Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Immunother ; 44(2): 63-70, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33443972

RESUMEN

Adoptive cell transfer (ACT) with tumor-infiltrating lymphocytes (TILs) can generate durable clinical responses in patients with metastatic melanoma and ongoing trials are evaluating efficacy in other advanced solid tumors. The aim of this study was to develop methods for the expansion of tumor-reactive TIL from resected soft tissue sarcoma to a degree required for the ACT. From 2015 to 2018, 70 patients were consented to an institutional review board-approved protocol, and fresh surgical specimens were taken directly from the operating room to the laboratory. Fragments of the tumor (1 mm3) or fresh tumor digest were placed in culture for a period of 4 weeks. Successfully propagated TIL from these cultures were collected and analyzed by flow cytometry. TIL were cocultured with autologous tumor and function was assessed by measurement of interferon-γ in the supernatant by enzyme-linked immunosorbent assay. Initial TIL cultures were further expanded using a rapid expansion protocol. Nearly all specimens generated an initial TIL culture (91% fragment method, 100% digest method). The phenotype of the TIL indicated a predominant CD3+ population after culture (43% fragment, 52% digest) and TIL were responsive to the autologous tumor (56% fragment, 40% digest). The cultured TIL expanded to a degree required for clinical use following rapid expansion protocol (median: 490-fold fragment, 403-fold digest). The data demonstrate the feasibility of TIL culture from fresh soft tissue sarcoma. The derived TIL have tumor-specific reactivity and can be expanded to clinically relevant numbers. An active ACT clinical trial using the methods described in this report is now approved for patients with metastatic soft tissue sarcoma.


Asunto(s)
Linfocitos Infiltrantes de Tumor/inmunología , Sarcoma/inmunología , Sarcoma/patología , Microambiente Tumoral , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores , Terapia Combinada/efectos adversos , Terapia Combinada/métodos , Citotoxicidad Inmunológica , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Femenino , Humanos , Inmunofenotipificación , Inmunoterapia/efectos adversos , Inmunoterapia/métodos , Linfocitos Infiltrantes de Tumor/metabolismo , Linfocitos Infiltrantes de Tumor/patología , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Estadificación de Neoplasias , Sarcoma/terapia , Adulto Joven
2.
Cancer Immunol Immunother ; 69(12): 2465-2476, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32556443

RESUMEN

Emm55 is a bacterial gene derived from Streptococcus pyogenes (S. pyogenes) that was cloned into a plasmid DNA vaccine (pAc/emm55). In this study, we investigated the anti-tumor efficacy of pAc/emm55 in a B16 murine melanoma model. Intralesional (IL) injections of pAc/emm55 significantly delayed tumor growth compared to the pAc/Empty group. There was a significant increase in the CD8+ T cells infiltrating into the tumors after pAc/emm55 treatment compared to the control group. In addition, we observed that IL injection of pAc/emm55 increased antigen-specific T cell infiltration into tumors. Depletion of CD4+ or CD8+ T cells abrogated the anti-tumor effect of pAc/emm55. Combination treatment of IL injection of pAc/emm55 with anti-PD-1 antibody significantly delayed tumor growth compared to either monotherapy. pAc/emm55 treatment combined with PD-1 blockade enhanced anti-tumor immune response and improved systemic anti-tumor immunity. Together, these strategies may lead to improvements in the treatment of patients with melanoma.


Asunto(s)
Antígenos Bacterianos/inmunología , Antineoplásicos Inmunológicos/administración & dosificación , Proteínas de la Membrana Bacteriana Externa/inmunología , Inmunoterapia/métodos , Melanoma Experimental/terapia , Animales , Antígenos Bacterianos/genética , Proteínas de la Membrana Bacteriana Externa/genética , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral/trasplante , Terapia Combinada/métodos , Femenino , Humanos , Inyecciones Intralesiones , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma Experimental/inmunología , Ratones , Plásmidos/administración & dosificación , Plásmidos/genética , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología
3.
Orthop J Sports Med ; 7(3): 2325967119834077, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30937320

RESUMEN

BACKGROUND: Many surgical procedures are intended to return patients to sport early, but it is unknown how realistic these expectations are after shoulder surgery. PURPOSE: To determine which of the commonly performed surgical interventions in the shoulder best facilitated return to sport, and which did not, by 6 months postoperatively. STUDY DESIGN: Cohort study; Level of evidence, 3. METHODS: The study was a retrospective analysis of prospectively collected data from patients who underwent shoulder surgery from a single surgeon over 12 years. To be included, at least 20 patients needed to have undergone that procedure and complete a questionnaire evaluating their shoulder's function preoperatively and 6 months postoperatively. The primary outcome was a change in the response to the question, "What is your current level of sport?" RESULTS: A total of 2261 surgical procedures in 13 categories met the inclusion criteria. Capsular release was the only procedure associated with improved patient-reported sporting level at 6 months (d = 0.18 [95% CI, 0.05-0.30]; P = .009). This represented a mean improvement of 41% from the preoperative sporting level. Bankart repair was associated with the greatest decrease in patient-reported sporting level at 6 months (mean decline of 21%) (d = -0.17 [95% CI, -0.34 to -0.01]; P = .034), followed by rotator cuff repair (mean decline of 13%) (d = -0.06 [95% CI, -0.03 to -0.10]; P = .0004). There were no significant changes in sporting level at 6 months postoperatively for rotator cuff repair with acromioplasty, polytetrafluoroethylene (PTFE) patch repair, acromioplasty, superior labral anterior to posterior (SLAP) repair, total shoulder arthroplasty, reverse total shoulder arthroplasty, rotator cuff repair with capsular release, rotator cuff repair with stabilization, calcific debridement, or hemiarthroplasty. CONCLUSION: Capsular release was the only surgical procedure that provided a significant improvement in patient-reported sporting level in a relatively short period of time (6 months). Patients who underwent rotator cuff repair and Bankart repair were the only surgical groups that reported a significant decline in sporting level 6 months postoperatively.

4.
PLoS One ; 13(4): e0196033, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29694419

RESUMEN

Intralesional (IL) injection of Rose Bengal (PV-10) induces regression of injected and uninjected lesions in several murine tumor models. In this study, we investigated the anti-tumor response of combining IL PV-10 with blockade of the PD-1 / PD-L1 pathway and the role of immune cell populations in eliciting this response. To investigate the role of T cell subsets in mediating an immune response, B16 or M05 melanoma-bearing mice received combination therapy as well as CD8+, CD4+, or CD25+ depleting antibodies. Tumor growth was measured. T cells were collected from spleens or tumors, and phenotype, activation markers, and reactivity were measured. Splenocytes from mice treated with combination therapy had increased OVA antigen-specific CD8+ T cells in M05-tumor-bearing mice. Depletion of CD4+ T cells or regulatory T cells (Tregs) in combination with IL PV-10 and anti-PD-1 antibody treatment resulted in an enhanced anti-tumor effect. Treatment with CD8+ depleting antibody abrogated anti-tumor immunity. These results support a clinical study for the safety and anti-tumor immune responses with combination therapy of IL PV-10 and PD-1/PD-L1 blockade.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Melanoma Experimental/tratamiento farmacológico , Rosa Bengala/administración & dosificación , Neoplasias Cutáneas/tratamiento farmacológico , Linfocitos T/inmunología , Animales , Anticuerpos Monoclonales/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Antígeno B7-H1/antagonistas & inhibidores , Antígenos CD4/metabolismo , Antígenos CD8/metabolismo , Línea Celular Tumoral , Citotoxicidad Inmunológica , Humanos , Inyecciones Intralesiones , Inyecciones Intraperitoneales , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Melanoma Experimental/inmunología , Ratones , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Rosa Bengala/farmacología , Neoplasias Cutáneas/inmunología , Linfocitos T/efectos de los fármacos , Resultado del Tratamiento
5.
Oncotarget ; 7(25): 37893-37905, 2016 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-27177220

RESUMEN

Intralesional (IL) therapy is under investigation to treat dermal and subcutaneous metastatic cancer. Rose bengal (RB) is a staining agent that was originally used by ophthalmologists and in liver function studies. IL injection of RB has been shown to induce regression of injected and uninjected tumors in murine models and clinical trials. In this study, we have shown a mechanism of tumor-specific immune response induced by IL RB. In melanoma-bearing mice, IL RB induced regression of injected tumor and inhibited the growth of bystander lesions mediated by CD8+ T cells. IL RB resulted in necrosis of tumor cells and the release of High Mobility Group Box 1 (HMGB1), with increased dendritic cell (DC) infiltration into draining lymph nodes and the activation of tumor-specific T cells. Treatment of DC with tumor supernatants increased the ability of DCs to stimulate T cell proliferation, and blockade of HMGB1 in the supernatants suppressed DC activity. Additionally, increased HMGB1 levels were measured in the sera of melanoma patients treated with IL RB. These results support the role of IL RB to activate dendritic cells at the site of tumor necrosis for the induction of a systemic anti-tumor immune response.


Asunto(s)
Células Dendríticas/efectos de los fármacos , Proteína HMGA1a/metabolismo , Melanoma/tratamiento farmacológico , Rosa Bengala/farmacología , Adulto , Anciano , Animales , Antineoplásicos/farmacología , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Proliferación Celular , Células Dendríticas/citología , Células Dendríticas/metabolismo , Femenino , Células HEK293 , Humanos , Leucocitos Mononucleares/citología , Masculino , Melanoma Experimental/metabolismo , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Células 3T3 NIH , Necrosis , Proyectos Piloto , Linfocitos T/citología , Adulto Joven
6.
PLoS One ; 11(4): e0153053, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27050669

RESUMEN

Tumor-infiltrating lymphocytes (TIL) has been associated with improved survival in cancer patients. Within the tumor microenvironment, regulatory cells and expression of co-inhibitory immune checkpoint molecules can lead to the inactivation of TIL. Hence, there is a need to develop strategies that disrupt these negative regulators to achieve robust anti-tumor immune responses. We evaluated the blockade of immune checkpoints and their effect on T cell infiltration and function. We examined the ability of TIL to induce tumor-specific immune responses in vitro and in vivo. TIL isolated from tumor bearing mice were tumor-specific and expressed co-inhibitory immune checkpoint molecules. Administration of monoclonal antibodies against immune checkpoints led to a significant delay in tumor growth. However, anti-PD-L1 antibody treated mice had a significant increase in T cell infiltration and IFN-γ production compared to other groups. Adoptive transfer of in vitro expanded TIL from tumors of anti-PD-L1 antibody treated mice led to a significant delay in tumor growth. Blockade of co-inhibitory immune checkpoints could be an effective strategy to improve TIL infiltration and function.


Asunto(s)
Traslado Adoptivo , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias Experimentales/terapia , Animales , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/inmunología , Femenino , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales/inmunología
7.
Cancer Res ; 76(6): 1381-90, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26719539

RESUMEN

Cancer immunotherapies, such as immune checkpoint blockade or adoptive T-cell transfer, can lead to durable responses in the clinic, but response rates remain low due to undefined suppression mechanisms. Solid tumors are characterized by a highly acidic microenvironment that might blunt the effectiveness of antitumor immunity. In this study, we directly investigated the effects of tumor acidity on the efficacy of immunotherapy. An acidic pH environment blocked T-cell activation and limited glycolysis in vitro. IFNγ release blocked by acidic pH did not occur at the level of steady-state mRNA, implying that the effect of acidity was posttranslational. Acidification did not affect cytoplasmic pH, suggesting that signals transduced by external acidity were likely mediated by specific acid-sensing receptors, four of which are expressed by T cells. Notably, neutralizing tumor acidity with bicarbonate monotherapy impaired the growth of some cancer types in mice where it was associated with increased T-cell infiltration. Furthermore, combining bicarbonate therapy with anti-CTLA-4, anti-PD1, or adoptive T-cell transfer improved antitumor responses in multiple models, including cures in some subjects. Overall, our findings show how raising intratumoral pH through oral buffers therapy can improve responses to immunotherapy, with the potential for immediate clinical translation.


Asunto(s)
Antineoplásicos/inmunología , Microambiente Tumoral/inmunología , Animales , Anticuerpos/inmunología , Bicarbonatos/farmacología , Antígeno CTLA-4/inmunología , Línea Celular Tumoral , Femenino , Concentración de Iones de Hidrógeno , Inmunoterapia/métodos , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Receptor de Muerte Celular Programada 1/inmunología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Microambiente Tumoral/efectos de los fármacos
8.
PLoS One ; 8(7): e68561, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23874673

RESUMEN

Intralesional (IL) injection of PV-10 has shown to induce regression of both injected and non-injected lesions in patients with melanoma. To determine an underlying immune mechanism, the murine B16 melanoma model and the MT-901 breast cancer model were utilized. In BALB/c mice bearing MT-901 breast cancer, injection of PV-10 led to regression of injected and untreated contralateral subcutaneous lesions. In a murine model of melanoma, B16 cells were injected into C57BL/6 mice to establish one subcutaneous tumor and multiple lung lesions. Treatment of the subcutaneous lesion with a single injection of IL PV-10 led to regression of the injected lesion as well as the distant B16 melanoma lung metastases. Anti-tumor immune responses were measured in splenocytes collected from mice treated with IL PBS or PV-10. Splenocytes isolated from tumor bearing mice treated with IL PV-10 demonstrated enhanced tumor-specific IFN-gamma production compared to splenocytes from PBS-treated mice in both models. In addition, a significant increase in lysis of B16 cells by T cells isolated after PV-10 treatment was observed. Transfer of T cells isolated from tumor-bearing mice treated with IL PV-10 led to tumor regression in mice bearing B16 melanoma. These studies establish that IL PV-10 therapy induces tumor-specific T cell-mediated immunity in multiple histologic subtypes and support the concept of combining IL PV10 with immunotherapy for advanced malignancies.


Asunto(s)
Neoplasias de la Mama/inmunología , Modelos Animales de Enfermedad , Melanoma/inmunología , Rosa Bengala/administración & dosificación , Traslado Adoptivo , Animales , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática , Femenino , Inyecciones Intralesiones , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
9.
Cancer Immunol Immunother ; 62(6): 1083-91, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23604104

RESUMEN

Pancreatic cancer is an extremely aggressive malignancy with a dismal prognosis. Cancer patients and tumor-bearing mice have multiple immunoregulatory subsets including regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSC) that may limit the effectiveness of anti-tumor immunotherapies for pancreatic cancer. It is possible that modulating these subsets will enhance anti-tumor immunity. The goal of this study was to explore depletion of immunoregulatory cells to enhance dendritic cell (DC)-based cancer immunotherapy in a murine model of pancreatic cancer. Flow cytometry results showed an increase in both Tregs and MDSC in untreated pancreatic cancer-bearing mice compared with control. Elimination of Tregs alone or in combination with DC-based vaccination had no effect on pancreatic tumor growth or survival. Gemcitabine (Gem) is a chemotherapeutic drug routinely used for the treatment for pancreatic cancer patients. Treatment with Gem led to a significant decrease in MDSC percentages in the spleens of tumor-bearing mice, but did not enhance overall survival. However, combination therapy with DC vaccination followed by Gem treatment led to a significant delay in tumor growth and improved survival in pancreatic cancer-bearing mice. Increased MDSC were measured in the peripheral blood of patients with pancreatic cancer. Treatment with Gem also led to a decrease of this population in pancreatic cancer patients, suggesting that combination therapy with DC-based cancer vaccination and Gem may lead to improved treatments for patients with pancreatic cancer.


Asunto(s)
Antimetabolitos Antineoplásicos/administración & dosificación , Carcinoma/mortalidad , Carcinoma/terapia , Células Dendríticas , Desoxicitidina/análogos & derivados , Inmunoterapia Adoptiva , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/terapia , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Carcinoma/patología , Línea Celular Tumoral , Terapia Combinada , Células Dendríticas/inmunología , Desoxicitidina/administración & dosificación , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Neoplasias Pancreáticas/patología , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Carga Tumoral/efectos de los fármacos , Carga Tumoral/inmunología , Gemcitabina
10.
J Immunol ; 189(11): 5147-54, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23100512

RESUMEN

Administration of nonmyeloablative chemotherapeutic agents or total body irradiation (TBI) prior to adoptive transfer of tumor-specific T cells may reduce or eliminate immunosuppressive populations such as T regulatory cells (Tregs) and myeloid-derived suppressor cells (MDSC). Little is known about these populations during immune reconstitution. This study was designed to understand the reconstitution rate and function of these populations post TBI in melanoma tumor-bearing mice. Reconstitution rate and suppressive activity of CD4(+)CD25(+)Foxp3(+) Tregs and CD11b(+)Gr1(+) MDSC following TBI-induced lymphopenia was measured in B16 melanoma tumor-bearing mice. To ablate the rapid reconstitution of suppressive populations, we treated mice with docetaxel, a known chemotherapeutic agent that targets MDSC, in combination with adoptive T cell transfer and dendritic cell immunotherapy. Both Treg and MDSC populations exhibited rapid reconstitution after TBI-induced lymphopenia. Although reconstituted Tregs were just as suppressive as Tregs from untreated mice, MDSC demonstrated enhanced suppressive activity of CD8(+) T cell proliferation compared with endogenous MDSC from tumor-bearing mice. TBI-induced lymphopenia followed by docetaxel treatment improved the efficacy of adoptive T cell transfer and dendritic cell immunotherapy in melanoma-bearing mice, inducing a significant reduction in tumor growth and enhancing survival. Tumor regression correlated with increased CTL activity and persistence of adoptively transferred T cells. Overall, these findings suggest that TBI-induced MDSC are highly immunosuppressive and blocking their rapid reconstitution may improve the efficacy of vaccination strategies and adoptive immunotherapy.


Asunto(s)
Inmunoterapia Adoptiva , Linfopenia/inmunología , Melanoma Experimental/terapia , Neoplasias Cutáneas/terapia , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Animales , Antígenos CD/genética , Antígenos CD/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Proliferación Celular , Rayos gamma/uso terapéutico , Linfopenia/etiología , Linfopenia/patología , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Bazo/inmunología , Bazo/patología , Linfocitos T Reguladores/trasplante , Irradiación Corporal Total
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA