Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Br J Cancer ; 123(4): 525-533, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32523090

RESUMEN

BACKGROUND: This phase 1 study examined the safety, maximum-tolerated dose (MTD) and antitumour activity of E7449, a novel PARP 1/2 and tankyrase 1/2 inhibitor. METHODS: E7449 was orally administered once daily in 28-day cycles to patients with advanced solid tumours (50-800-mg doses). Archival tumour samples from consenting patients were evaluated for the expression of 414 genes in a biomarker panel (2X-121 drug-response predictor [DRP]) found to be predictive of the response to E7449 in cell lines. RESULTS: Forty-one patients were enrolled (13 pancreatic, 5 ovarian, 4 each with breast, lung or colorectal cancer and 11 with other tumour types). The most common grade ≥3 treatment-related adverse event was fatigue (n = 7, 17.1%). Five patients experienced a dose-limiting toxicity (fatigue, n = 4, 800 mg; anaphylaxis, n = 1, 600 mg) for an MTD of 600 mg. E7449 exhibited antitumour activity in solid tumours, including 2 partial responses (PRs), and stable disease (SD) in 13 patients, which was durable (>23 weeks) for 8 patients. In 13 patients, the 2X-121 DRP identified those achieving PR and durable SD. E7449 showed good tolerability, promising antitumour activity and significant concentration-dependent PARP inhibition following 50-800-mg oral dosing. CONCLUSION: The results support further clinical investigation of E7449 and its associated biomarker 2X-121 DRP. CLINICAL TRIAL REGISTRATION: www.ClinicalTrials.gov code: NCT01618136.


Asunto(s)
Biomarcadores de Tumor/genética , Isoquinolinas/administración & dosificación , Neoplasias/tratamiento farmacológico , Inhibidores de Poli(ADP-Ribosa) Polimerasas/administración & dosificación , Quinazolinonas/administración & dosificación , Administración Oral , Adulto , Anciano , Compuestos Azo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Isoquinolinas/efectos adversos , Isoquinolinas/farmacología , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Neoplasias/genética , Inhibidores de Poli(ADP-Ribosa) Polimerasas/efectos adversos , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Quinazolinonas/efectos adversos , Quinazolinonas/farmacología , Análisis de Supervivencia , Resultado del Tratamiento
2.
Arthritis Rheumatol ; 75(12): 2185-2194, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37459248

RESUMEN

OBJECTIVE: Obexelimab is an investigational, bifunctional, noncytolytic monoclonal antibody that binds CD19 and FcyRIIb to inhibit B cells, plasmablasts, and plasma cells. This trial evaluated the efficacy and safety of obexelimab in the treatment of patients with systemic lupus erythematosus (SLE). METHODS: During screening, patients with active, non-organ-threatening SLE received corticosteroid injections to ameliorate symptoms while immunosuppressants were withdrawn (≤10 mg/day prednisone equivalent and ≤400 mg/day hydroxychloroquine allowed). Patients with improved disease activity were randomized 1:1 to obexelimab 5 mg/kg intravenously or placebo once every 2 weeks until week 32 or loss of improvement (LOI). RESULTS: In this study, 104 patients were randomized. Analysis of the primary endpoint, proportion of patients reaching week 32 without LOI, used an efficacy-evaluable (EE) population defined as patients who completed the study or withdrew for flare or treatment-related toxicity. This endpoint did not reach statistical significance: 21 of 50 obexelimab-treated patients (42.0%) versus 12 of 42 patients (28.6%) treated with a placebo (P = 0.183). Time to LOI was increased in obexelimab-treated patients versus patients treated with a placebo in the EE (hazard ratio [HR] 0.53, P = 0.025) and intention-to-treat (HR 0.59, P = 0.062) populations. In obexelimab-treated patients, B cells decreased approximately 50%, and trough concentration and inclusion in baseline gene expression clusters with high B cell pathway modules were associated with increased time to LOI. Obexelimab was associated with infusion reactions but was generally safe and well-tolerated. CONCLUSION: Although the primary endpoint was not reached, secondary analysis showed time to LOI was significantly increased in obexelimab-treated patients, and analysis of patient subsets defined by gene expression patterns at baseline suggests a responding subpopulation.


Asunto(s)
Anticuerpos Monoclonales , Lupus Eritematoso Sistémico , Humanos , Anticuerpos Monoclonales/uso terapéutico , Método Doble Ciego , Inmunosupresores/uso terapéutico , Lupus Eritematoso Sistémico/tratamiento farmacológico , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inducido químicamente , Prednisona/uso terapéutico , Resultado del Tratamiento
3.
Mol Cancer Ther ; 21(6): 914-924, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35313332

RESUMEN

Stimulator of interferon genes (STING) is an innate immune receptor activated by natural or synthetic agonists to elicit antitumoral immune response via type I IFNs and other inflammatory cytokines. Bacillus Calmette-Guerin (BCG) is the standard of care as intravesical therapy for patients with high-risk non-muscle invasive bladder cancer (NMIBC). There are limited options available for patients with NMIBC who developed BCG unresponsiveness. In this study, we characterized in vitro and in vivo antitumor effects of E7766, a macrocyle-bridged STING agonist, via intravesical instillation in two syngeneic orthotopic murine NMIBC tumor models resistant to therapeutic doses of BCG and anti-PD-1 agents. E7766 bound to recombinant STING protein with a Kd value of 40 nmol/L and induced IFNß expression in primary human peripheral blood mononuclear cells harboring any of seven major STING genotypes with EC50 values of 0.15 to 0.79 µmol/L. Intravesical E7766 was efficacious in both NMIBC models with induction of effective immunologic memory in the treated animals. Pharmacologic activation of the STING pathway in the bladder resulted in IFN pathway activation, infiltration of T cells and natural killer (NK) cells, dendritic cell activation, and antigen presentation in bladder epithelium, leading to the antitumor activity and immunity. In addition, measurements of the pharmacodynamic markers, Ifnß1 and CXCL10, in bladder, urine, and plasma, and of STING pathway intactness in cancer cells, supported this mode of action. Taken together, our studies reveal an antitumor immune effect of pharmacologic activation of the STING pathway in bladder epithelium and thus provide a rationale for subsequent clinical studies in patients with NMIBC.


Asunto(s)
Fosfatidilinositol 3-Quinasas , Neoplasias de la Vejiga Urinaria , Animales , Vacuna BCG/farmacología , Línea Celular Tumoral , Proliferación Celular , Humanos , Leucocitos Mononucleares/metabolismo , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Vejiga Urinaria/metabolismo , Vejiga Urinaria/patología , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/patología
4.
Cell Immunol ; 270(1): 62-9, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21550027

RESUMEN

A phase II trial was conducted in subjects with human papillomavirus (HPV) associated high-grade cervical dysplasia testing the safety and efficacy of a microparticle encapsulated pDNA vaccine. Amolimogene expresses T cell epitopes from E6 and E7 proteins of HPV types 16 and 18. An analysis was performed on a subset of HLA-A2+ subjects to test whether CD8+ T cells specific to HPV 16, 18, 6 and 11 were increased in response to amolimogene immunization. Of the 21 subjects receiving amolimogene, 11 had elevated CD8+ T cell responses to HPV 16 and/or 18 peptides and seven of these also had increases to corresponding HPV 6 and/or 11 peptides. In addition, T cells primed and expanded in vitro with an HPV 18 peptide demonstrated cross-reactivity to the corresponding HPV 11 peptide. These data demonstrate that treatment with amolimogene elicits T cell responses to HPV 16, 18, 6 and 11.


Asunto(s)
Presentación de Antígeno , Antígenos Virales/inmunología , Vacunas contra el Cáncer/inmunología , Papillomavirus Humano 16/inmunología , Papillomavirus Humano 18/inmunología , Linfocitos T/inmunología , Displasia del Cuello del Útero/inmunología , Displasia del Cuello del Útero/terapia , Neoplasias del Cuello Uterino/inmunología , Neoplasias del Cuello Uterino/terapia , Vacunas de ADN/inmunología , Adulto , Antígenos Virales/genética , Vacunas contra el Cáncer/administración & dosificación , Método Doble Ciego , Femenino , Papillomavirus Humano 11/inmunología , Papillomavirus Humano 6/inmunología , Humanos , Inmunización , Epítopos Inmunodominantes , Plásmidos , Poliglactina 910 , Células Precursoras de Linfocitos T/inmunología , Neoplasias del Cuello Uterino/virología , Vacunas de ADN/administración & dosificación , Adulto Joven , Displasia del Cuello del Útero/virología
7.
Eur J Cancer ; 75: 213-221, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28237867

RESUMEN

BACKGROUND: Lenvatinib significantly prolonged progression-free survival (PFS) versus placebo in the phase III Study of (E7080) LEnvatinib in differentiated Cancer of the Thyroid (SELECT) of patients with radioiodine-refractory differentiated thyroid cancer. This exploratory analysis investigated potential predictive biomarkers of lenvatinib efficacy and target engagement. PATIENTS AND METHODS: Circulating cytokine/angiogenic factors (CAFs) in blood samples collected at baseline and throughout treatment were analysed from patients randomised to receive lenvatinib or placebo from August 5, 2011 to October 4, 2012. For CAF biomarker analyses, patients were dichotomised by baseline levels. Tumour tissues were analysed for BRAF and NRAS/KRAS/HRAS mutations. RESULTS: Tumours and CAFs were analysed from 183/392 (47%) and 387/392 (99%) patients, respectively. Lenvatinib PFS benefit was maintained in all assessments. For lenvatinib-treated patients, interaction-term analyses revealed that low baseline Ang2 level was predictive of tumour shrinkage (Pinteraction = 0.016) and PFS (Pinteraction = 0.018). Vascular endothelial growth factor and fibroblast growth factor 23 (FGF23) were significantly upregulated with lenvatinib, and FGF23 upregulation on cycle 1/day 15 was associated with longer PFS. In mutation analyses, no significant differences in clinical outcomes were observed. BRAFWT may be a negative prognostic factor for PFS in placebo-treated patients with papillary thyroid cancer (P = 0.019). CONCLUSION: The lenvatinib PFS benefit was maintained regardless of baseline CAF or BRAF/RAS status. Baseline Ang2 was predictive of PFS in a subgroup of lenvatinib-treated patients, indicating that Ang2 may be predictive of lenvatinib sensitivity. BRAFWT may be a poor prognostic factor in patients with radioiodine-refractory papillary thyroid cancer. Improved PFS associated with upregulated FGF23 suggests that lenvatinib-induced FGF receptor inhibition contributes to lenvatinib efficacy. Trial registration ID of the main study, SELECT: ClinicalTrials.gov: NCT01321554.


Asunto(s)
Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/metabolismo , Compuestos de Fenilurea/uso terapéutico , Quinolinas/uso terapéutico , Neoplasias de la Tiroides/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Angiopoyetina 2/metabolismo , Supervivencia sin Enfermedad , Método Doble Ciego , Femenino , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/metabolismo , GTP Fosfohidrolasas/metabolismo , Humanos , Estimación de Kaplan-Meier , Masculino , Proteínas de la Membrana/metabolismo , Persona de Mediana Edad , Mutación/genética , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Neoplasias de la Tiroides/genética , Regulación hacia Arriba/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Adulto Joven
8.
Nat Commun ; 8(1): 103, 2017 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-28740126

RESUMEN

Muscle-invasive bladder cancer (MIBC) is an aggressive disease with limited therapeutic options. Although immunotherapies are approved for MIBC, the majority of patients fail to respond, suggesting existence of complementary immune evasion mechanisms. Here, we report that the PPARγ/RXRα pathway constitutes a tumor-intrinsic mechanism underlying immune evasion in MIBC. Recurrent mutations in RXRα at serine 427 (S427F/Y), through conformational activation of the PPARγ/RXRα heterodimer, and focal amplification/overexpression of PPARγ converge to modulate PPARγ/RXRα-dependent transcription programs. Immune cell-infiltration is controlled by activated PPARγ/RXRα that inhibits expression/secretion of inflammatory cytokines. Clinical data sets and an in vivo tumor model indicate that PPARγHigh/RXRαS427F/Y impairs CD8+ T-cell infiltration and confers partial resistance to immunotherapies. Knockdown of PPARγ or RXRα and pharmacological inhibition of PPARγ significantly increase cytokine expression suggesting therapeutic approaches to reviving immunosurveillance and sensitivity to immunotherapies. Our study reveals a class of tumor cell-intrinsic "immuno-oncogenes" that modulate the immune microenvironment of cancer.Muscle-invasive bladder cancer (MIBC) is a potentially lethal disease. Here the authors characterize diverse genetic alterations in MIBC that convergently lead to constitutive activation of PPARgamma/RXRalpha and result in immunosurveillance escape by inhibiting CD8+ T-cell recruitment.


Asunto(s)
Evasión Inmune/inmunología , Monitorización Inmunológica , PPAR gamma/inmunología , Receptor alfa X Retinoide/inmunología , Neoplasias de la Vejiga Urinaria/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Citocinas/genética , Citocinas/inmunología , Citocinas/metabolismo , Perfilación de la Expresión Génica/métodos , Células HCT116 , Humanos , Immunoblotting , Inmunoterapia/métodos , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Ratones , Microscopía Fluorescente , Mutación/inmunología , Invasividad Neoplásica , PPAR gamma/química , PPAR gamma/genética , Multimerización de Proteína/inmunología , Receptor alfa X Retinoide/química , Receptor alfa X Retinoide/genética , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/terapia
9.
Methods Mol Biol ; 302: 237-52, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15937356

RESUMEN

The interferon (IFN)-gamma enzyme-linked immunospot (ELISPOT) assay has become a useful tool for immunologists seeking to quantify immune responses on a per-cell basis. The assay is sensitive and allows for the enumeration of low-frequency T-cells. Many have applied this assay to clinical trials as a way to measure biological activity in a patient cohort. It is critical that each laboratory attempting to use the assay in their facility perform rigorous development and qualification work to establish an assay that suits their particular needs. This chapter serves as a demonstration of two practical and slightly different approaches to using the ELISPOT assay to monitor immune activity in the human periphery: (1) assays using whole samples of peripheral blood mononuclear cells with and without the use of additional antigen presenting cells and (2) assays using enriched T-cell populations. Detailed protocols and procedures will be covered, as well as a demonstration of results obtained from three separate applications.


Asunto(s)
Ensayo de Inmunoadsorción Enzimática/métodos , Interferón gamma/análisis , Interferón gamma/biosíntesis , Células Presentadoras de Antígenos/inmunología , Antígeno HLA-A2 , Humanos , Técnicas In Vitro , Leucocitos Mononucleares/inmunología , Subgrupos de Linfocitos T/inmunología
10.
Clin Cancer Res ; 8(5): 1028-37, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-12006515

RESUMEN

High-grade dysplasia induced by high-risk types of human papillomavirus (HPV) precedes invasive cancer in anal squamous epithelium just as it does in the cervix. A therapeutic HPV vaccine strategy as a potential treatment for anal dysplasia was tested in a standard Phase I dose escalation trial. The primary objective was to evaluate the safety of the agent; additional study aims were to evaluate the histological response, immune response, and effect on anal HPV-16 infection. Each subject was treated with four i.m. injections of 50-400 microg of ZYC101 at 3-week intervals. ZYC101 is composed of plasmid DNA encapsulated in biodegradable polymer microparticles. The plasmid DNA encodes for multiple HLA-A2-restricted epitopes derived from the HPV-16 E7 protein, one of two HPV oncoproteins consistently expressed in neoplastic cells. Fifty-six potential anal dysplasia subjects were screened to identify 12 eligible subjects with HPV-16 anal infection and a HLA-A2 haplotype. The investigational agent was well tolerated in all subjects at all dose levels tested. Three subjects experienced partial histological responses, including one of three subjects receiving the 200-microg dose and two subjects at the 400-microg dose level. Using a direct Elispot, 10 of 12 subjects demonstrated increased immune response to the peptide epitopes encoded within ZYC101; each continued to show elevated immune responses 6 months after the initiation of therapy. These results support the continued investigation of a therapeutic vaccination strategy for anal dysplasia.


Asunto(s)
Neoplasias del Ano/inmunología , Proteínas Oncogénicas Virales/inmunología , Infecciones por Papillomavirus/inmunología , Infecciones Tumorales por Virus/inmunología , Adulto , Anciano , Neoplasias del Ano/tratamiento farmacológico , Neoplasias del Ano/virología , ADN Viral/efectos de los fármacos , ADN Viral/genética , ADN Viral/metabolismo , Relación Dosis-Respuesta a Droga , Eritema/inducido químicamente , Fatiga/inducido químicamente , Femenino , Fiebre/inducido químicamente , Antígeno HLA-A2/inmunología , Cefalea/inducido químicamente , Humanos , Masculino , Microesferas , Persona de Mediana Edad , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/uso terapéutico , Dolor/inducido químicamente , Papillomaviridae/genética , Papillomaviridae/crecimiento & desarrollo , Papillomaviridae/inmunología , Proteínas E7 de Papillomavirus , Infecciones por Papillomavirus/tratamiento farmacológico , Infecciones por Papillomavirus/virología , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/uso terapéutico , Plásmidos/administración & dosificación , Plásmidos/genética , Factores de Tiempo , Resultado del Tratamiento , Infecciones Tumorales por Virus/diagnóstico , Infecciones Tumorales por Virus/tratamiento farmacológico , Vacunas de ADN/efectos adversos , Vacunas de ADN/inmunología , Vacunas de ADN/uso terapéutico
11.
Cell Rep ; 13(5): 1033-45, 2015 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-26565915

RESUMEN

Recurrent mutations in the spliceosome are observed in several human cancers, but their functional and therapeutic significance remains elusive. SF3B1, the most frequently mutated component of the spliceosome in cancer, is involved in the recognition of the branch point sequence (BPS) during selection of the 3' splice site (ss) in RNA splicing. Here, we report that common and tumor-specific splicing aberrations are induced by SF3B1 mutations and establish aberrant 3' ss selection as the most frequent splicing defect. Strikingly, mutant SF3B1 utilizes a BPS that differs from that used by wild-type SF3B1 and requires the canonical 3' ss to enable aberrant splicing during the second step. Approximately 50% of the aberrantly spliced mRNAs are subjected to nonsense-mediated decay resulting in downregulation of gene and protein expression. These findings ascribe functional significance to the consequences of SF3B1 mutations in cancer.


Asunto(s)
Empalme Alternativo , Mutación , Neoplasias/genética , Fosfoproteínas/genética , Ribonucleoproteína Nuclear Pequeña U2/genética , Alelos , Secuencia de Aminoácidos , Secuencia de Bases , Células HEK293 , Humanos , Datos de Secuencia Molecular , Tasa de Mutación , Degradación de ARNm Mediada por Codón sin Sentido , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Factores de Empalme de ARN , Ribonucleoproteína Nuclear Pequeña U2/química , Ribonucleoproteína Nuclear Pequeña U2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA