a valuable resource for the brain tumor research community, aiding in the selection of appropriate models to study cancer immunology.

6.
Genome Med ; 16(1): 17, 2024 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-38268001

RESUMEN

BACKGROUND: Despite advancements in the successful use of immunotherapy in treating a variety of solid tumors, applications in treating brain tumors have lagged considerably. This is due, at least in part, to the lack of well-characterized antigens expressed within brain tumors that can mediate tumor rejection; the low mutational burden of these tumors that limits the abundance of targetable neoantigens; and the immunologically "cold" tumor microenvironment that hampers the generation of sustained and productive immunologic responses. The field of mRNA-based therapeutics has experienced a boon following the universal approval of COVID-19 mRNA vaccines. mRNA-based immunotherapeutics have also garnered widespread interest for their potential to revolutionize cancer treatment. In this study, we developed a novel and scalable approach for the production of personalized mRNA-based therapeutics that target multiple tumor rejection antigens in a single therapy for the treatment of refractory brain tumors. METHODS: Tumor-specific neoantigens and aberrantly overexpressed tumor-associated antigens were identified for glioblastoma and medulloblastoma tumors using our cancer immunogenomics pipeline called Open Reading Frame Antigen Network (O.R.A.N). Personalized tumor antigen-specific mRNA vaccine was developed for each individual tumor model using selective gene capture and enrichment strategy. The immunogenicity and efficacy of the personalized mRNA vaccines was evaluated in combination with anti-PD-1 immune checkpoint blockade therapy or adoptive cellular therapy with ex vivo expanded tumor antigen-specific lymphocytes in highly aggressive murine GBM models. RESULTS: Our results demonstrate the effectiveness of the antigen-specific mRNA vaccines in eliciting robust anti-tumor immune responses in GBM hosts. Our findings substantiate an increase in tumor-infiltrating lymphocytes characterized by enhanced effector function, both intratumorally and systemically, after antigen-specific mRNA-directed immunotherapy, resulting in a favorable shift in the tumor microenvironment from immunologically cold to hot. Capacity to generate personalized mRNA vaccines targeting human GBM antigens was also demonstrated. CONCLUSIONS: We have established a personalized and customizable mRNA-therapeutic approach that effectively targets a plurality of tumor antigens and demonstrated potent anti-tumor response in preclinical brain tumor models. This platform mRNA technology uniquely addresses the challenge of tumor heterogeneity and low antigen burden, two key deficiencies in targeting the classically immunotherapy-resistant CNS malignancies, and possibly other cold tumor types.


Asunto(s)
Neoplasias Encefálicas , Vacunas contra el Cáncer , Neoplasias Cerebelosas , Meduloblastoma , Humanos , Animales , Ratones , Vacunas de ARNm , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Vacunas contra el Cáncer/genética , Antígenos de Neoplasias/genética , Microambiente Tumoral/genética
7.
Cancer Immunol Immunother ; 62(11): 1649-62, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23982483

RESUMEN

Adoptive cell transfer (ACT) using ex vivo-expanded anti-tumor T cells such as tumor-infiltrated lymphocytes or genetically engineered T cells potently eradicates established tumors. However, these two approaches possess obvious limitations. Therefore, we established a novel methodology using total tumor RNA (ttRNA) to prime dendritic cells (DC) as a platform for the ex vivo generation of anti-tumor T cells. We evaluated the antigen-specific expansion and recognition of T cells generated by the ttRNA-DC-T platform, and directly modulated the differentiation status of these ex vivo-expanded T cells with a cytokine cocktail. Furthermore, we evaluated the persistence and in vivo anti-tumor efficacy of these T cells through murine xenograft and syngeneic tumor models. During ex vivo culture, IL-2 preferentially expanded CD4 subset, while IL-7 enabled homeostatic proliferation from the original precursors. T cells tended to lose CD62L during ex vivo culture using IL-2; however, IL-12 could maintain high levels of CD62L by increasing expression on effector T cells (Tem). In addition, we validated that OVA RNA-DC only selectively expanded T cells in an antigen-specific manner. A cytokine cocktail excluding the use of IL-2 greatly increased CD62Lhigh T cells which specifically recognized tumor cells, engrafted better in a xenograft model and exhibited superior anti-tumor activities in a syngeneic intracranial model. ACT using the ex vivo ttRNA-DC-T platform in conjunction with a cytokine cocktail generated potent CD62Lhigh anti-tumor T cells and imposes a novel T cell-based therapeutic with the potential to treat brain tumors and other cancers.


Asunto(s)
Citocinas/inmunología , Células Dendríticas/inmunología , Neoplasias/inmunología , Linfocitos T/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Citocinas/farmacología , Citotoxicidad Inmunológica/efectos de los fármacos , Citotoxicidad Inmunológica/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/trasplante , Femenino , Citometría de Flujo , Humanos , Inmunofenotipificación , Inmunoterapia Adoptiva/métodos , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Subunidad gamma Común de Receptores de Interleucina/genética , Selectina L/inmunología , Selectina L/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Neoplasias/patología , Neoplasias/terapia , ARN Neoplásico/inmunología , Linfocitos T/metabolismo , Linfocitos T/trasplante
8.
ACS Chem Neurosci ; 14(2): 235-245, 2023 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-36571847

RESUMEN

Aggregated α-synuclein, a major constituent of Lewy bodies plays a crucial role in the pathogenesis of α-synucleinopathies (SPs) such as Parkinson's disease (PD). PD is affected by the innate and adaptive arms of the immune system, and recently both active and passive immunotherapies targeted against α-synuclein are being trialed as potential novel treatment strategies. Specifically, dendritic cell-based vaccines have shown to be an effective treatment for SPs in animal models. Here, we report on the development of adoptive cellular therapy (ACT) for SP and demonstrate that adoptive transfer of pre-activated T-cells generated from immunized mice can improve survival and behavior, reduce brain microstructural impairment via magnetic resonance imaging (MRI), and decrease α-synuclein pathology burden in a peripherally induced preclinical SP model (M83) when administered prior to disease onset. This study provides preclinical evidence for ACT as a potential immunotherapy for LBD, PD and other related SPs, and future work will provide necessary understanding of the mechanisms of its action.


Asunto(s)
Enfermedad de Parkinson , Sinucleinopatías , Vacunas , Ratones , Animales , alfa-Sinucleína/genética , Sinucleinopatías/patología , Ratones Transgénicos , Enfermedad de Parkinson/terapia , Enfermedad de Parkinson/patología , Modelos Animales de Enfermedad
9.
J Immunother Cancer ; 11(2)2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36750252

RESUMEN

BACKGROUND: Glioma-induced immune dysregulation of the hematopoietic system has been described in a limited number of studies. In this study, our group further demonstrates that gliomas interrupt the cellular differentiation programming and outcomes of hematopoietic stem and progenitor cells (HSPCs) in the bone marrow. HSPCs from glioma-bearing mice are reprogrammed and driven towards expansion of myeloid lineage precursors and myeloid-derived suppressor cells (MDSCs) in secondary lymphoid organs. However, we found this expansion is reversed by immunotherapy. Adoptive cellular therapy (ACT) has been demonstrably efficacious in multiple preclinical models of central nervous system (CNS) malignancies, and here we describe how glioma-induced dysfunction is reversed by this immunotherapeutic platform. METHODS: The impact of orthotopic KR158B-luc glioma on HSPCs was evaluated in an unbiased fashion using single cell RNAseq (scRNAseq) of lineage- cells and phenotypically using flow cytometry. Mature myeloid cell frequencies and function were also evaluated using flow cytometry. Finally, ACT containing total body irradiation, tumor RNA-pulsed dendritic cells, tumor-reactive T cells and HSPCs isolated from glioma-bearing or non-tumor-bearing mice were used to evaluate cell fate differentiation and survival. RESULTS: Using scRNAseq, we observed an altered HSPC landscape in glioma-bearing versus non-tumor-bearing mice . In addition, an expansion of myeloid lineage subsets, including granulocyte macrophage precursors (GMPs) and MDSCs, were observed in glioma-bearing mice relative to non-tumor-bearing controls. Furthermore, MDSCs from glioma-bearing mice demonstrated increased suppressive capacity toward tumor-specific T cells as compared with MDSCs from non-tumor-bearing hosts. Interestingly, treatment with ACT overcame these suppressive properties. When HSPCs from glioma-bearing mice were transferred in the context of ACT, we observed significant survival benefit and long-term cures in orthotopic glioma models compared with mice treated with ACT using non-glioma-bearing HSPCs.


Asunto(s)
Neoplasias del Sistema Nervioso Central , Glioma , Ratones , Animales , Línea Celular Tumoral , Glioma/patología , Inmunoterapia , Células Madre Hematopoyéticas , Linfocitos T
10.
Hematol Oncol Clin North Am ; 36(1): 237-252, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34801163

RESUMEN

Glioblastoma has emerged as an immunotherapy-refractory tumor based on negative phase III studies of anti-programmed cell death-1 therapy among newly diagnosed as well as recurrent patients. In addition, although much work on vaccine and cellular approaches is ongoing, therapeutic benefit with these approaches has been underwhelming. Much scientific insight into the multitiered layers of immunosuppression exploited by glioblastoma tumors is emerging that sheds light on the explanation for the disappointing results to date and highlights possible therapeutic avenues that may offer a better likelihood of therapeutic benefit for immune-based therapies.


Asunto(s)
Neoplasias Encefálicas , Vacunas contra el Cáncer , Neoplasias del Sistema Nervioso Central , Glioblastoma , Neoplasias Encefálicas/terapia , Vacunas contra el Cáncer/uso terapéutico , Sistema Nervioso Central , Neoplasias del Sistema Nervioso Central/terapia , Glioblastoma/terapia , Humanos , Inmunoterapia , Recurrencia Local de Neoplasia
11.
Nat Cancer ; 3(1): 11-24, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35121998

RESUMEN

Pediatric central nervous system tumors are the most common solid malignancies in childhood, and aggressive therapy often leads to long-term sequelae in survivors, making these tumors challenging to treat. Immunotherapy has revolutionized prospects for many cancer types in adults, but the intrinsic complexity of treating pediatric patients and the scarcity of clinical studies of children to inform effective approaches have hampered the development of effective immunotherapies in pediatric settings. Here, we review recent advances and ongoing challenges in pediatric brain cancer immunotherapy, as well as considerations for efficient clinical translation of efficacious immunotherapies into pediatric settings.


Asunto(s)
Neoplasias Encefálicas , Neoplasias del Sistema Nervioso Central , Neoplasias Encefálicas/terapia , Neoplasias del Sistema Nervioso Central/terapia , Niño , Humanos , Factores Inmunológicos , Inmunoterapia/efectos adversos , Sobrevivientes
12.
Cells ; 10(5)2021 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-33919157

RESUMEN

Our understanding of the relationship between the immune system and cancers has undergone significant discovery recently. Immunotherapy with T cell therapies and checkpoint blockade has meaningfully changed the oncology landscape. While remarkable clinical advances in adaptive immunity are occurring, modulation of innate immunity has proven more difficult. The myeloid compartment, including macrophages, neutrophils, and dendritic cells, has a significant impact on the persistence or elimination of tumors. Myeloid cells, specifically in the tumor microenvironment, have direct contact with tumor tissue and coordinate with tumor-reactive T cells to either stimulate or antagonize cancer immunity. However, the myeloid compartment comprises a broad array of cells in various stages of development. In addition, hematopoietic stem and progenitor cells at various stages of myelopoiesis in distant sites undergo significant modulation by tumors. Understanding how tumors exert their influence on myeloid progenitors is critical to making clinically meaningful improvements in these pathways. Therefore, this review will cover recent developments in our understanding of how solid tumors modulate myelopoiesis to promote the formation of pro-tumor immature myeloid cells. Then, it will cover some of the potential avenues for capitalizing on these mechanisms to generate antitumor immunity.


Asunto(s)
Células Madre Hematopoyéticas/inmunología , Inmunoterapia/métodos , Células Mieloides/inmunología , Mielopoyesis/inmunología , Neoplasias/terapia , Microambiente Tumoral/inmunología , Animales , Células Madre Hematopoyéticas/citología , Humanos , Factores Inmunológicos , Células Mieloides/citología
13.
Expert Rev Neurother ; 21(2): 205-219, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33225764

RESUMEN

INTRODUCTION: Children with high-grade gliomas (pHGGs) represent a clinical population in substantial need of new therapeutic options given the inefficacy and toxicity of current standard-of-care modalities. Although immunotherapy has emerged as a promising modality, it has yet to elicit a significant survival benefit for pHGG patients. While preclinical studies address a variety of underlying challenges, translational clinical trial design and management also need to reflect the most updated progress and lessons from the field. AREAS COVERED: The authors will focus our discussion on the design of clinical trials, the management of potential toxicities, immune monitoring, and novel biomarkers. Clinical trial design should integrate appropriate patient populations, novel, and preclinically optimized trial design, and logical treatment combinations, particularly those which synergize with standard of care modalities. However, there are caveats due to the nature of immunotherapy trials, such as patient selection bias, evidenced by the frequent exclusion of patients on high-dose corticosteroids. Robust immune-modulating effects of modern immunotherapy can have toxicities. As such, it is important to understand and manage these, especially in pHGG patients. EXPERT OPINION: Adequate integration of these considerations should allow us to effectively gain insights on biological activity, safety, and biomarkers associated with benefits for patients.


Asunto(s)
Neoplasias Encefálicas , Glioma , Neoplasias Encefálicas/terapia , Niño , Glioma/terapia , Humanos , Inmunoterapia
14.
Clin Cancer Res ; 26(21): 5689-5700, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32788225

RESUMEN

PURPOSE: Immunotherapy has been demonstrably effective against multiple cancers, yet tumor escape is common. It remains unclear how brain tumors escape immunotherapy and how to overcome this immune escape. EXPERIMENTAL DESIGN: We studied KR158B-luc glioma-bearing mice during treatment with adoptive cellular therapy (ACT) with polyclonal tumor-specific T cells. We tested the immunogenicity of primary and escaped tumors using T-cell restimulation assays. We used flow cytometry and RNA profiling of whole tumors to further define escape mechanisms. To treat immune-escaped tumors, we generated escape variant-specific T cells through the use of escape variant total tumor RNA and administered these cells as ACT. In addition, programmed cell death protein-1 (PD-1) checkpoint blockade was studied in combination with ACT. RESULTS: Escape mechanisms included a shift in immunogenic tumor antigens, downregulation of MHC class I, and upregulation of checkpoint molecules. Polyclonal T cells specific for escape variants displayed greater recognition of escaped tumors than primary tumors. When administered as ACT, these T cells prolonged median survival of escape variant-bearing mice by 60%. The rational combination of ACT with PD-1 blockade prolonged median survival of escape variant glioma-bearing mice by 110% and was dependent upon natural killer cells and T cells. CONCLUSIONS: These findings suggest that the immune landscape of brain tumors are markedly different postimmunotherapy yet can still be targeted with immunotherapy.


Asunto(s)
Glioma/terapia , Receptor de Muerte Celular Programada 1/inmunología , Linfocitos T/inmunología , Escape del Tumor/efectos de los fármacos , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Antígenos de Neoplasias/inmunología , Línea Celular Tumoral , Glioma/genética , Glioma/inmunología , Glioma/patología , Xenoinjertos , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inmunoterapia Adoptiva/efectos adversos , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Ratones , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Linfocitos T/efectos de los fármacos , Escape del Tumor/inmunología , Microambiente Tumoral/efectos de los fármacos
15.
Neuro Oncol ; 21(6): 730-741, 2019 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-30668768

RESUMEN

BACKGROUND: The changes induced in host immunity and the tumor microenvironment by chemotherapy have been shown to impact immunotherapy response in both a positive and a negative fashion. Temozolomide is the most common chemotherapy used to treat glioblastoma (GBM) and has been shown to have variable effects on immune response to immunotherapy. Therefore, we aimed to determine the immune modulatory effects of temozolomide that would impact response to immune checkpoint inhibition in the treatment of experimental GBM. METHODS: Immune function and antitumor efficacy of immune checkpoint inhibition were tested after treatment with metronomic dose (MD) temozolomide (25 mg/kg × 10 days) or standard dose (SD) temozolomide (50 mg/kg × 5 days) in the GL261 and KR158 murine glioma models. RESULTS: SD temozolomide treatment resulted in an upregulation of markers of T-cell exhaustion such as LAG-3 and TIM-3 in lymphocytes which was not seen with MD temozolomide. When temozolomide treatment was combined with programmed cell death 1 (PD-1) antibody therapy, the MD temozolomide/PD-1 antibody group demonstrated a decrease in exhaustion markers in tumor infiltrating lymphocytes that was not observed in the SD temozolomide/PD-1 antibody group. Also, the survival advantage of PD-1 antibody therapy in a murine syngeneic intracranial glioma model was abrogated by adding SD temozolomide to treatment. However, when MD temozolomide was added to PD-1 inhibition, it preserved the survival benefit that was seen by PD-1 antibody therapy alone. CONCLUSION: The peripheral and intratumoral immune microenvironments are distinctively affected by dose modulation of temozolomide.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Neoplasias Encefálicas/inmunología , Glioblastoma/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Linfocitos T/inmunología , Temozolomida/farmacología , Animales , Antineoplásicos Alquilantes/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Humanos , Inmunoterapia , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Linfocitos T/efectos de los fármacos , Linfocitos T/patología , Células Tumorales Cultivadas , Microambiente Tumoral
16.
Nat Commun ; 10(1): 3029, 2019 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-31292444

RESUMEN

With improving biofabrication technology, 3D bioprinted constructs increasingly resemble real tissues. However, the fundamental principles describing how cell-generated forces within these constructs drive deformations, mechanical instabilities, and structural failures have not been established, even for basic biofabricated building blocks. Here we investigate mechanical behaviours of 3D printed microbeams made from living cells and extracellular matrix, bioprinting these simple structural elements into a 3D culture medium made from packed microgels, creating a mechanically controlled environment that allows the beams to evolve under cell-generated forces. By varying the properties of the beams and the surrounding microgel medium, we explore the mechanical behaviours exhibited by these structures. We observe buckling, axial contraction, failure, and total static stability, and we develop mechanical models of cell-ECM microbeam mechanics. We envision these models and their generalizations to other fundamental 3D shapes to facilitate the predictable design of biofabricated structures using simple building blocks in the future.


Asunto(s)
Bioimpresión/métodos , Técnicas de Cultivo de Célula/métodos , Impresión Tridimensional , Ingeniería de Tejidos/métodos , Resinas Acrílicas/química , Animales , Materiales Biocompatibles , Línea Celular Tumoral , Matriz Extracelular , Geles/química , Ensayo de Materiales , Metacrilatos/química , Ratones , Células 3T3 NIH
17.
Enferm. nefrol ; 26(2): 159-166, Abr-Jun 2023. tab, graf
Artículo en Español | IBECS (España) | ID: ibc-222845

RESUMEN

Introducción: Una de las funciones en la gestión de unidades de hemodiálisis, es evaluar la satisfacción del usuario sobre la atención que brinda el equipo de enfermería para identificar y corregir áreas deficitarias, y que además permite evaluar los resultados del cuidado que constituyen el eje de las presta-ciones asistenciales.Objetivo: Determinar la relación entre el nivel de satisfacción y la calidad de atención de salud percibida en pacientes con tratamiento de hemodiálisis de un hospital público de Perú.Material y Método: Estudio de enfoque cuantitativo, diseño no experimental, tipo descriptivo y correlacional de corte transversal. Se trabajó con una muestra representativa de pacientes en hemodiálisis, a quienes se les aplicó la encuesta SERVQUAL modificada para evaluar la calidad de atención; y un cuestionario de satisfacción al usuario validados con una confiabilidad Alfa de Cronbach de 0,84 y 0,91 respectiva-mente.Resultados: Se estudiaron 60 pacientes, con edad media de 56,6±15,4 años. El coeficiente Rho de Spearman encontró una correlación directa entre las variables calidad de aten-ción percibida y satisfacción del paciente en hemodiálisis (R=0,385, p=0,002). Por otro lado, el 58,3% de pacientes re-firieron un nivel de insatisfacción leve respecto a la atención que reciben, y las dimensiones por mejorar fueron seguridad y accesibilidad. Conclusión: Existe una asociación significativa entre las va-riables de estudio; es decir, que niveles altos de calidad de atención se corresponden con niveles altos de satisfacción en usuarios con tratamiento de hemodiálisis en un hospital público.(AU)


Introduction: One of the functions in the management of hemodialysis units is to evaluate user satisfaction regarding the care provided by the nursing team to identify and correct deficient areas, and which also makes it possible to evaluate the results of care that constitute the axis of the benefits. Objective: To determine the relationship between the level of satisfaction and the quality of health care in patients with hemodialysis treatment in a public hospital.Material and Method: study with a quantitative approach, non-experimental design, descriptive and correlational cross-sectional type. We worked with a representative sample of hemodialysis patients, to whom the SERVQUAL survey was applied to assess the quality of care; and a user satisfaction questionnaire validated with a Cronbach’s Alpha reliability of 0.84 and 0.91 respectively. Results: 60 patients were studied, with a mean age of 56.6±15.4 years. Spearman’s Rho coefficient (R=0.385) found a direct correlation between the quality of care variables and patient satisfaction in hemodialysis (p=0.002). On the other hand, 58.3% of patients reported a level of slight dissatisfaction regarding the care they receive, and the dimensions to be improved were safety and accessibility. Conclusion: There is a statistically significant correlation between the study variables, which implies that high levels of quality of care correspond to high levels of satisfaction in users with hemodialysis treatment in a public hospital.(AU)


Asunto(s)
Humanos , Masculino , Femenino , Persona de Mediana Edad , Diálisis Renal , Satisfacción del Paciente , Atención al Paciente , Calidad de la Atención de Salud , Terapia de Reemplazo Renal , Epidemiología Descriptiva , Estudios Transversales , Perú , Nefrología , Enfermedades Renales , 24960
18.
Nat Commun ; 9(1): 4313, 2018 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-30333482

RESUMEN

Immune checkpoint blockade using anti-PD-1 monoclonal antibodies has shown considerable promise in the treatment of solid tumors, but brain tumors remain notoriously refractory to treatment. In CNS malignancies that are completely resistant to PD-1 blockade, we found that bone marrow-derived, lineage-negative hematopoietic stem and progenitor cells (HSCs) that express C-C chemokine receptor type 2 (CCR2+) reverses treatment resistance and sensitizes mice to curative immunotherapy. HSC transfer with PD-1 blockade increases T-cell frequency and activation within tumors in preclinical models of glioblastoma and medulloblastoma. CCR2+HSCs preferentially migrate to intracranial brain tumors and differentiate into antigen-presenting cells within the tumor microenvironment and cross-present tumor-derived antigens to CD8+ T cells. HSC transfer also rescues tumor resistance to adoptive cellular therapy in medulloblastoma and glioblastoma. Our studies demonstrate a novel role for CCR2+HSCs in overcoming brain tumor resistance to PD-1 checkpoint blockade and adoptive cellular therapy in multiple invasive brain tumor models.


Asunto(s)
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Trasplante de Células Madre Hematopoyéticas , Inmunoterapia Adoptiva , Meduloblastoma/terapia , Animales , Neoplasias Encefálicas/inmunología , Diferenciación Celular , Movimiento Celular , Células Dendríticas/inmunología , Resistencia a Antineoplásicos , Femenino , Glioblastoma/inmunología , Activación de Linfocitos , Meduloblastoma/inmunología , Ratones Transgénicos , Linfocitos T/fisiología
19.
Clin Cancer Res ; 24(16): 3955-3966, 2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-29712687

RESUMEN

Purpose: Adoptive T-cell immunotherapy (ACT) has emerged as a viable therapeutic for peripheral and central nervous system (CNS) tumors. In peripheral cancers, optimal efficacy of ACT is reliant on dendritic cells (DCs) in the tumor microenvironment. However, the CNS is largely devoid of resident migratory DCs to function as antigen-presenting cells during immunotherapy. Herein, we demonstrate that cellular interactions between adoptively transferred tumor-reactive T cells and bone marrow-derived hematopoietic stem and progenitor cells (HSPCs) lead to the generation of potent intratumoral DCs within the CNS compartment.Experimental Design: We evaluated HSPC differentiation during ACT in vivo in glioma-bearing hosts and HSPC proliferation and differentiation in vitro using a T-cell coculture system. We utilized FACS, ELISAs, and gene expression profiling to study the phenotype and function of HSPC-derived cells ex vivo and in vivo To demonstrate the impact of HSPC differentiation and function on antitumor efficacy, we performed survival experiments.Results: Transfer of HSPCs with concomitant ACT led to the production of activated CD86+CD11c+MHCII+ cells consistent with DC phenotype and function within the brain tumor microenvironment. These intratumoral DCs largely supplanted abundant host myeloid-derived suppressor cells. We determined that during ACT, HSPC-derived cells in gliomas rely on T-cell-released IFNγ to differentiate into DCs, activate T cells, and reject intracranial tumors.Conclusions: Our data support the use of HSPCs as a novel cellular therapy. Although DC vaccines induce robust immune responses in the periphery, our data demonstrate that HSPC transfer uniquely generates intratumoral DCs that potentiate T-cell responses and promote glioma rejection in situClin Cancer Res; 24(16); 3955-66. ©2018 AACR.


Asunto(s)
Neoplasias del Sistema Nervioso Central/terapia , Glioma/terapia , Células Madre Hematopoyéticas/inmunología , Inmunoterapia Adoptiva , Animales , Antígeno B7-2/inmunología , Antígeno CD11c/inmunología , Diferenciación Celular/genética , Línea Celular Tumoral , Proliferación Celular/genética , Neoplasias del Sistema Nervioso Central/genética , Neoplasias del Sistema Nervioso Central/inmunología , Neoplasias del Sistema Nervioso Central/patología , Técnicas de Cocultivo , Células Dendríticas/inmunología , Células Dendríticas/trasplante , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica/genética , Glioma/genética , Glioma/inmunología , Glioma/patología , Células Madre Hematopoyéticas/metabolismo , Antígenos de Histocompatibilidad Clase II/inmunología , Humanos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Microambiente Tumoral/inmunología
20.
Oncoimmunology ; 6(1): e1256527, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28197373

RESUMEN

While RNA-pulsed dendritic cell (DC) vaccines have shown promise, the advancement of cellular therapeutics is fraught with developmental challenges. To circumvent the challenges of cellular immunotherapeutics, we developed clinically translatable nanoliposomes that can be combined with tumor-derived RNA to generate personalized tumor RNA-nanoparticles (NPs) with considerable scale-up capacity. RNA-NPs bypass MHC restriction, are amenable to central distribution, and can provide near immediate immune induction. We screened commercially available nanoliposomal preparations and identified the cationic lipid 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP) as an efficient mRNA courier to antigen-presenting cells (APCs). When administered intravenously, RNA-NPs mediate systemic activation of APCs in reticuloendothelial organs such as the spleen, liver, and bone marrow. RNA-NPs increase percent expression of MHC class I/II, B7 co-stimulatory molecules, and maturation markers on APCs (all vital for T-cell activation). RNA-NPs also increase activation markers on tumor APCs and elicit potent expansion of antigen-specific T-cells superior to peptide vaccines formulated in complete Freund's adjuvant. We demonstrate that both model antigen-encoding and physiologically-relevant tumor-derived RNA-NPs expand potent antitumor T-cell immunity. RNA-NPs were shown to induce antitumor efficacy in a vaccine model and functioned as a suitable alternative to DCs in a stringent cellular immunotherapy model for a radiation/temozolomide resistant invasive murine high-grade glioma. Although cancer vaccines have suffered from weak immunogenicity, we have advanced a RNA-NP formulation that systemically activates host APCs precipitating activated T-cell frequencies necessary to engender antitumor efficacy. RNA-NPs can thus be harnessed as a more feasible and effective immunotherapy to re-program host-immunity.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA