Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Craniofac Surg ; 34(3): e247-e249, 2023 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-36441840

RESUMEN

Immune checkpoint inhibitors (ICI) have become an essential part of cancer treatment. Anti-programmed cell death receptor-1 (PD-1) is a monoclonal antibody that targets PD-1. For patients with inoperable cutaneous squamous cell carcinoma, anti-PD-1 ICI treatment has been approved as monotherapy or in adjunct with other treatment modalities. A patient primarily treated with PD-1 inhibition for local advanced moderately differentiated cutaneous squamous cell carcinoma involving the parotid and the neck is presented. Surgical therapy would be extensive including pinnectomy, radical parotidectomy, neck dissection, resection of the temporal and mastoid bones, and free flap reconstruction. Anti-PD-1 ICI was started as a first-line treatment and a complete clinical response was observed after 5 cycles of treatment. The patient is disease-free within the follow-up period of 17 months. Although a complete response to anti-PD-1 receptor antibodies was observed, off-target toxicities are a risk and not all patients will benefit from a response.


Asunto(s)
Antineoplásicos Inmunológicos , Carcinoma de Células Escamosas , Neoplasias Cutáneas , Humanos , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/cirugía , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/cirugía , Antineoplásicos Inmunológicos/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico
2.
Cancers (Basel) ; 13(17)2021 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-34503145

RESUMEN

Adenoid cystic carcinoma (ACC) is a rare cancer of secretory glands. Recurrent or metastatic (R/M) ACC is generally considered resistant to cytotoxic chemotherapy. Recent phase II studies have reported improved objective response rates (ORR) with the use of the multi-kinase inhibitor lenvatinib. We sought to evaluate real-world experience of R/M ACC patients treated with lenvatinib monotherapy within the UK National Health Service (NHS) to determine the response rates by Response Evaluation Criteria of Solid Tumour (RECIST) and clinical outcomes. Twenty-three R/M ACC patients from eleven cancer centres were included. All treatment assessments for clinical decision making related to drug therapy were undertaken at the local oncology centre. Central radiology review was performed by an independent clinical trial radiologist and blinded to the clinical decision making. In contrast to previously reported ORR of 12-15%, complete or partial response was not observed in any patients. Eleven patients (52.4%) had stable disease and 5 patients (23.8%) had progression of disease as the best overall response. The median time on treatment was 4 months and the median survival from discontinuation was 1 month. The median PFS and OS from treatment initiation were 4.5 months and 12 months respectively. Multicentre collaborative studies such as this are required to evaluate rare cancers with no recommended standard of care therapy and variable disease courses.

3.
J Oral Pathol Med ; 49(9): 956-959, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32017243

RESUMEN

AIMS: Oral and/or oropharyngeal acute mucositis during and after chemo-radiotherapy (chemo-RT) for head and neck squamous cell carcinoma (HNSCC) can be extremely painful, sometimes requiring nasogastric feeding to enable adequate nutrition. The MASCC/ISOO evidence-based guidelines recommend benzydamine mouthwash for mucositis prevention in RT (recently updated to include chemo-RT), and a Cochrane systematic review found other agents to be effective in prophylaxis. Diclofenac mouthwash is licensed for painful oral mucosal inflammatory conditions but to our knowledge has not been assessed in chemo-RT-associated oral mucositis. METHOD: A clinical observation and service evaluation study in 10 patients undergoing chemo-RT for HNSCC to assess the potential value of diclofenac mouthwash (0.74 mg/mL) in reducing symptoms. Patients used 20ml of mouthwash up to 4 times a day starting in week 3 (of a 6-week course of treatment), recording pain and discomfort scores using a visual analogue scale on days 0, 1,7 and 14 (until the end of week 4). As per our current clinical practice, oral mucositis was not clinically scored as an outcome. Statistical analysis was performed using a one-way ANOVA. RESULTS: Using diclofenac mouthwash, 9/10 patients experienced pain score reduction from day 0 (mean score 6.75 ± SD 1.83) to day 2 (5.05 ± SD 1.62) and day 14 (4.09 ± SD 1.96). CONCLUSIONS: Diclofenac mouthwash may be beneficial for managing chemo-RT-induced oral mucositis. While a prospective randomised clinical trial is needed, it can be prescribed for this condition within its current licence.


Asunto(s)
Neoplasias de Cabeza y Cuello , Traumatismos por Radiación , Estomatitis , Quimioradioterapia , Diclofenaco/uso terapéutico , Neoplasias de Cabeza y Cuello/radioterapia , Humanos , Antisépticos Bucales/uso terapéutico , Dolor/tratamiento farmacológico , Estudios Prospectivos , Traumatismos por Radiación/tratamiento farmacológico , Estomatitis/tratamiento farmacológico , Estomatitis/etiología , Estomatitis/prevención & control
4.
Mol Ther ; 27(6): 1139-1152, 2019 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-31053413

RESUMEN

A clinical oncolytic herpes simplex virus (HSV) encoding granulocyte-macrophage colony-stimulating factor (GM-CSF), talimogene laherparepvec, causes regression of injected and non-injected melanoma lesions in patients and is now licensed for clinical use in advanced melanoma. To date, limited data are available regarding the mechanisms of human anti-tumor immune priming, an improved understanding of which could inform the development of future combination strategies with improved efficacy. This study addressed direct oncolysis and innate and adaptive human immune-mediated effects of a closely related HSV encoding GM-CSF (HSVGM-CSF) alone and in combination with histone deacetylase inhibition. We found that HSVGM-CSF supported activation of anti-melanoma immunity via monocyte-mediated type I interferon production, which activates NK cells, and viral maturation of immature dendritic cells (iDCs) into potent antigen-presenting cells for cytotoxic T lymphocyte (CTL) priming. Addition of the histone deacetylase inhibitor valproic acid (VPA) to HSVGM-CSF treatment of tumor cells increased viral replication, viral GM-CSF production, and oncolysis and augmented the development of anti-tumor immunity. Mechanistically, VPA increased expression of activating ligands for NK cell recognition and induced expression of tumor-associated antigens, supporting innate NK cell killing and CTL priming. These data support the clinical combination of talimogene laherparepvec with histone deacetylase inhibition to enhance oncolysis and anti-tumor immunity.


Asunto(s)
Inhibidores de Histona Desacetilasas/uso terapéutico , Melanoma/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos/metabolismo , Simplexvirus/inmunología , Neoplasias Cutáneas/terapia , Ácido Valproico/uso terapéutico , Antígenos de Neoplasias/efectos de los fármacos , Antígenos de Neoplasias/metabolismo , Antineoplásicos Inmunológicos/uso terapéutico , Productos Biológicos/uso terapéutico , Supervivencia Celular/genética , Células Dendríticas/inmunología , Quimioterapia Combinada , Vectores Genéticos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Herpesvirus Humano 1 , Humanos , Interferón Tipo I/metabolismo , Células Asesinas Naturales/inmunología , Células MCF-7 , Melanoma/patología , Virus Oncolíticos/genética , Simplexvirus/genética , Neoplasias Cutáneas/patología , Linfocitos T Citotóxicos/inmunología
5.
Mol Ther ; 22(11): 1936-48, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25059678

RESUMEN

Previously, we showed that vesicular stomatitis virus (VSV) engineered to express a cDNA library from human melanoma cells (ASMEL, Altered Self Melanoma Epitope Library) was an effective systemic therapy to treat subcutaneous (s.c.) murine B16 melanomas. Here, we show that intravenous treatment with the same ASMEL VSV-cDNA library was an effective treatment for established intra-cranial (i.c.) melanoma brain tumors. The optimal combination of antigens identified from the ASMEL which treated s.c. B16 tumors (VSV-N-RAS+VSV-CYTC-C+VSV-TYRP-1) was ineffective against i.c. B16 brain tumors. In contrast, combination of VSV-expressed antigens-VSV-HIF-2α+VSV-SOX-10+VSV-C-MYC+VSV-TYRP1-from ASMEL which was highly effective against i.c. B16 brain tumors, had no efficacy against the same tumors growing subcutaneously. Correspondingly, i.c. B16 tumors expressed a HIF-2α(Hi), SOX-10(Hi), c-myc(Hi), TYRP1, N-RAS(lo)Cytc(lo) antigen profile, which differed significantly from the HIF-2α(lo), SOX-10(lo), c-myc(lo), TYRP1, N-RAS(Hi)Cytc(Hi) phenotype of s.c. B16 tumors, and was imposed upon the tumor cells by CD11b(+) cells within the local brain tumor microenvironment. Combining T-cell costimulation with systemic VSV-cDNA treatment, long-term cures of mice with established i.c. tumors were achieved in about 75% of mice. Our data show that the anatomical location of a tumor profoundly affects the profile of antigens that it expresses.


Asunto(s)
Antígenos Virales de Tumores/metabolismo , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/terapia , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Vesiculovirus/genética , Administración Intravenosa , Animales , Línea Celular Tumoral , Biblioteca de Genes , Vectores Genéticos/administración & dosificación , Humanos , Inmunoterapia/métodos , Ratones , Ratones Endogámicos C57BL , Especificidad de Órganos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Microambiente Tumoral , Vesiculovirus/metabolismo
6.
Mol Ther ; 22(10): 1851-63, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24957982

RESUMEN

Optimum clinical protocols require systemic delivery of oncolytic viruses in the presence of an intact immune system. We show that preconditioning with immune modulators, or loading virus onto carrier cells ex vivo, enhances virus-mediated antitumor activity. Our early trials of systemic reovirus delivery showed that after infusion reovirus could be recovered from blood cells--but not from plasma--suggesting that rapid association with blood cells may protect virus from neutralizing antibody. We therefore postulated that stimulation of potential carrier cells directly in vivo before intravenous viral delivery would enhance delivery of cell-associated virus to tumor. We show that mobilization of the CD11b(+) cell compartment by granulocyte macrophage-colony stimulating factor immediately before intravenous reovirus, eliminated detectable tumor in mice with small B16 melanomas, and achieved highly significant therapy in mice bearing well-established tumors. Unexpectedly, cytokine conditioning therapy was most effective in the presence of preexisting neutralizing antibody. Consistent with this, reovirus bound by neutralizing antibody effectively accessed monocytes/macrophages and was handed off to tumor cells. Thus, preconditioning with cytokine stimulated recipient cells in vivo for enhanced viral delivery to tumors. Moreover, preexisting neutralizing antibody to an oncolytic virus may, therefore, even be exploited for systemic delivery to tumors in the clinic.


Asunto(s)
Citocinas/metabolismo , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Virus Oncolíticos/genética , Transducción Genética , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/metabolismo , Anticuerpos Antivirales , Antígeno CD11b/metabolismo , Citocinas/farmacología , Citotoxicidad Inmunológica/efectos de los fármacos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Inmunidad/efectos de los fármacos , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Orthoreovirus Mamífero 3/genética , Orthoreovirus Mamífero 3/inmunología , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , Melanoma Experimental/mortalidad , Melanoma Experimental/patología , Melanoma Experimental/terapia , Ratones , Monocitos/efectos de los fármacos , Monocitos/inmunología , Monocitos/metabolismo , Viroterapia Oncolítica , Virus Oncolíticos/inmunología , Receptores Fc/genética , Receptores Fc/metabolismo , Carga Tumoral
7.
Nat Med ; 19(12): 1625-1631, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24240185

RESUMEN

Tumor recurrence represents a major clinical challenge. Our data show that emergent recurrent tumors acquire a phenotype radically different from that of their originating primary tumors. This phenotype allows them to evade a host-derived innate immune response elicited by the progression from minimal residual disease (MRD) to actively growing recurrence. Screening for this innate response predicted accurately in which mice recurrence would occur. Premature induction of recurrence resensitized MRD to the primary therapy, suggesting a possible paradigm shift for clinical treatment of dormant disease in which the current expectant approach is replaced with active attempts to uncover MRD before evolution of the escape phenotype is complete. By combining screening with second-line treatments targeting innate insensitivity, up to 100% of mice that would have otherwise relapsed were cured. These data may open new avenues for early detection and appropriately timed, highly targeted treatment of tumor recurrence irrespective of tumor type or frontline treatment.


Asunto(s)
Inmunidad Innata/fisiología , Melanoma Experimental/diagnóstico , Recurrencia Local de Neoplasia/diagnóstico , Neoplasias Cutáneas/diagnóstico , Linfocitos T/inmunología , Escape del Tumor/fisiología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Detección Precoz del Cáncer/métodos , Femenino , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/inmunología , Neoplasia Residual , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Células Tumorales Cultivadas
8.
J R Soc Med ; 106(8): 310-4, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23824333

RESUMEN

Viruses that selectively replicate in cancer cells, leading to the death of the cell, are being studied for their potential as cancer therapies. Some of these viruses are naturally occurring but cause little if any illness in humans; others have been engineered to make them specifically able to kill cancer cells while sparing normal cells. These oncolytic viruses may be selective for cancer cells because viral receptors are over-expressed on the surface of cancer cells or because antiviral pathways are distorted in cancer cells. Additionally, when oncolytic viruses kill cancer cells, it can stimulate an antitumour immune response from the host that can enhance efficacy. Numerous early phase trials of at least six oncolytic viruses have been reported with no evidence of concerning toxicity either as single agents or in combination with chemotherapies and radiotherapy. Three oncolytic viruses have reached randomized testing in cancer patients; reolysin in head and neck cancer and JX594 in hepatocellular cancers, while results from the first-phase III trial of T-vec in metastatic melanoma are expected shortly.


Asunto(s)
Neoplasias/terapia , Viroterapia Oncolítica , Virus Oncolíticos , Neoplasias de Cabeza y Cuello/terapia , Humanos , Neoplasias Hepáticas/terapia , Melanoma/terapia
11.
Sci Transl Med ; 4(138): 138ra77, 2012 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-22700953

RESUMEN

Oncolytic viruses, which preferentially lyse cancer cells and stimulate an antitumor immune response, represent a promising approach to the treatment of cancer. However, how they evade the antiviral immune response and their selective delivery to, and replication in, tumor over normal tissue has not been investigated in humans. Here, we treated patients with a single cycle of intravenous reovirus before planned surgery to resect colorectal cancer metastases in the liver. Tracking the viral genome in the circulation showed that reovirus could be detected in plasma and blood mononuclear, granulocyte, and platelet cell compartments after infusion. Despite the presence of neutralizing antibodies before viral infusion in all patients, replication-competent reovirus that retained cytotoxicity was recovered from blood cells but not plasma, suggesting that transport by cells could protect virus for potential delivery to tumors. Analysis of surgical specimens demonstrated greater, preferential expression of reovirus protein in malignant cells compared to either tumor stroma or surrounding normal liver tissue. There was evidence of viral factories within tumor, and recovery of replicating virus from tumor (but not normal liver) was achieved in all four patients from whom fresh tissue was available. Hence, reovirus could be protected from neutralizing antibodies after systemic administration by immune cell carriage, which delivered reovirus to tumor. These findings suggest new preclinical and clinical scheduling and treatment combination strategies to enhance in vivo immune evasion and effective intravenous delivery of oncolytic viruses to patients in vivo.


Asunto(s)
Virus Oncolíticos/fisiología , Anciano , Anticuerpos Neutralizantes/inmunología , Plaquetas/virología , Neoplasias Colorrectales/cirugía , Neoplasias Colorrectales/terapia , Femenino , Genoma Viral/genética , Granulocitos/virología , Humanos , Neoplasias Hepáticas/cirugía , Neoplasias Hepáticas/terapia , Masculino , Persona de Mediana Edad , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Virus Oncolíticos/crecimiento & desarrollo , Replicación Viral/genética , Replicación Viral/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA