Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Clin Lab Med ; 43(2): 299-321, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37169447

RESUMEN

Predictive biomarker testing on metastatic breast cancer is essential for determining patient eligibility for targeted therapeutics. The National Comprehensive Cancer Network currently recommends assessment of specific biomarkers on metastatic tumor subtypes, including hormone receptors, HER2, and BRCA1/2 mutations, on all newly metastatic breast cancers subtypes; programmed death-ligand 1 on metastatic triple-negative carcinomas; and PIK3CA mutation status on estrogen receptor-positive carcinomas. In select circumstances mismatch repair protein deficiency and/or microsatellite insufficiency, tumor mutation burden, and NTRK translocation status are also testing options. Novel biomarker testing, such as detecting PIK3CA mutations in circulating tumor DNA, is expanding in this rapidly evolving arena.


Asunto(s)
Neoplasias de la Mama , Carcinoma , Humanos , Femenino , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/genética , Neoplasias de la Mama/terapia , Proteína BRCA1 , Proteína BRCA2 , Biomarcadores de Tumor/genética , Carcinoma/genética
2.
Surg Pathol Clin ; 15(1): 105-120, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35236627

RESUMEN

Predictive biomarker testing on metastatic breast cancer is essential for determining patient eligibility for targeted therapeutics. The National Comprehensive Cancer Network currently recommends assessment of specific biomarkers on metastatic tumor subtypes, including hormone receptors, HER2, and BRCA1/2 mutations, on all newly metastatic breast cancers subtypes; programmed death-ligand 1 on metastatic triple-negative carcinomas; and PIK3CA mutation status on estrogen receptor-positive carcinomas. In select circumstances mismatch repair protein deficiency and/or microsatellite insufficiency, tumor mutation burden, and NTRK translocation status are also testing options. Novel biomarker testing, such as detecting PIK3CA mutations in circulating tumor DNA, is expanding in this rapidly evolving arena.


Asunto(s)
Neoplasias de la Mama , Carcinoma , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/genética , Neoplasias de la Mama/terapia , Carcinoma/genética , Femenino , Humanos , Mutación
3.
JAMA Netw Open ; 4(4): e216322, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33856473

RESUMEN

Importance: Overtreatment of early-stage breast cancer with favorable tumor biology in older patients may be harmful without affecting recurrence and survival. Guidelines that recommend deimplementation of sentinel lymph node biopsy (SLNB) (Choosing Wisely) and radiotherapy (RT) (National Comprehensive Cancer Network) have been published. Objective: To describe the use rates and association with disease recurrence of SLNB and RT in older women with breast cancer. Design, Setting, and Participants: This cohort study obtained patient and clinical data from an integrated cancer registry and electronic health record of a single health care system in Pennsylvania. The cohort was composed of consecutive female patients 70 years or older who were diagnosed with early-stage, estrogen receptor-positive, ERBB2 (formerly HER2)-negative, clinically node-negative breast cancer from January 1, 2010, to December 31, 2018, who were treated at 15 community and academic hospitals within the health system. Exposures: Sentinel lymph node biopsy and adjuvant RT. Main Outcomes and Measures: Primary outcomes were 5-year locoregional recurrence-free survival (LRFS) rate and disease-free survival (DFS) rate after SLNB and after RT. Secondary outcomes included recurrence rate, subgroups that may benefit from SLNB or RT, and use rate of SLNB and RT over time. Propensity scores were used to create 2 cohorts to separately evaluate the association of SLNB and RT with recurrence outcomes. Cox proportional hazards regression model was used to estimate hazard ratios (HRs). Results: From 2010 to 2018, a total of 3361 women 70 years or older (median [interquartile range {IQR}] age, 77.0 [73.0-82.0] years) with estrogen receptor-positive, ERBB2-negative, clinically node-negative breast cancer were included in the study. Of these women, 2195 (65.3%) received SLNB and 1828 (54.4%) received adjuvant RT. Rates of SLNB steadily increased (1.0% per year), a trend that persisted after the 2016 adoption of the Choosing Wisely guideline. Rates of RT decreased slightly (3.4% per year). To examine patient outcomes and maximize follow-up time, the analysis was limited to cases from 2010 to 2014, identifying 2109 patients with a median (IQR) follow-up time of 4.1 (2.5-5.7) years. In the propensity score-matched cohorts, no association was found between SLNB and either LRFS (HR, 1.26; 95% CI, 0.37-4.30; P = .71) or DFS (HR, 1.92; 95% CI, 0.86-4.32; P = .11). In addition, RT was not associated with LRFS (HR, 0.33; 95% CI, 0.09-1.24; P = .10) or DFS (HR, 0.99; 95% CI, 0.46-2.10; P = .97). Subgroup analysis showed that stratification by tumor grade or comorbidity was not associated with LRFS or DFS. Low absolute rates of recurrence were observed when comparing the groups that received SLNB (3.5%) and those that did not (4.5%) as well as the groups that received RT (2.7%) and those that did not (5.5%). Conclusions and Relevance: This study found that receipt of SLNB or RT was not associated with improved LRFS or DFS in older patients with ER-positive, clinically node-negative breast cancer. Despite limited follow-up time and wide 95% CIs, this study supports the continued deimplementation of both SLNB and RT in accordance with the Choosing Wisely and National Comprehensive Cancer Network guidelines.


Asunto(s)
Neoplasias de la Mama/terapia , Radioterapia Adyuvante/estadística & datos numéricos , Biopsia del Ganglio Linfático Centinela/estadística & datos numéricos , Procedimientos Innecesarios , Anciano , Anciano de 80 o más Años , Supervivencia sin Enfermedad , Femenino , Humanos , Recurrencia Local de Neoplasia , Evaluación de Resultado en la Atención de Salud , Supervivencia sin Progresión , Receptor ErbB-2 , Receptores de Estrógenos , Sistema de Registros , Estudios Retrospectivos
4.
J Immunother Cancer ; 6(1): 57, 2018 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-29914571

RESUMEN

Immune checkpoint antagonists (CTLA-4 and PD-1/PD-L1) and CAR T-cell therapies generate unparalleled durable responses in several cancers and have firmly established immunotherapy as a new pillar of cancer therapy. To extend the impact of immunotherapy to more patients and a broader range of cancers, targeting additional mechanisms of tumor immune evasion will be critical. Adenosine signaling has emerged as a key metabolic pathway that regulates tumor immunity. Adenosine is an immunosuppressive metabolite produced at high levels within the tumor microenvironment. Hypoxia, high cell turnover, and expression of CD39 and CD73 are important factors in adenosine production. Adenosine signaling through the A2a receptor expressed on immune cells potently dampens immune responses in inflamed tissues. In this article, we will describe the role of adenosine signaling in regulating tumor immunity, highlighting potential therapeutic targets in the pathway. We will also review preclinical data for each target and provide an update of current clinical activity within the field. Together, current data suggest that rational combination immunotherapy strategies that incorporate inhibitors of the hypoxia-CD39-CD73-A2aR pathway have great promise for further improving clinical outcomes in cancer patients.


Asunto(s)
Adenosina/metabolismo , Biomarcadores de Tumor , Neoplasias/inmunología , Neoplasias/metabolismo , Transducción de Señal , Animales , Ensayos Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Humanos , Inmunomodulación/efectos de los fármacos , Inmunoterapia , Neoplasias/diagnóstico , Neoplasias/terapia , Transducción de Señal/efectos de los fármacos , Resultado del Tratamiento , Escape del Tumor
5.
Clin Cancer Res ; 24(3): 511-520, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28801472

RESUMEN

Immunotherapy is revolutionizing the management of multiple solid tumors, and early data have revealed the clinical activity of programmed cell death-1/programmed death ligand-1 (PD-1/PD-L1) antagonists in small numbers of patients with metastatic breast cancer. Clinical activity appears more likely if the tumor is triple negative, PD-L1+, and/or harbors higher levels of tumor-infiltrating leukocytes. Responses to atezolizumab and pembrolizumab appear to be durable in metastatic triple-negative breast cancer (TNBC), suggesting that these agents may transform the lives of responding patients. Current clinical efforts are focused on developing immunotherapy combinations that convert nonresponders to responders, deepen those responses that do occur, and surmount acquired resistance to immunotherapy. Identifying biomarkers that can predict the potential for response to single-agent immunotherapy, identify the best immunotherapy combinations for a particular patient, and guide salvage immunotherapy in patients with progressive disease are high priorities for clinical development. Smart clinical trials testing rational immunotherapy combinations that include robust biomarker evaluations will accelerate clinical progress, moving us closer to effective immunotherapy for almost all patients with breast cancer. Clin Cancer Res; 24(3); 511-20. ©2017 AACR.


Asunto(s)
Neoplasias de la Mama/terapia , Inmunoterapia , Animales , Biomarcadores de Tumor , Neoplasias de la Mama/etiología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/mortalidad , Estudios Clínicos como Asunto , Terapia Combinada , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Inmunomodulación , Inmunoterapia/efectos adversos , Inmunoterapia/métodos , Terapia Molecular Dirigida , Resultado del Tratamiento
6.
Int Immunopharmacol ; 46: 112-123, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28282575

RESUMEN

The tumor microenvironment (TME) is established and maintained through complex interactions between tumor cells and host stromal elements. Therefore, therapies that target multiple cellular components of the tumor may be most effective. Sorafenib, a multi-kinase inhibitor, alters signaling pathways in both tumor cells and host stromal cells. Thus, we explored the potential immune-modulating effects of sorafenib in a murine HER-2-(neu) overexpressing breast tumor model alone and in combination with a HER-2 targeted granulocyte-macrophage colony-stimulating factor (GM-CSF)-secreting vaccine (3T3neuGM). In vitro, sorafenib inhibited the growth of HER-2 overexpressing NT2.5 tumor cells, inducing apoptosis. Sorafenib also interfered with ERK MAPK, p38 MAPK, and STAT3 signaling, as well as cyclin D expression, but did not affect HER-2 or AKT signaling. In vivo, single agent sorafenib disrupted the tumor-associated vasculature and induced tumor cell apoptosis, effectively inducing the regression of established NT2.5 tumors in immune competent FVB/N mice. Immune depletion studies demonstrated that both CD4+ and CD8+ T cells were required for tumor regression. Sorafenib treatment did not impact the rate of tumor clearance induced by vaccination with 3T3neuGM in tumor-bearing FVB/N mice relative to either sorafenib treatment or vaccination alone. In vivo studies further demonstrated that sorafenib enhanced the accumulation of both CD4+ and CD8+ T cells into the TME of vaccinated mice. Together, these findings suggest that GM-CSF-secreting cellular immunotherapy may be integrated with sorafenib without impairing vaccine-based immune responses.


Asunto(s)
Neoplasias de la Mama/terapia , Linfocitos T CD4-Positivos/trasplante , Linfocitos T CD8-positivos/trasplante , Vacunas contra el Cáncer/inmunología , Inmunoterapia Adoptiva/métodos , Niacinamida/análogos & derivados , Compuestos de Fenilurea/uso terapéutico , Receptor ErbB-2/metabolismo , Animales , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Terapia Combinada , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Humanos , Inmunidad Celular , Ratones , Ratones Endogámicos , Neoplasias Experimentales , Niacinamida/uso terapéutico , Receptor ErbB-2/inmunología , Transducción de Señal/efectos de los fármacos , Sorafenib , Carga Tumoral , Microambiente Tumoral
7.
J Transl Med ; 10: 70, 2012 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-22490302

RESUMEN

The development of cancer has historically been attributed to genomic alterations of normal host cells. Accordingly, the aim of most traditional cancer therapies has been to destroy the transformed cells themselves. There is now widespread appreciation that the progressive growth and metastatic spread of cancer cells requires the cooperation of normal host cells (endothelial cells, fibroblasts, other mesenchymal cells, and immune cells), both local to, and at sites distant from, the site at which malignant transformation occurs. It is the balance of these cellular interactions that both determines the natural history of the cancer, and influences its response to therapy. This active tumor-host dynamic has stimulated interest in the tumor microenvironment as a key target for both cancer diagnosis and therapy. Recent data has demonstrated both that the presence of CD8⁺ T cells within a tumor is associated with a good prognosis, and that the eradication of all malignantly transformed cells within a tumor requires that the intra-tumoral concentration of cytolytically active CD8⁺ effector T cells remain above a critical concentration until every tumor cell has been killed. These findings have stimulated two initiatives in the field of cancer immunotherapy that focus on the tumor microenvironment. The first is the development of the immune score as part of the routine diagnostic and prognostic evaluation of human cancers, and the second is the development of combinatorial immune-based therapies that reduce tumor-associated immune suppression to unleash pre-existing or therapeutically-induced tumor immunity. In support of these efforts, the Society for the Immunotherapy of Cancer (SITC) is sponsoring a workshop entitled "Focus on the Target: The Tumor Microenvironment" to be held October 24-25, 2012 in Bethesda, Maryland. This meeting should support development of the immune score, and result in a position paper highlighting opportunities for the development of integrative cancer immunotherapies that sculpt the tumor microenvironment to promote definitive tumor rejection.


Asunto(s)
Inmunoterapia/métodos , Neoplasias/inmunología , Neoplasias/terapia , Microambiente Tumoral/inmunología , Proliferación Celular , Congresos como Asunto , Progresión de la Enfermedad , Humanos , Inmunidad/inmunología , Neoplasias/patología
8.
Curr Opin Investig Drugs ; 10(12): 1315-24, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19943203

RESUMEN

Whole tumor cells that secrete GM-CSF have been tested in clinical trials and have demonstrated early evidence of safety and clinical activity. The intradermal administration of these cells induces a massive infiltration of dendritic cells, which process and present tumor antigens to activate tumor-specific CD4+ and CD8+ T-cells. However, trial design flaws limit the phase III evaluation of this vaccine platform. Preclinical and clinical data suggest the development of GM-CSF-secreting tumor vaccines should be continued in combination with drugs that enhance vaccine activity by mitigating immune tolerance or augmenting costimulatory pathways of T-cell activation.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Neoplasias/terapia , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/efectos adversos , Ensayos Clínicos como Asunto/métodos , Células Dendríticas/inmunología , Diseño de Fármacos , Evaluación Preclínica de Medicamentos , Humanos , Tolerancia Inmunológica/efectos de los fármacos , Neoplasias/inmunología
9.
Curr Opin Mol Ther ; 9(5): 490-7, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17932813

RESUMEN

GemVax AS, a subsidiary of Pharmexa A/S, is developing GV-1001, an injectable formulation of a promiscuous MHC class II peptide derived from the telomerase reverse transcriptase catalytic subunit (hTERT), for the potential treatment of solid tumors, including pancreatic, liver and NSCLC. GV-1001 is currently undergoing phase II clinical trials for pancreatic, liver and NSCLC as well as a phase III trial for pancreatic cancer.


Asunto(s)
Adenocarcinoma/terapia , Neoplasias Pancreáticas/terapia , Telomerasa/inmunología , Adenocarcinoma/inmunología , Animales , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/efectos adversos , Vacunas contra el Cáncer/inmunología , Evaluación Preclínica de Medicamentos , Humanos , Inyecciones , Neoplasias Pancreáticas/inmunología , Telomerasa/administración & dosificación , Telomerasa/efectos adversos , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/efectos adversos , Vacunas de Subunidad/inmunología
10.
Expert Rev Vaccines ; 4(6): 831-41, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16372879

RESUMEN

The genomic revolution has advanced our understanding of breast cancer biology and the molecular basis of antitumor immunity. Trastuzumab, the first monoclonal antibody for breast cancer, is now a routine part of clinical care. Breast cancer vaccines may be more effective by actively recruiting both humoral and cellular immunity to the therapeutic effort. However, immunization alone is unlikely to have significant activity against established breast cancers, where it is limited by potent mechanisms of immune tolerance and the immunobiology of breast cancer itself. The next generation of clinical studies should integrate breast cancer vaccines with standard breast cancer drugs or novel immunotherapeutics in strategic doses and schedules that abrogate immune tolerance and groom the tumor microenvironment for a productive immune response.


Asunto(s)
Antígenos de Neoplasias/inmunología , Neoplasias de la Mama/terapia , Vacunas contra el Cáncer/uso terapéutico , Inmunoterapia/tendencias , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Antineoplásicos/administración & dosificación , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/inmunología , Vacunas contra el Cáncer/inmunología , Ciclofosfamida/administración & dosificación , Ciclofosfamida/uso terapéutico , Esquema de Medicación , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Tolerancia Inmunológica , Paclitaxel/administración & dosificación , Paclitaxel/uso terapéutico , Ensayos Clínicos Controlados Aleatorios como Asunto , Linfocitos T/inmunología , Trastuzumab
11.
Breast Cancer Res Treat ; 76(2): 145-56, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12452452

RESUMEN

PURPOSE: We studied sequential dose-dense doxorubicin, paclitaxel, and 5-fluorouracil (A-T-F) before high dose chemotherapy (HDC) with autologous peripheral blood stem cell support (PBSCT). Our aims were to determine the maximum tolerated dose (MTD) of 5-FU in the dose-dense regimen and to determine the impact of dose-dense chemotherapy on HDC/PBSCT. METHODS: Patients with Stage IIIB or Stage II or IIIA breast cancer with > or = 4 involved ipsilateral lymph nodes were treated with nine cycles of chemotherapy at 14-day intervals. The regimen was doxorubicin at 80 mg/m2 x 3, followed by paclitaxel at 140 mg/m2 over 96 h x 3, then 5-FU at doses of 1285, 1470, or 1655 mg/m2 by continuous intravenous infusion over 72 h x 3. Patients then underwent a G-CSF-stimulated peripheral blood stem cell (PBSC) apheresis prior to receiving HDC with autologous PBSCT. RESULTS: We identified 1285 mg/m2 as the MTD of 5-FU in this regimen. 5-FU-related DLTs included hand-foot syndrome, mucositis, and facial edema with somnolence. Unexpectedly, 3/19 treated patients developed congestive heart failure that prevented planned HDC. Compared to standard dose doxorubicin-containing adjuvant therapy, the dose-dense regimen also decreased CD34+ PBSC yields by about 40% (p = 0.049), requiring that 50% of patients have a supplemental bone marrow harvest. There was no difference in time to neutrophil, platelet, and red blood cell recovery after HDC. CONCLUSIONS: This regimen resulted in an unacceptably high rate of cardiac toxicity and is not recommended for further testing. It may be feasible to use a different schedule of 5-FU-containing dose-dense chemotherapy, particularly for the induction therapy of high-risk primary breast cancer prior to novel targeted therapies.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Adulto , Recuento de Células Sanguíneas , Diarrea/inducido químicamente , Doxorrubicina/administración & dosificación , Esquema de Medicación , Eritema/inducido químicamente , Femenino , Fluorouracilo/administración & dosificación , Fluorouracilo/efectos adversos , Humanos , Dosis Máxima Tolerada , Persona de Mediana Edad , Paclitaxel/administración & dosificación , Trastornos del Sueño-Vigilia/inducido químicamente , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA