Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Cancers (Basel) ; 14(9)2022 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-35565467

RESUMEN

Under cellular distress, multiple facets of normal homeostatic signaling are altered or disrupted. In the context of the immune landscape, external and internal stressors normally promote the expression of natural killer group 2 member D (NKG2D) ligands that allow for the targeted recognition and killing of cells by NKG2D receptor-bearing effector populations. The presence or absence of NKG2D ligands can heavily influence disease progression and impact the accessibility of immunotherapy options. In cancer, tumor cells are known to have distinct regulatory mechanisms for NKG2D ligands that are directly associated with tumor progression and maintenance. Therefore, understanding the regulation of NKG2D ligands in cancer will allow for targeted therapeutic endeavors aimed at exploiting the stress response pathway. In this review, we summarize the current understanding of regulatory mechanisms controlling the induction and repression of NKG2D ligands in cancer. Additionally, we highlight current therapeutic endeavors targeting NKG2D ligand expression and offer our perspective on considerations to further enhance the field of NKG2D ligand biology.

2.
Sci Rep ; 11(1): 21133, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34702850

RESUMEN

Chemotherapeutic drugs such as the alkylating agent Temozolomide (TMZ), in addition to reducing tumor mass, can also sensitize tumors to immune recognition by transient upregulation of multiple stress induced NKG2D ligands (NKG2DL). However, the potential for an effective response by innate lymphocyte effectors such as NK and γδ T cells that recognize NKG2DL is limited by the drug's concomitant lymphodepleting effects. We have previously shown that modification of γδ T cells with a methylguanine DNA methyltransferase (MGMT) transgene confers TMZ resistance via production of O6-alkylguanine DNA alkyltransferase (AGT) thereby enabling γδ T cell function in therapeutic concentrations of TMZ. In this study, we tested this strategy which we have termed Drug Resistant Immunotherapy (DRI) to examine whether combination therapy of TMZ and MGMT-modified γδ T cells could improve survival outcomes in four human/mouse xenograft models of primary and refractory GBM. Our results confirm that DRI leverages the innate response of γδ T cells to chemotherapy-induced stress associated antigen expression and achieves synergies that are significantly greater than either individual approach.


Asunto(s)
Neoplasias Encefálicas/terapia , Glioma/terapia , Inmunoterapia , Receptores de Antígenos de Linfocitos T gamma-delta , Linfocitos T , Temozolomida/farmacología , Transgenes , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Glioma/metabolismo , Glioma/patología , Humanos , Ratones Desnudos , O(6)-Metilguanina-ADN Metiltransferasa/biosíntesis , O(6)-Metilguanina-ADN Metiltransferasa/economía , Linfocitos T/enzimología , Linfocitos T/trasplante , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Biol Blood Marrow Transplant ; 25(2): 233-238, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30219699

RESUMEN

Filgrastim (FIL) is the most common growth factor combined with plerixafor for autologous hematopoietic progenitor cell mobilization, but requires daily, multi-injection administration. We adopted a standardized mobilization regimen with pegfilgrastim (PEG) and upfront plerixafor, allowing for a single injection given the long half-life and slow elimination of PEG. Between 2015 and 2017, a total of 235 patients with lymphoma or plasma cell dyscrasias underwent mobilization with PEG 6 mg on day 1 and upfront plerixafor 24 mg on day 3, followed by apheresis on day 4 regardless of peripheral blood CD34+ cells. The median CD34+ cells/mm3 in peripheral blood on first day of collection was 48 and median collection yield was 7.27 × 106 CD34+ cells/kg (range, 0.32 to 39.6 × 106 CD34+ cells/kg) after a mean of 1.6 apheresis collections. Overall, 83% of patients achieved the mobilization target, and 95% reached the minimum necessary CD34+ cell yield to proceed with transplantation (2 × 106 CD34+ cells/kg). Because FIL is weight-based and dosed daily, the cost comparison with PEG is influenced by patient weight and number of apheresis sessions required. A cost simulation using actual patient data indicates that PEG is associated with lower cost than FIL for the majority of patients. Autologous hematopoietic progenitor cell mobilization with PEG and plerixafor is practical, effective, and not associated with increased cost compared with FIL mobilization.


Asunto(s)
Costos y Análisis de Costo , Filgrastim , Movilización de Célula Madre Hematopoyética/economía , Linfoma , Trasplante de Células Madre de Sangre Periférica/economía , Polietilenglicoles , Adulto , Anciano , Femenino , Filgrastim/administración & dosificación , Filgrastim/economía , Humanos , Linfoma/economía , Linfoma/patología , Linfoma/terapia , Masculino , Persona de Mediana Edad , Polietilenglicoles/administración & dosificación , Polietilenglicoles/economía , Trasplante Autólogo
7.
PLoS One ; 11(12): e0166891, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27907031

RESUMEN

BACKGROUND: Approximately fifty percent of patients with acute myeloid leukemia can be cured with current therapeutic strategies which include, standard dose chemotherapy for patients at standard risk of relapse as assessed by cytogenetic and molecular analysis, or high-dose chemotherapy with allogeneic hematopoietic stem cell transplant for high-risk patients. Despite allogeneic hematopoietic stem cell transplant about 25% of patients still succumb to disease relapse, therefore, novel strategies are needed to improve the outcome of patients with acute myeloid leukemia. METHODS AND FINDINGS: We developed an immunotherapeutic strategy targeting the CD33 myeloid antigen, expressed in ~ 85-90% of patients with acute myeloid leukemia, using chimeric antigen receptor redirected T-cells. Considering that administration of CAR T-cells has been associated with cytokine release syndrome and other potential off-tumor effects in patients, safety measures were here investigated and reported. We genetically modified human activated T-cells from healthy donors or patients with acute myeloid leukemia with retroviral supernatant encoding the inducible Caspase9 suicide gene, a ΔCD19 selectable marker, and a humanized third generation chimeric antigen receptor recognizing human CD33. ΔCD19 selected inducible Caspase9-CAR.CD33 T-cells had a 75±3.8% (average ± standard error of the mean) chimeric antigen receptor expression, were able to specifically lyse CD33+ targets in vitro, including freshly isolated leukemic blasts from patients, produce significant amount of tumor-necrosis-factor-alpha and interferon-gamma, express the CD107a degranulation marker, and proliferate upon antigen specific stimulation. Challenging ΔCD19 selected inducible Caspase9-CAR.CD33 T-cells with programmed-death-ligand-1 enriched leukemia blasts resulted in significant killing like observed for the programmed-death-ligand-1 negative leukemic blasts fraction. Since the administration of 10 nanomolar of a non-therapeutic dimerizer to activate the suicide gene resulted in the elimination of only 76.4±2.0% gene modified cells in vitro, we found that co-administration of the dimerizer with either the BCL-2 inhibitor ABT-199, the pan-BCL inhibitor ABT-737, or mafosfamide, resulted in an additive effect up to complete cell elimination. CONCLUSIONS: This strategy could be investigated for the safety of CAR T-cell applications, and targeting CD33 could be used as a 'bridge" therapy for patients coming to allogeneic hematopoietic stem cell transplant, as anti-leukemia activity from infusing CAR.CD33 T-cells has been demonstrated in an ongoing clinical trial. Albeit never performed in the clinical setting, our future plan is to investigate the utility of iC9-CAR.CD33 T-cells as part of the conditioning therapy for an allogeneic hematopoietic stem cell transplant for acute myeloid leukemia, together with other myelosuppressive agents, whilst the activation of the inducible Caspase9 suicide gene would grant elimination of the infused gene modified T-cells prior to stem cell infusion to reduce the risk of engraftment failure as the CD33 is also expressed on a proportion of the donor stem cell graft.


Asunto(s)
Citotoxicidad Inmunológica , Leucemia Mieloide Aguda/terapia , Receptores de Antígenos de Linfocitos T/inmunología , Proteínas Recombinantes de Fusión/inmunología , Lectina 3 Similar a Ig de Unión al Ácido Siálico/inmunología , Linfocitos T/inmunología , Antígeno B7-H1/farmacología , Compuestos de Bifenilo/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Caspasa 9/genética , Caspasa 9/inmunología , Ingeniería Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Reprogramación Celular , Ensayos Clínicos como Asunto , Ciclofosfamida/análogos & derivados , Ciclofosfamida/farmacología , Vectores Genéticos , Humanos , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/patología , Proteína 1 de la Membrana Asociada a los Lisosomas/genética , Proteína 1 de la Membrana Asociada a los Lisosomas/inmunología , Células Mieloides/efectos de los fármacos , Células Mieloides/inmunología , Células Mieloides/patología , Nitrofenoles/farmacología , Piperazinas/farmacología , Cultivo Primario de Células , Receptores de Antígenos de Linfocitos T/genética , Proteínas Recombinantes de Fusión/genética , Lectina 3 Similar a Ig de Unión al Ácido Siálico/antagonistas & inhibidores , Lectina 3 Similar a Ig de Unión al Ácido Siálico/genética , Sulfonamidas/farmacología , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/inmunología
8.
Medicine (Baltimore) ; 95(39): e4909, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27684826

RESUMEN

INTRODUCTION: CD3+ γδ+ T cells comprise 2% to 5% of circulating T cells with Vγ9Vδ2+ cells the dominant circulating subtype. Vγ9Vδ2+ cells recognize non-peptide phosphoantigens and stress-associated NKG2D ligands expressed on malignant cells. Strategies that incorporate the tumoricidal properties of γδ T cells represent a promising immunotherapeutic strategy for treatment of solid malignancies including neuroblastoma (NB). In this prospective, non-randomized Phase I trial, we assessed whether circulating Vγ9Vδ2+ cells could be safely expanded using intravenous ZOL (Zoledronate [Zometa]) and subcutaneous Interleukin-2 (IL-2) in patients with refractory NB. METHODS: Patients 2 to 21 years of age with refractory neuroblastoma with no known curative therapeutic options received ZOL on day 1, and IL-2 on days 1 to 5 and 15 to 19 of each 28-day cycle (n = 4). Lymphocyte immunophenotyping was assessed weekly. Immunophenotyping studies from the treatment group were compared with healthy pediatric controls (n = 16; range, 5y-15y) and of untreated NB disease controls (n = 9; range, 4m-18y). RESULTS: Treatment was well tolerated with no unexpected grade 3 and 4 toxicities. Lymphocyte subset counts did not differ significantly between volunteers and disease controls with the exception of γδ+ T cell counts that were significantly higher in healthy volunteers (212 + 93 vs. 89 + 42, P = 0.05). Study patients showed increases in circulating γδ+ T cell count (3-10 fold) after the first week, increasing into the range seen in healthy volunteers (125 + 37, P = 0.1940). Interestingly, all ZOL + IL-2 treated patients showed significant increases in CD3+CD4+CD27CD127 T cells that rose weekly in 2 patients throughout the 4 weeks of observation (maximum 41% and 24% of total CD3+CD4+ T cells, respectively). CONCLUSIONS: In summary, combined ZOL and IL-2 is well tolerated and restored γδ+ T cell counts to the normal range with a moderate expansion of Natural Killer cells. Progressive increases in circulating CD4+ T cells with a regulatory phenotype cells may offset beneficial effects of this therapy.


Asunto(s)
Antineoplásicos/administración & dosificación , Inmunoterapia/métodos , Activación de Linfocitos/efectos de los fármacos , Neuroblastoma/terapia , Linfocitos T/efectos de los fármacos , Adolescente , Niño , Preescolar , Difosfonatos/administración & dosificación , Esquema de Medicación , Femenino , Humanos , Imidazoles/administración & dosificación , Inmunofenotipificación , Interleucina-2/administración & dosificación , Recuento de Linfocitos , Masculino , Estudios Prospectivos , Receptores de Antígenos de Linfocitos T gamma-delta/efectos de los fármacos , Adulto Joven , Ácido Zoledrónico
9.
J Immunol Res ; 2016: 2568125, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27610392

RESUMEN

We designed multimodal tumor vaccine that consists of irradiated tumor cells infected with the oncolytic IL-12-expressing HSV-1 virus, M002. This vaccine was tested against the syngeneic neuroblastoma mouse model Neuro 2a injected into the right caudate nucleus of the immunocompetent A/J mice. Mice were vaccinated via intramuscular injection of multimodal vaccine or uninfected irradiated tumor cells at seven and 14 days after tumor establishment. While there was no survival difference between groups vaccinated with cell-based vaccine applied following tumor injection, a premunition prime/boost vaccination strategy produced a significant survival advantage in both groups and sustained immune response to an intracranial rechallenge of the same tumor. The syngeneic but unrelated H6 hepatocellular tumor cell line grew unrestricted in vaccinated mice, indicative of vaccine-mediated specific immunity to Neuro 2a tumors. Longitudinal analyses of tumor-infiltrating lymphocytes revealed a primary adaptive T cell response involving both CD4+ and CD8+ T cell subsets. Spleen cell mononuclear preparations from vaccinated mice were significantly more cytotoxic to Neuro 2a tumor cells than spleen cells from control mice as demonstrated in a four-hour in vitro cytotoxicity assay. These results strongly suggest that an irradiated whole cell tumor vaccine incorporating IL-12-expressing M002 HSV can produce a durable, specific immunization in a murine model of intracranial tumor.


Asunto(s)
Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Herpesvirus Humano 1/genética , Interleucina-12/genética , Neuroblastoma/genética , Neuroblastoma/inmunología , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/terapia , Vacunas contra el Cáncer/administración & dosificación , Línea Celular Tumoral , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Vectores Genéticos/genética , Inmunidad Celular/inmunología , Inmunofenotipificación , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Linfocitos Infiltrantes de Tumor/patología , Ratones , Clasificación del Tumor , Neuroblastoma/patología , Neuroblastoma/terapia , Virus Oncolíticos/genética , Fenotipo , Análisis de Supervivencia , Resultado del Tratamiento , Vacunación
10.
Gynecol Oncol ; 140(2): 245-52, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26616223

RESUMEN

OBJECTIVE: The purpose of this study was to evaluate the safety, efficacy, and immunogenicity of a plasmid vaccine, pNGVL4a-CRT-E7(detox), administered either intradermally, intramuscularly, or directly into the cervical lesion, in patients with HPV16-associated CIN2/3. METHODS: Eligible patients with HPV16(+) CIN2/3 were enrolled in treatment cohorts evaluating pNGVL4a-CRT-E7(detox), administered by either particle-mediated epidermal delivery (PMED), intramuscular injection (IM), or cervical intralesional injection, at study weeks 0, 4, and 8. Patients were monitored for local injection site and systemic toxicity. A standard therapeutic resection was performed at week 15. The primary endpoints were safety and tolerability. Secondary endpoints included histologic regression and change in cervical HPV viral load. Exploratory endpoints included immune responses in the blood and in the target tissue. RESULTS: Thirty-two patients with HPV16(+) CIN2/3 were enrolled onto the treatment phase of the study, and were vaccinated. Twenty-two of 32 patients (69%) experienced vaccine-specific related adverse events. The most frequent vaccine-related events were constitutional and local injection site in nature, and were grade 1 or less in severity. Histologic regression to CIN 1 or less occurred in 8 of 27 (30%) patients who received all vaccinations and underwent LEEP. In subject-matched comparisons, intraepithelial CD8+ T cell infiltrates increased after vaccination in subjects in the intralesional administration cohort. CONCLUSION: pNGVL4a-CRT-E7(detox) was well-tolerated, elicited the most robust immune response when administered intralesionally, and demonstrated preliminary evidence of potential clinical efficacy.


Asunto(s)
Papillomavirus Humano 16/aislamiento & purificación , Infecciones por Papillomavirus/terapia , Vacunas contra Papillomavirus/administración & dosificación , Displasia del Cuello del Útero/terapia , Neoplasias del Cuello Uterino/terapia , Vacunas de ADN/administración & dosificación , Adulto , Linfocitos T CD8-positivos/inmunología , Relación Dosis-Respuesta Inmunológica , Femenino , Humanos , Infecciones por Papillomavirus/inmunología , Infecciones por Papillomavirus/patología , Infecciones por Papillomavirus/virología , Vacunas contra Papillomavirus/efectos adversos , Proyectos Piloto , Neoplasias del Cuello Uterino/inmunología , Neoplasias del Cuello Uterino/virología , Vacunas de ADN/efectos adversos , Carga Viral , Adulto Joven , Displasia del Cuello del Útero/inmunología , Displasia del Cuello del Útero/virología
11.
Cell Rep ; 12(10): 1668-77, 2015 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-26321643

RESUMEN

Mutations of the Janus family kinase JAK3 gene cause severe combined immunodeficiency (SCID). JAK3 deficiency in humans is characterized by the absence of circulating T cells and natural killer (NK) cells with normal numbers of poorly functioning B cells (T(-)B(+)NK(-)). Using SCID patient-specific induced pluripotent stem cells (iPSCs) and a T cell in vitro differentiation system, we demonstrate a complete block in early T cell development of JAK3-deficient cells. Correction of the JAK3 mutation by CRISPR/Cas9-enhanced gene targeting restores normal T cell development, including the production of mature T cell populations with a broad T cell receptor (TCR) repertoire. Whole-genome sequencing of corrected cells demonstrates no CRISPR/Cas9 off-target modifications. These studies describe an approach for the study of human lymphopoiesis and provide a foundation for gene correction therapy in humans with immunodeficiencies.


Asunto(s)
Terapia Genética , Janus Quinasa 3/genética , Inmunodeficiencia Combinada Grave/terapia , Proteínas Bacterianas/genética , Secuencia de Bases , Proteína 9 Asociada a CRISPR , Células Cultivadas , Preescolar , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Análisis Mutacional de ADN , Endonucleasas/genética , Marcación de Gen , Humanos , Células Madre Pluripotentes Inducidas/enzimología , Masculino , Mutación Missense , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Inmunodeficiencia Combinada Grave/genética , Linfocitos T/fisiología
12.
PLoS One ; 10(5): e0122387, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25955158

RESUMEN

Human γδ T cells are potent effectors against glioma cell lines in vitro and in human/mouse xenograft models of glioblastoma, however, this effect has not been investigated in an immunocompetent mouse model. In this report, we established GL261 intracranial gliomas in syngeneic WT C57BL/6 mice and measured circulating γδ T cell count, phenotype, Vγ/Vδ repertoire, tumor histopathology, NKG2D ligands expression, and T cell invasion at day 10-12 post-injection and at end stage. Circulating γδ T cells transiently increased and upregulated Annexin V expression at post-tumor day 10-12 followed by a dramatic decline in γδ T cell count at end stage. T cell receptor repertoire showed no changes in Vγ1, Vγ4, Vγ7 or Vδ1 subsets from controls at post-tumor day 10-12 or at end stage except for an end-stage increase in the Vδ4 population. Approximately 12% of γδ T cells produced IFN-γ. IL-17 and IL-4 producing γδ T cells were not detected. Tumor progression was the same in TCRδ-/- C57BL/6 mice as that observed in WT mice, suggesting that γδ T cells exerted neither a regulatory nor a sustainable cytotoxic effect on the tumor. WT mice that received an intracranial injection of γδ T cells 15m following tumor placement showed evidence of local tumor growth inhibition but this was insufficient to confer a survival advantage over untreated controls. Taken together, our findings suggest that an early nonspecific proliferation of γδ T cells followed by their depletion occurs in mice implanted with syngeneic GL261 gliomas. The mechanism by which γδ T cell expansion occurs remains a subject for further investigation of the mechanisms responsible for this immune response in the setting of high-grade glioma.


Asunto(s)
Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Encéfalo/metabolismo , Glioma/inmunología , Glioma/patología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Encéfalo/inmunología , Neoplasias Encefálicas/sangre , Línea Celular Tumoral , Modelos Animales de Enfermedad , Glioma/sangre , Humanos , Interleucina-17/inmunología , Interleucina-4/inmunología , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos T gamma-delta/análisis , Receptores de Antígenos de Linfocitos T gamma-delta/sangre , Subgrupos de Linfocitos T/patología
13.
Cancer Immunol Immunother ; 64(5): 551-62, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25676710

RESUMEN

The standard treatment of high-grade glioma presents a combination of radiotherapy, chemotherapy and surgery. Immunotherapy is proposed as a potential adjunct to standard cytotoxic regimens to target remaining microscopic disease following resection. We have shown ex vivo expanded/activated γδ T cells to be a promising innate lymphocyte therapy based on their recognition of stress antigens expressed on gliomas. However, successful integration of γδ T cell therapy protocols requires understanding the efficacy and safety of adoptively transferred immune cells in the post-treatment environment. The unique features of γδ T cell product and the environment (hypoxia, inflammation) can affect levels of expression of key cell receptors and secreted factors and either promote or hinder the feasibility of γδ T cell therapy. We investigated the potential for the γδ T cells to injure normal brain tissue that may have been stressed by treatment. We evaluated γδ T cell toxicity by assessing actual and correlative toxicity indicators in several available models including: (1) expression of stress markers on normal primary human astrocytes (as surrogate for brain parenchyma) after irradiation and temozolomide treatment, (2) cytotoxicity of γδ T cells on normal and irradiated primary astrocytes, (3) microglial activation and expression of stress-induced ligands in mouse brain after whole-brain irradiation and (4) expression of stress-induced markers on human brain tumors and on normal brain tissue. The lack of expression of stress-induced ligands in all tested models suggests that γδ T cell therapy is safe for brain tumor patients who undergo standard cytotoxic therapies.


Asunto(s)
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Inmunoterapia Adoptiva/métodos , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Linfocitos T/trasplante , Animales , Astrocitos/inmunología , Astrocitos/metabolismo , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/radioterapia , Línea Celular Tumoral , Tratamiento Basado en Trasplante de Células y Tejidos/efectos adversos , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Citotoxicidad Inmunológica/inmunología , Glioblastoma/inmunología , Glioblastoma/radioterapia , Humanos , Inmunoterapia Adoptiva/efectos adversos , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Microglía/inmunología , Microglía/metabolismo , Subfamilia K de Receptores Similares a Lectina de Células NK/biosíntesis , Linfocitos T/inmunología
14.
Front Pharmacol ; 5: 254, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25505885

RESUMEN

Adoptive T-cell therapy can involve donor lymphocyte infusion after allogeneic hematopoietic stem cell transplantation, the administration of tumor infiltrating lymphocyte expanded ex-vivo, or more recently the use of T cell receptor or chimeric antigen receptor redirected T cells. However, cellular therapies can pose significant risks, including graft-vs.-host-disease and other on and off-target effects, and therefore strategies need to be implemented to permanently reverse any sign of toxicity. A suicide gene is a genetically encoded molecule that allows selective destruction of adoptively transferred cells. Suicide gene addition to cellular therapeutic products can lead to selective ablation of gene-modified cells, preventing collateral damage to contiguous cells and/or tissues. The "ideal" suicide gene would ensure the safety of gene modified cellular applications by granting irreversible elimination of "all" and "only" the cells responsible for the unwanted toxicity. This review presents the suicide gene safety systems reported to date, with a focus on the state-of-the-art and potential applications regarding two of the most extensively validated suicide genes, including the clinical setting: herpes-simplex-thymidine-kinase and inducible-caspase-9.

15.
Mol Ther ; 22(8): 1416-1422, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24895997

RESUMEN

Exploration of cancer immunotherapy strategies that incorporate γδ T cells as primary mediators of antitumor immunity are just beginning to be explored and with a primary focus on the use of manufactured phosphoantigen-stimulated Vγ9Vδ2 T cells. Increasing evidence, however, supports a critical role for Vδ1+ γδ T cells, a minor subset in peripheral blood with distinct innate recognition properties that possess powerful tumoricidal activity. They are activated by a host of ligands including stress-induced self-antigens, glycolipids presented by CD1c/d, and potentially many others that currently remain unidentified. In contrast to Vγ9Vδ2 T cells, tumor-reactive Vδ1+ T cells are not as susceptible to activation-induced cell death and can persist in the circulation for many years, potentially offering durable immunity to some cancers. In addition, specific populations of Vδ1+ T cells can also exhibit immunosuppressive and regulatory properties, a function that can also be exploited for therapeutic purposes. This review explores the biology, function, manufacturing strategies, and potential therapeutic role of Vδ1+ T cells. We also discuss clinical experience with Vδ1+ T cells in the setting of cancer, as well as the potential of and barriers to the development of Vδ1+ T cell-based adoptive cell therapy strategies.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Linfocitos T/citología , Humanos , Neoplasias/terapia , Receptores de Antígenos de Linfocitos T gamma-delta/sangre
16.
PLoS One ; 9(5): e97335, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24828440

RESUMEN

Human induced pluripotent stem cells (hiPSCs) have enormous potential for the treatment of inherited and acquired disorders. Recently, antigen-specific T lymphocytes derived from hiPSCs have been reported. However, T lymphocyte populations with broad T cell receptor (TCR) diversity have not been generated. We report that hiPSCs derived from skin biopsy are capable of producing T lymphocyte populations with a broad TCR repertoire. In vitro T cell differentiation follows a similar developmental program as observed in vivo, indicated by sequential expression of CD7, intracellular CD3 and surface CD3. The γδ TCR locus is rearranged first and is followed by rearrangement of the αß locus. Both γδ and αß T cells display a diverse TCR repertoire. Upon activation, the cells express CD25, CD69, cytokines (TNF-α, IFN-γ, IL-2) and cytolytic proteins (Perforin and Granzyme-B). These results suggest that most, if not all, mechanisms required to generate functional T cells with a broad TCR repertoire are intact in our in vitro differentiation protocol. These data provide a foundation for production of patient-specific T cells for the treatment of acquired or inherited immune disorders and for cancer immunotherapy.


Asunto(s)
Diferenciación Celular/inmunología , Células Madre Pluripotentes Inducidas/inmunología , Activación de Linfocitos/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Linfocitos T/inmunología , Línea Celular , Granzimas/inmunología , Humanos , Interferón gamma/inmunología , Interleucina-2/inmunología , Perforina/inmunología , Factor de Necrosis Tumoral alfa/inmunología
17.
Biol Blood Marrow Transplant ; 20(4): 581-6, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24370862

RESUMEN

Allogeneic hematopoietic stem cell transplantation for patients with a hemoglobinopathy can be curative but is limited by donor availability. Although positive results are frequently observed in those with an HLA-matched sibling donor, use of unrelated donors has been complicated by poor engraftment, excessive regimen-related toxicity, and graft-versus-host disease (GVHD). As a potential strategy to address these obstacles, a pilot study was designed that incorporated both a reduced-intensity conditioning and mesenchymal stromal cells (MSCs). Six patients were enrolled, including 4 with high-risk sickle cell disease (SCD) and 2 with transfusion-dependent thalassemia major. Conditioning consisted of fludarabine (150 mg/m(2)), melphalan (140 mg/m(2)), and alemtuzumab (60 mg for patients weighing > 30 kg and .9 mg/kg for patients weighing <30 kg). Two patients received HLA 7/8 allele matched bone marrow and 4 received 4-5/6 HLA matched umbilical cord blood as the source of HSCs. MSCs were of bone marrow origin and derived from a parent in 1 patient and from an unrelated third-party donor in the remaining 5 patients. GVHD prophylaxis consisted of cyclosporine A and mycophenolate mofetil. One patient had neutropenic graft failure, 2 had autologous hematopoietic recovery, and 3 had hematopoietic recovery with complete chimerism. The 2 SCD patients with autologous hematopoietic recovery are alive. The remaining 4 died either from opportunistic infection, GVHD, or intracranial hemorrhage. Although no infusion-related toxicity was seen, the cotransplantation of MSCs was not sufficient for reliable engraftment in patients with advanced hemoglobinopathy. Although poor engraftment has been observed in nearly all such trials to date in this patient population, there was no evidence to suggest that MSCs had any positive impact on engraftment. Because of the lack of improved engraftment and unacceptably high transplant-related mortality, the study was prematurely terminated. Further investigations into understanding the mechanisms of graft resistance and development of strategies to overcome this barrier are needed to move this field forward.


Asunto(s)
Anemia de Células Falciformes/terapia , Trasplante de Células Madre de Sangre del Cordón Umbilical , Trasplante de Células Madre Mesenquimatosas , Agonistas Mieloablativos/uso terapéutico , Acondicionamiento Pretrasplante/métodos , Talasemia beta/terapia , Adolescente , Alemtuzumab , Anemia de Células Falciformes/inmunología , Anemia de Células Falciformes/mortalidad , Anemia de Células Falciformes/patología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Niño , Femenino , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/mortalidad , Enfermedad Injerto contra Huésped/patología , Enfermedad Injerto contra Huésped/prevención & control , Antígenos HLA/inmunología , Prueba de Histocompatibilidad , Humanos , Masculino , Melfalán/uso terapéutico , Análisis de Supervivencia , Trasplante Homólogo , Insuficiencia del Tratamiento , Donante no Emparentado , Vidarabina/análogos & derivados , Vidarabina/uso terapéutico , Talasemia beta/inmunología , Talasemia beta/mortalidad , Talasemia beta/patología
18.
PLoS One ; 8(8): e68729, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23950874

RESUMEN

Vδ2(neg) γδ T cells, of which Vδ1+ γδ T cells are by far the largest subset, are important effectors against CMV infection. Malignant gliomas often contain CMV genetic material and proteins, and evidence exists that CMV infection may be associated with initiation and/or progression of glioblastoma multiforme (GBM). We sought to determine if Vδ1+ γδ T cells were cytotoxic to GBM and the extent to which their cytotoxicity was CMV dependent. We examined the cytotoxic effect of ex vivo expanded/activated Vδ1+ γδ T cells from healthy CMV seropositive and CMV seronegative donors on unmanipulated and CMV-infected established GBM cell lines and cell lines developed from short- term culture of primary tumors. Expanded/activated Vδ1+ T cells killed CMV-negative U251, U87, and U373 GBM cell lines and two primary tumor explants regardless of the serologic status of the donor. Experimental CMV infection did not increase Vδ1+ T cell--mediated cytotoxicity and in some cases the cell lines were more resistant to lysis when infected with CMV. Flow cytometry analysis of CMV-infected cell lines revealed down-regulation of the NKG2D ligands ULBP-2, and ULBP-3 as well as MICA/B in CMV-infected cells. These studies show that ex vivo expanded/activated Vδ1+ γδ T cells readily recognize and kill established GBM cell lines and primary tumor-derived GBM cells regardless of whether CMV infection is present, however, CMV may enhance the resistance GBM cell lines to innate recognition possibly contributing to the poor immunogenicity of GBM.


Asunto(s)
Citotoxicidad Inmunológica/inmunología , Glioblastoma/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Linfocitos T/inmunología , Línea Celular , Línea Celular Tumoral , Citomegalovirus/clasificación , Citomegalovirus/genética , Citomegalovirus/inmunología , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/metabolismo , Infecciones por Citomegalovirus/virología , Citometría de Flujo , Proteínas Ligadas a GPI/inmunología , Proteínas Ligadas a GPI/metabolismo , Glioblastoma/patología , Glioblastoma/virología , Humanos , Hibridación in Situ , Péptidos y Proteínas de Señalización Intercelular/inmunología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Activación de Linfocitos/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Linfocitos T/metabolismo , Linfocitos T/virología , Células Tumorales Cultivadas
20.
PLoS One ; 8(1): e51805, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23326319

RESUMEN

Classical approaches to immunotherapy that show promise in some malignancies have generally been disappointing when applied to high-grade brain tumors such as glioblastoma multiforme (GBM). We recently showed that ex vivo expanded/activated γδ T cells recognize NKG2D ligands expressed on malignant glioma and are cytotoxic to glioma cell lines and primary GBM explants. In addition, γδ T cells extend survival and slow tumor progression when administered to immunodeficient mice with intracranial human glioma xenografts. We now show that temozolomide (TMZ), a principal chemotherapeutic agent used to treat GBM, increases the expression of stress-associated NKG2D ligands on TMZ-resistant glioma cells, potentially rendering them vulnerable to γδ T cell recognition and lysis. TMZ is also highly toxic to γδ T cells, however, and to overcome this cytotoxic effect γδ T cells were genetically modified using a lentiviral vector encoding the DNA repair enzyme O(6)-alkylguanine DNA alkyltransferase (AGT) from the O(6)-methylguanine methyltransferase (MGMT) cDNA, which confers resistance to TMZ. Genetic modification of γδ T cells did not alter their phenotype or their cytotoxicity against GBM target cells. Importantly, gene modified γδ T cells showed greater cytotoxicity to two TMZ resistant GBM cell lines, U373(TMZ-R) and SNB-19(TMZ-R) cells, in the presence of TMZ than unmodified cells, suggesting that TMZ exposed more receptors for γδ T cell-targeted lysis. Therefore, TMZ resistant γδ T cells can be generated without impairing their anti-tumor functions in the presence of high concentrations of TMZ. These results provide a mechanistic basis for combining chemotherapy and γδ T cell-based drug resistant cellular immunotherapy to treat GBM.


Asunto(s)
Dacarbazina/análogos & derivados , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Linfocitos T/inmunología , Antineoplásicos Alquilantes/farmacología , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/terapia , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Células Cultivadas , Citotoxicidad Inmunológica/efectos de los fármacos , Citotoxicidad Inmunológica/genética , Citotoxicidad Inmunológica/inmunología , Dacarbazina/farmacología , Difosfonatos/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Resistencia a Antineoplásicos/inmunología , Citometría de Flujo , Proteínas Ligadas a GPI/inmunología , Proteínas Ligadas a GPI/metabolismo , Ingeniería Genética , Vectores Genéticos/genética , Glioblastoma/inmunología , Glioblastoma/patología , Glioblastoma/terapia , Humanos , Imidazoles/farmacología , Péptidos y Proteínas de Señalización Intercelular/inmunología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Interleucina-2/farmacología , Lentivirus/genética , O(6)-Metilguanina-ADN Metiltransferasa/genética , O(6)-Metilguanina-ADN Metiltransferasa/inmunología , O(6)-Metilguanina-ADN Metiltransferasa/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Linfocitos T/metabolismo , Temozolomida , Ácido Zoledrónico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...