Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 12: 729085, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34630403

RESUMEN

Recently, many discoveries have elucidated the cellular and molecular diversity in the leukemic microenvironment and improved our knowledge regarding their complex nature. This has allowed the development of new therapeutic strategies against leukemia. Advances in biotechnology and the current understanding of T cell-engineering have led to new approaches in this fight, thus improving cell-mediated immune response against cancer. However, most of the investigations focus only on conventional cytotoxic cells, while ignoring the potential of unconventional T cells that until now have been little studied. γδ T cells are a unique lymphocyte subpopulation that has an extensive repertoire of tumor sensing and may have new immunotherapeutic applications in a wide range of tumors. The ability to respond regardless of human leukocyte antigen (HLA) expression, the secretion of antitumor mediators and high functional plasticity are hallmarks of γδ T cells, and are ones that make them a promising alternative in the field of cell therapy. Despite this situation, in particular cases, the leukemic microenvironment can adopt strategies to circumvent the antitumor response of these lymphocytes, causing their exhaustion or polarization to a tumor-promoting phenotype. Intervening in this crosstalk can improve their capabilities and clinical applications and can make them key components in new therapeutic antileukemic approaches. In this review, we highlight several characteristics of γδ T cells and their interactions in leukemia. Furthermore, we explore strategies for maximizing their antitumor functions, aiming to illustrate the findings destined for a better mobilization of γδ T cells against the tumor. Finally, we outline our perspectives on their therapeutic applicability and indicate outstanding issues for future basic and clinical leukemia research, in the hope of contributing to the advancement of studies on γδ T cells in cancer immunotherapy.


Asunto(s)
Antineoplásicos Inmunológicos/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunoterapia Adoptiva , Linfocitos Intraepiteliales/efectos de los fármacos , Linfocitos Intraepiteliales/trasplante , Leucemia/terapia , Animales , Antineoplásicos Inmunológicos/efectos adversos , Proliferación Celular/efectos de los fármacos , Humanos , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Inmunoterapia Adoptiva/efectos adversos , Linfocitos Intraepiteliales/inmunología , Linfocitos Intraepiteliales/metabolismo , Leucemia/genética , Leucemia/inmunología , Leucemia/metabolismo , Activación de Linfocitos/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Fenotipo , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/metabolismo , Transducción de Señal , Microambiente Tumoral
2.
Front Immunol ; 12: 635076, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34025642

RESUMEN

Hyperforin is a major active constituent of Hypericum perforatum L. extract, which is widely used for the treatment of depressive disorders. Recent studies have reported that hyperforin reduced inflammation in stroke and suppressed proliferation and differentiation in keratinocytes. Psoriasis is a chronic immune-mediated inflammatory skin disease in which the IL-23/IL-17 axis plays an important role. To investigate the underlying inflammatory mechanisms and response of hyperforin in psoriasis, we use imiquimod (IMQ)-induced mice model, in vitro cultured murine splenic γδ T cells, and HaCaT cells in this study. Data showed that hyperforin reduced epidermal thickness and decreased IMQ-induced pathological scores of cutaneous skin lesions in mice. Meanwhile we proved that hyperforin suppressed infiltration of CD3+ T cells and downregulated expression of Il1, Il6, Il23, Il17a, Il22, antimicrobial peptides (AMPs) in the skin lesion. Hyperforin significantly inhibited imiquimod-induced splenomegaly, reduced serum levels of TNF-α and IL-6, and IL-17A in splenocytes and draining lymph nodes. Our study also suggested that hyperforin lessened the infiltration of γδ T cell and CCR6+ γδ T cells in spleen and lymph nodes. Hyperforin also suppressed the typical psoriasis-like inflammatory responses and the infiltration of IL-17A+ cells in dermal γδ T cells of IMQ treated Tcrd-/- mice transferred with γδ T cells. In vitro studies, hyperforin reduced the expression and secretion of IL-17A in γδ T cells, and suppressed the activation of MAPK/STAT3 pathways in human keratinocyte HaCaT cells and γδ T cells. In conclusion, hyperforin alleviates IMQ-induced inflammation in psoriasis through suppressing the immune responses exerted by IL-17 A-producing γδ T cells and related cytokines by modulating MAPK/STAT3 pathways. Our study provided a novel therapeutic tragedy for psoriasis by which hyperforin attenuates psoriasis-related inflammatory responses.


Asunto(s)
Antiinflamatorios/farmacología , Interleucina-17/metabolismo , Linfocitos Intraepiteliales/efectos de los fármacos , Floroglucinol/análogos & derivados , Psoriasis/prevención & control , Piel/efectos de los fármacos , Terpenos/farmacología , Traslado Adoptivo , Animales , Modelos Animales de Enfermedad , Genes Codificadores de los Receptores de Linfocitos T , Células HaCaT , Humanos , Imiquimod , Interleucina-17/genética , Linfocitos Intraepiteliales/inmunología , Linfocitos Intraepiteliales/metabolismo , Linfocitos Intraepiteliales/trasplante , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Floroglucinol/farmacología , Fosforilación , Psoriasis/inducido químicamente , Psoriasis/inmunología , Psoriasis/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Piel/inmunología , Piel/metabolismo
3.
J Immunother Cancer ; 9(5)2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33986124

RESUMEN

BACKGROUND: T cell-based immunotherapies using chimeric antigen receptors (CAR) or bispecific antibodies (BsAb) have produced impressive responses in hematological malignancies. However, major hurdles remained, including cytokine release syndrome, neurotoxicity, on-target off-tumor effects, reliance on autologous T cells, and failure in most solid tumors. BsAb armed T cells offer a safe alternative. METHODS: We generated ex vivo armed T cells (EATs) using IgG-[L]-scFv-platformed BsAb, where the anti-CD3 (huOKT3) scFv was attached to the light chain of a tumor-binding IgG. BsAb density on EAT, in vitro cytotoxicity, cytokine release, in vivo trafficking into tumors, and their antitumor activities were evaluated in multiple cancer cell lines and patient-derived xenograft mouse models. The efficacy of EATs after cryopreservation was studied, and gamma delta (γδ) T cells were investigated as unrelated alternative effector T cells. RESULTS: The antitumor potency of BsAb armed T cells was substantially improved using the IgG-[L]-scFv BsAb platform. When compared with separate BsAb and T cell injection, EATs released less TNF-α, and infiltrated tumors faster, while achieving robust antitumor responses. The in vivo potency of EAT therapy depended on BsAb dose for arming, EAT cell number per injection, total number of EAT doses, and treatment schedule intensity. The antitumor efficacy of EATs was preserved following cryopreservation, and EATs using γδ T cells were safe and as effective as αß T cell-EATs. CONCLUSIONS: EATs exerted potent antitumor activities against a broad spectrum of human cancer targets with remarkable safety. The antitumor potency of EATs depended on BsAb dose, cell number and total dose, and schedule. EATs were equally effective after cryopreservation, and the feasibility of third-party γδ-EATs offered an alternative for autologous T cell sources.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Citocinas/metabolismo , Inmunoterapia Adoptiva , Linfocitos Intraepiteliales/trasplante , Activación de Linfocitos , Linfocitos Infiltrantes de Tumor/trasplante , Neoplasias/terapia , Animales , Anticuerpos Biespecíficos/genética , Anticuerpos Biespecíficos/metabolismo , Línea Celular Tumoral , Movimiento Celular , Técnicas de Cocultivo , Citotoxicidad Inmunológica , Humanos , Linfocitos Intraepiteliales/inmunología , Linfocitos Intraepiteliales/metabolismo , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Masculino , Ratones Endogámicos BALB C , Ratones Noqueados , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/metabolismo , Fenotipo , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Scand J Immunol ; 93(3): e12996, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33205443

RESUMEN

Intestinal T cells form a central part of the front-line defence against foreign organisms and need to be situated in the mucosa where infection occurs. It is well accepted that immunization by a mucosal route favours localization of antigen-specific effector T cells in the mucosal epithelium, while systemic immunization does not. The aim of the study is to determine how homing receptors are specifically involved in retaining effector T cells in the small intestine after oral immunization. We here demonstrate that the chemokine receptor CXCR6, integrins ß7 and CD29 contribute differentially to the epithelial retention phenotype of CD8+ T cells in the small intestine of mice. CD8+ intraepithelial lymphocytes (IELs) of unvaccinated mice are predominantly ß7 single positives, and subcutaneous immunization-induced antigen-specific CD8+ effector IELs are mainly composed of CXCR6+ , CD29+ and CXCR6+ CD29+ cells. Strikingly, the majority of oral immunization-induced antigen-specific CD8+ effector IELs exhibit a distinct, tissue-specific CXCR6+ ß7+ double-positive phenotype, cytotoxic potential and enhanced intraepithelial localization. Transfer of antigen-specific CD8+ T cells preactivated with certain immuno-stimuli (such as monophosphoryl lipid A) results in increased accumulation of donor IELs with the CXCR6+ ß7+ phenotype. As ß7 exclusively paired with αE on IELs, our results strongly suggest that CXCR6 may cooperate with the heterodimer αEß7 to preferentially retain intestinally induced effector IELs in the epithelium. The identification of this novel IEL phenotype has significant implications for the development of vaccines and therapeutic strategies to enhance gut immunity.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Cadenas beta de Integrinas/metabolismo , Intestino Delgado/inmunología , Linfocitos Intraepiteliales/inmunología , Receptores CXCR6/metabolismo , Traslado Adoptivo , Animales , Linfocitos T CD8-positivos/trasplante , Integrina beta1/metabolismo , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Intestino Delgado/citología , Linfocitos Intraepiteliales/trasplante , Listeria monocytogenes/inmunología , Activación de Linfocitos/inmunología , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/metabolismo , Ratones , Ratones Endogámicos C57BL , Vacunación
5.
World J Gastroenterol ; 26(20): 2559-2569, 2020 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-32523311

RESUMEN

γδT cells are unconventional T lymphocytes that bridge innate and adaptive immunity. Based on the composition of T cell receptor and the cytokines produced, γδT cells can be divided into diverse subsets that may be present at different locations, including the liver, epithelial layer of the gut, the dermis and so on. Many of these cells perform specific functions in liver diseases, such as viral hepatitis, autoimmune liver diseases, non-alcoholic fatty liver disease, liver cirrhosis and liver cancers. In this review, we discuss the distribution, subsets, functions of γδT cells and the relationship between the microbiota and γδT cells in common hepatic diseases. As γδT cells have been used to cure hematological and solid tumors, we are interested in γδT cell-based immunotherapies to treat liver diseases.


Asunto(s)
Microbioma Gastrointestinal/inmunología , Inmunoterapia/métodos , Linfocitos Intraepiteliales/inmunología , Hepatopatías/inmunología , Animales , Modelos Animales de Enfermedad , Humanos , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Linfocitos Intraepiteliales/trasplante , Hígado/inmunología , Hígado/patología , Hepatopatías/microbiología , Hepatopatías/patología , Hepatopatías/terapia
6.
Cancer Immunol Immunother ; 69(12): 2571-2587, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32588076

RESUMEN

As ideal cells that can be used for adoptive cell therapy, γδ T cells are a group of homogeneous cells with high proliferative and tumor killing ability. However, γδ T cells are apt to apoptosis and show decreased cytotoxicity under persistent stimulation in vitro and cannot aggregate at tumor sites efficiently in vivo, both of which are two main obstacles to tumor adoptive immunotherapy. In this study, we found that the immune checkpoint T-cell immunoglobulin domain and mucin domain 3 (TIM-3) were up-regulated significantly on γδ T cells during their ex vivo expansion and this up-regulation contributed to the dysfunction of γδ T cells. Although the killing ability of γδ T cells against breast cancer cells which exhibited a high level of epithelial cell adhesion molecule (EpCAM) was enhanced, the level of TIM-3 on γδ T cells was also further up-regulated under the application of the bispecific antibody MT110 (anti-CD3 × anti-EpCAM) which can redirect T cells to target cells. Besides, these γδ T cells with up-regulated TIM-3 exhibited an increased susceptibility to apoptosis. By reinvigorating dysfunctional γδ T cells and promoting them to accumulate at tumor sites, the combined use of TIM-3 inhibitor and MT110 could further enhance the anti-tumor effect of the adoptively transfused γδ T cells. These results may have clinical implications for the design of new translational anti-tumor regimens aimed at combining checkpoint blockade and immune cell redirection.


Asunto(s)
Neoplasias de la Mama/terapia , Receptor 2 Celular del Virus de la Hepatitis A/antagonistas & inhibidores , Inmunoterapia Adoptiva/métodos , Linfocitos Intraepiteliales/trasplante , Anticuerpos de Cadena Única/farmacología , Adulto , Animales , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/sangre , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Células Cultivadas , Técnicas de Cocultivo , Terapia Combinada/métodos , Molécula de Adhesión Celular Epitelial/metabolismo , Femenino , Voluntarios Sanos , Receptor 2 Celular del Virus de la Hepatitis A/metabolismo , Humanos , Linfocitos Intraepiteliales/efectos de los fármacos , Linfocitos Intraepiteliales/inmunología , Linfocitos Intraepiteliales/metabolismo , Ratones , Persona de Mediana Edad , Cultivo Primario de Células , Anticuerpos de Cadena Única/uso terapéutico , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
8.
JCI Insight ; 4(24)2019 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-31689241

RESUMEN

γδ T cells account for a large fraction of human intestinal intraepithelial lymphocytes (IELs) endowed with potent antitumor activities. However, little is known about their origin, phenotype, and clinical relevance in colorectal cancer (CRC). To determine γδ IEL gut specificity, homing, and functions, γδ T cells were purified from human healthy blood, lymph nodes, liver, skin, and intestine, either disease-free, affected by CRC, or generated from thymic precursors. The constitutive expression of NKp46 specifically identifies a subset of cytotoxic Vδ1 T cells representing the largest fraction of gut-resident IELs. The ontogeny and gut-tropism of NKp46+/Vδ1 IELs depends both on distinctive features of Vδ1 thymic precursors and gut-environmental factors. Either the constitutive presence of NKp46 on tissue-resident Vδ1 intestinal IELs or its induced expression on IL-2/IL-15-activated Vδ1 thymocytes are associated with antitumor functions. Higher frequencies of NKp46+/Vδ1 IELs in tumor-free specimens from CRC patients correlate with a lower risk of developing metastatic III/IV disease stages. Additionally, our in vitro settings reproducing CRC tumor microenvironment inhibited the expansion of NKp46+/Vδ1 cells from activated thymic precursors. These results parallel the very low frequencies of NKp46+/Vδ1 IELs able to infiltrate CRC, thus providing insights to either follow-up cancer progression or to develop adoptive cellular therapies.


Asunto(s)
Neoplasias Colorrectales/inmunología , Mucosa Intestinal/citología , Linfocitos Intraepiteliales/inmunología , Receptor 1 Gatillante de la Citotoxidad Natural/metabolismo , Linfocitos T Citotóxicos/inmunología , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Animales , Antígenos Ly/metabolismo , Colon/citología , Colon/inmunología , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/terapia , Progresión de la Enfermedad , Femenino , Humanos , Íleon/citología , Íleon/inmunología , Inmunoterapia Adoptiva/métodos , Mucosa Intestinal/inmunología , Linfocitos Intraepiteliales/metabolismo , Linfocitos Intraepiteliales/trasplante , Masculino , Ratones , Persona de Mediana Edad , Estadificación de Neoplasias , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Globulina de Unión a Hormona Sexual , Linfocitos T Citotóxicos/metabolismo , Linfocitos T Citotóxicos/trasplante , Adulto Joven
9.
Expert Opin Biol Ther ; 19(9): 887-895, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31220420

RESUMEN

Introduction: Cancer immunotherapy relies on the development of an efficient and long-lasting anti-tumor response, generally mediated by cytotoxic T cells. γδ T cells possess distinctive features that justify their use in cancer immunotherapy. Areas covered: Here we will review our current knowledge on the functions of human γδ T cells that may be relevant in tumor immunity and the most recent advances in our understanding of how these functions are regulated in the tumor microenvironment. We will also discuss the major achievements and limitations of γδ T cell-based immunotherapy of cancer. Expert opinion: Several small-scale clinical trials have been conducted in cancer patients using either in vivo activation of γδ T cells or adoptive transfer of ex vivo-expanded γδ T cells. Both strategies are safe and give some clinical benefit to patients, thus providing a proof of principle for their utilization in addition to conventional therapies. However, low objective response rates have been obtained in both settings and therefore larger and well-controlled trials are needed. Discovering the factors which influence the success of γδ T cell-based immunotherapy will lead to a better understanding of their mechanism of action and to harness these cells for effective and durable anti-tumor responses.


Asunto(s)
Inmunoterapia Adoptiva , Linfocitos Intraepiteliales/inmunología , Neoplasias/terapia , Animales , Humanos , Linfocitos Intraepiteliales/trasplante , Neoplasias/inmunología , Microambiente Tumoral
10.
J Immunother Cancer ; 7(1): 36, 2019 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-30736852

RESUMEN

BACKGROUND: Cholangiocarcinoma (CCA) is a highly aggressive and fatal tumor. CCA occurs in the epithelial cells of bile ducts. Due to increasing incidences, CCA accounts for 3% of all gastrointestinal malignancies. In addition to comprehensive treatments for cancer, such as surgery, chemotherapy, and radiotherapy, during the past few years, cellular immunotherapy has played an increasingly important role. As a result of our research, we have discovered the γδ T cell-based immunotherapy for CCA. CASE PRESENTATION: A 30-year-old male ( https://www.clinicaltrials.gov/ ID: NCT02425735) was diagnosed with recurrent mediastinal lymph node metastasis after liver transplantation because of Cholangiocarcinoma (stage IV). In the course of his therapy sessions, he only received allogenic γδ T cell immunotherapy from August, 2017 through February, 2018 (8 infusions in total). γδ T cells were expanded from peripheral blood mononuclear cells (PBMCs) of healthy donor, and ~ 4 × 108 cells were adoptive transferred to the patient. CONCLUSION: In the above case report of the Cholangiocarcinoma (stage IV) patient who had received liver transplantation and afterward was diagnosed with recurrent mediastinal lymph node metastasis, we clinically proved that allogenic γδ T cell treatment had no adverse effects. We observed that allogenic γδ T cell treatments positively regulated peripheral immune functions of the patient, depleted tumor activity, improved quality of life, and prolonged his life span. After 8 γδ T cell treatments, the size of lymph nodes was remarkably reduced with activity depletion. This clinical work suggested that allogenic γδ T cell immunotherapy could be developed into a promising therapy drug for CCA.


Asunto(s)
Neoplasias de los Conductos Biliares/terapia , Colangiocarcinoma/terapia , Inmunoterapia , Linfocitos Intraepiteliales/trasplante , Adulto , Neoplasias de los Conductos Biliares/inmunología , Neoplasias de los Conductos Biliares/patología , Colangiocarcinoma/inmunología , Colangiocarcinoma/patología , Humanos , Trasplante de Hígado , Ganglios Linfáticos/patología , Metástasis Linfática/patología , Masculino
11.
Methods Mol Biol ; 1884: 57-72, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30465195

RESUMEN

Recent clinical trials have yielded promising results suggesting that γδ T cell62-based immunotherapies can be effective against hematological malignancies. Human T cells expressing Vγ9Vδ2+ receptors are particularly attractive candidates for this application, since they can be readily expanded in vitro in large quantities for adoptive transfer and do not require HLA-matching of donors and recipients. While it is well established that Vγ9Vδ2+ T cells are potently cytolytic against many human cancers and it has been shown that they can control transplanted human tumors in xenogeneic model systems, little is known about the parameters that determine the antitumor efficacy of adoptively transferred Vγ9Vδ2+ T cells in physiologically relevant scenarios. In particular, it may be important to separate their immunosurveillance functions from those employed in the context of an established tumor. Moreover, it is critical to understand how the presence of an immunosuppressive environment, such as one where tumor-infiltrating T cells are held in check by inhibitory ligands, affects the functions of Vγ9Vδ2+ T cells. This chapter describes how to establish Epstein-Barr virus (EBV) infection of human umbilical cord blood mononuclear cells (CBMCs) within immunodeficient mice, so as to drive the in vivo formation of human B cell lymphomas that contain an immunosuppressive environment. Details are provided on how to expand human Vγ9Vδ2+ T cells from peripheral blood mononuclear cells (PBMCs), administer them to the mice, and evaluate tumors and other tissues.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Inmunoterapia Adoptiva/métodos , Linfocitos Intraepiteliales/trasplante , Neoplasias/terapia , Animales , Técnicas de Cultivo de Célula/instrumentación , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Sangre Fetal/citología , Células HEK293 , Herpesvirus Humano 4/inmunología , Humanos , Linfocitos Intraepiteliales/inmunología , Linfocitos Intraepiteliales/virología , Ratones , Ratones Endogámicos NOD , Neoplasias/inmunología , Neoplasias/patología , Neoplasias/virología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Quimera por Trasplante/inmunología
12.
J Immunother Cancer ; 5: 9, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28239463

RESUMEN

BACKGROUND: Human γδ T cells expressing Vγ2Vδ2 T cell receptors monitor foreign- and self-prenyl pyrophosphate metabolites in isoprenoid biosynthesis to mediate immunity to microbes and tumors. Adoptive immunotherapy with Vγ2Vδ2 T cells has been used to treat cancer patients with partial and complete remissions. Most clinical trials and preclinical studies have used continuous zoledronate exposure to expand Vγ2Vδ2 cells where zoledronate is slowly diluted over the course of the culture. Zoledronate inhibits farnesyl diphosphate synthase (FDPS) in monocytes causing isopentenyl pyrophosphate to accumulate that then stimulates Vγ2Vδ2 cells. Because zoledronate inhibition of FDPS is also toxic for T cells, we hypothesized that a short period of exposure would reduce T cell toxicity but still be sufficient for monocytes uptake. Additionally, IL-15 increases the anti-tumor activity of murine αß T cells in mice but its effect on the in vivo anti-tumor activity of human Vγ2Vδ2 cells has not been assessed. METHODS: Human Vγ2Vδ2 T cells were expanded by pulse or continuous zoledronate stimulation with IL-2 or IL-15. Expanded Vγ2Vδ2 cells were tested for their expression of effector molecules and killing of tumor cells as well as their in vivo control of human prostate cancer tumors in immunodeficient NSG mice. RESULTS: Pulse zoledronate stimulation with either IL-2 or IL-15 resulted in more uniform expansion of Vγ2Vδ2 cells with higher purity and cell numbers as compared with continuous exposure. The Vγ2Vδ2 cells had higher levels of CD107a and perforin and increased tumor cytotoxicity. Adoptive immunotherapy with Vγ2Vδ2 cells derived by pulse stimulation controlled human PC-3 prostate cancer tumors in NSG mice significantly better than those derived by continuous stimulation, halting tumor growth. Although pulse zoledronate stimulation with IL-15 preserved early memory subsets, adoptive immunotherapy with IL-15-derived Vγ2Vδ2 cells equally inhibited PC-3 tumor growth as those derived with IL-2. CONCLUSIONS: Pulse zoledronate stimulation maximizes the purity, quantity, and quality of expanded Vγ2Vδ2 cells for adoptive immunotherapy but there is no advantage to using IL-15 over IL-2 in our humanized mouse model. Pulse zoledronate stimulation is a simple modification to existing protocols that will enhance the effectiveness of adoptively transferred Vγ2Vδ2 cells by increasing their numbers and anti-tumor activity.


Asunto(s)
Difosfonatos/farmacología , Imidazoles/farmacología , Inmunoterapia Adoptiva , Interleucina-15/farmacología , Interleucina-2/farmacología , Linfocitos Intraepiteliales/trasplante , Neoplasias/terapia , Adulto , Animales , Línea Celular Tumoral , Femenino , Humanos , Linfocitos Intraepiteliales/efectos de los fármacos , Ratones , Neoplasias/patología , Carga Tumoral/efectos de los fármacos , Ácido Zoledrónico
13.
Oncotarget ; 8(4): 5954-5964, 2017 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-27992381

RESUMEN

Multiple myeloma (MM) is an incurable hematological malignancy, although bortezomib has markedly improved its outcomes. Growing clinical evidence indicates that enhancing induced natural killer (NK) or γδ T cells for infusion is useful in the treatment of MM. However, whether combination treatment with bortezomib and induced NK and γδ T cells further improves outcomes in MM, and how the treatments should be combined, remain unclear. Herein, we found that low-dose bortezomib did not suppress the viability of induced NK and γδ T cells, but did induce MM cell apoptosis. Importantly, low-dose bortezomib increased the expression of NKG2D and DNAM-1 ligands on MM cells, which sensitized the multiple myeloma cells to lysis by induced NK and γδ T cells. Our results suggested that combination treatment with low-dose bortezomib and induced NK or γδ T cells had a synergistic cytotoxic effect on MM cells. This study provided a proof of principle for the design of future trials and investigation of this combination therapeutic strategy for MM treatment.


Asunto(s)
Antígenos de Diferenciación de Linfocitos T/metabolismo , Antineoplásicos/farmacología , Bortezomib/farmacología , Linfocitos Intraepiteliales/citología , Células Asesinas Naturales/citología , Mieloma Múltiple/metabolismo , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Anciano , Línea Celular Tumoral , Supervivencia Celular , Terapia Combinada , Relación Dosis-Respuesta a Droga , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Linfocitos Intraepiteliales/efectos de los fármacos , Linfocitos Intraepiteliales/trasplante , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/trasplante , Ligandos , Activación de Linfocitos/efectos de los fármacos , Masculino , Persona de Mediana Edad , Mieloma Múltiple/terapia , Regulación hacia Arriba
14.
J Neurooncol ; 129(2): 231-41, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27393349

RESUMEN

Nitrogen-containing bisphosphonates (N-BPs), which prevent bone resorption, exert direct and γδT cell (GDT)-mediated antitumor effects against several tumor cell types, including glioblastoma (GBM). However, limited information is available regarding the antitumor effects of N-BPs in GBM. Specifically, the antitumor effects of minodronate (MDA), a third-generation N-BP, in GBM are yet unclear. This study aimed to investigate the antitumor effects of MDA in GBM in vitro and in vivo. We performed growth inhibition and apoptosis detection assays using the GBM cell lines U87MG and U138MG. Apoptosis inhibition assays were also conducted. In vivo xenograft assays were performed in highly immunodeficient NOD.Cg-Prkdc(scid) Il2rg(tm1Sug)/Jic mice subcutaneously implanted with U87MG and U138MG cells. Growth inhibition and apoptosis detection assays demonstrated that MDA inhibited GBM cell growth via apoptosis, which was markedly enhanced by ex vivo expanded GDT. A pan-caspase inhibitor, z-VAD-fmk, inhibited MDA-induced U138MG apoptosis and MDA/GDT-induced U87MG and U138MG apoptosis. But z-VAD-fmk increased MDA-induced U87MG apoptosis. MDA/GDT-mediated apoptosis was blocked by the anti-T cell receptor (TCR) Vγ9, mevalonate pathway inhibitor, granzyme B inhibitor, and antitumor necrosis factor (TNF)-α. In vivo xenograft assays showed that combined intraperitoneal administration of MDA/GDT induced antitumor effects on unestablished U87MG-derived subcutaneous tumors. MDA exerted direct and GDT-mediated anti-GBM apoptotic effects in a caspase-dependent manner. GDT recognized MDA-exposed GBM cells via TCRVγ9 and induced apoptosis via granzyme B and TNF-α release. Because MDA elicited anti-GBM effects in synergy with GDT in vivo, a combination of MDA and ex vivo-generated GDT could be an effective treatment in patients with GBM.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/terapia , Difosfonatos/uso terapéutico , Glioblastoma/terapia , Imidazoles/uso terapéutico , Linfocitos Intraepiteliales/fisiología , Linfocitos Intraepiteliales/trasplante , Clorometilcetonas de Aminoácidos/farmacología , Animales , Anexina A5/metabolismo , Apoptosis/efectos de los fármacos , Inhibidores de Caspasas/farmacología , Recuento de Células , Línea Celular Tumoral , Proliferación Celular , Difosfonatos/farmacología , Femenino , Humanos , Masculino , Ratones Endogámicos NOD , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA