Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Breast Cancer ; 17(5): 326-335, 2017 08.
Article in English | MEDLINE | ID: mdl-28365334

ABSTRACT

Locoregional recurrence (LRR) of breast cancer can occur after multidisciplinary treatment of a primary breast cancer. With modern multidisciplinary breast cancer treatment, the incidence of isolated LRR is decreasing. Improvements in systemic therapy are driving the decrease in LRR. LRR does still occur, however. LRR reflects biology of the cancer, as does systemic recurrence. LRR of breast cancer is frequently associated with systemic disease recurrence and poor prognosis. Given this associated poor prognosis, historically, it has been unclear whether patients with LRR would benefit from aggressive therapy with curative intent. Findings in retrospective studies suggest that prognosis for patients with LRR is not universally poor, and some patients may benefit from aggressive locoregional and systemic therapy. The challenge remains to assess prognosis and appropriately treat patients with locoregional breast cancer recurrence.


Subject(s)
Breast Neoplasms/therapy , Neoplasm Recurrence, Local/therapy , Combined Modality Therapy , Female , Humans , Prognosis
2.
Cancer Metastasis Rev ; 33(2-3): 791-807, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24944077

ABSTRACT

Endocrine therapy of breast cancer is perhaps the oldest form of effective and well-tolerated targeted cancer systemic treatment, in both the adjuvant and metastatic disease settings. The most commonly used endocrine therapy agents are selective estrogen receptor modulators, aromatase inhibitors, and selective estrogen receptor downregulators. De novo or acquired resistance to these agents is a significant clinical problem. Preclinical and clinical investigations to understand this resistance have yielded significant advances in understanding cell signaling and the possible mechanisms of resistance. These mechanisms of resistance are as diverse as the biology of breast cancer and can arise from alterations in any of the cell signaling pathway components. A growing understanding of these mechanisms has provided rationale for development of strategies to overcome the resistance. Many of these mechanisms of resistance involve adaptive upregulation of alternate signaling pathways, such as growth factor signaling, and cross talk between estrogen receptor and growth factor signaling. Clinical trials are focusing on cotargeting these alternate pathways along with estrogen receptor signaling. It is becoming evident that, as with all cancer therapy, strategies to overcome resistance need to be individualized, and it is important to identify biomarkers to guide the use of these strategies. This manuscript systemically reviews the recent preclinical and clinical trials on the novel and pathway-driven agents that have shown significant promise in enhancing the efficacy and overcoming the resistance in the hormonal treatment of breast cancer. Future directions including biomarker selection and the role of next generation sequencing will be discussed.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm , Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/metabolism , Clinical Trials as Topic , Female , Humans , Receptors, Estrogen/metabolism , Receptors, Growth Factor/metabolism , Signal Transduction/drug effects , Treatment Outcome
3.
Apoptosis ; 12(1): 55-71, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17136498

ABSTRACT

Apo2L/TRAIL is actively investigated as a novel targeted agent to directly induce apoptosis of susceptible cancer cells. Apo2L/TRAIL-refractory cells can be sensitized to the cytotoxic effect of this ligand by cytotoxic chemotherapeutics. The aim of this study was to evaluate the in vitro tumoricidal activity of the Apo2L/TRAIL + Trichostatin A in cultured thoracic cancer cells and to elucidate the molecular basis of the synergistic cytotoxicity of this combination. Concurrent exposure of cultured cancer cells to sublethal concentrations of Apo2L/TRAIL and Trichostatin A resulted in profound enhancement of Apo2L/TRAIL-mediated cytotoxicity in all cell lines regardless of their intrinsic susceptibility to this ligand. This combination was not toxic to primary normal cells. While Apo2L/TRAIL alone or Trichostatin A alone mediated < 20% cell death, 60 to 90% of cancer cells were apoptotic following treatment with TSA + Apo2L/TRAIL combinations. Complete translocation of Bax from the cytosol to the mitochondria compartment was mainly observed in combination-treated cells and this was correlated with robust elevation of caspase 9 proteolytic activity indicative of activation of the mitochondria apoptogenic effect. Profound TSA + Apo2L/TRAIL-mediated cytotoxicity and apoptosis were completely abrogated by either Bcl2 over-expression or by the selective caspase 9 inhibitor, highlighting the essential role of mitochondria-dependent apoptosis signaling cascade in this process. Moreover, increased caspase 8 activity observed in cells treated with the TSA + Apo2L/TRAIL combination was completely suppressed by Bcl-2 over-expression or by the selective caspase 9 inhibitor indicating that the elevated caspase 8 activity in combination-treated cells was secondary to a mitochondria-mediated amplification feedback loop of caspase activation. These finding form the basis for further development of HDAC inhibitors + Apo2L/TRAIL combination as novel targeted therapy for thoracic malignancies.


Subject(s)
Apoptosis/drug effects , Apoptosis/physiology , Hydroxamic Acids/administration & dosage , TNF-Related Apoptosis-Inducing Ligand/administration & dosage , Thoracic Neoplasms/drug therapy , Thoracic Neoplasms/metabolism , Caspases/metabolism , Cell Line, Tumor , Drug Synergism , Enzyme Activation , Enzyme Inhibitors/administration & dosage , Histone Deacetylase Inhibitors , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Recombinant Proteins/administration & dosage , Thoracic Neoplasms/pathology
4.
Cancer J ; 12(4): 257-73, 2006.
Article in English | MEDLINE | ID: mdl-16925970

ABSTRACT

PURPOSE: Despite adequately expressing functional receptors for tumor necrosis factor receptor apoptosis-inducing ligand (TRAIL), many cultured tumor cells are refractory to the cytotoxic effect of this ligand. Cytotoxic chemotherapeutic drugs have been shown to synergize with Apo2L/TRAIL to mediate apoptosis in cancer cells. The main goal of this study was to evaluate the effect of either cisplatin or paclitaxel, two common used chemotherapeutic agents for solid tumors, on enhancing Apo2L/TRAIL cytotoxicity in a panel-cultured thoracic cancer cells and to examine the role of the mitochondria-dependent caspase activation cascade in mediating apoptosis of combination-treated cells. METHODS: Cultured thoracic cancer cells were treated with cisplatin/Apo2L/TRAIL or paclitaxel/Apo2L/TRAIL sequential combinations in vitro. Cell viability and apoptosis were determined by 4,5-dimethylthiazo-2-yl)-2,5-diphenyl tetrazolium bromide and terminal deoxynucleotidyltransferase-mediated dUTP nick end labeling assays. Stable transfectants expressing high levels of Bcl-2 were created by retroviral gene transfer. Specific proteolytic activity of caspases 3, 6, 8, and 9 were measured by commercially available kits using fluorescent substrates. RESULTS: All cell lines preferentially expressed high levels of DR4 and/or DR5 and low levels of DcR1/DcR2; all of which were not altered by chemotherapeutic drug treatments. Pretreatment of these cancer cells with sublethal concentrations of either cisplatin or paclitaxel increased their susceptibility to Apo2L/TRAIL by twofold to >20-fold. Profound synergistic induction of apoptosis was observed in combination-treated cells. Viability of primary normal cells was affected by neither Apo2L/TRAIL nor the combinations of chemotherapy and Apo2L/TRAIL. Overexpression of Bcl-2 or inhibition of caspase 9 activity completely abrogated combination-induced cytotoxicity and apoptosis, indicating the essential role of the mitochondria-dependent death signaling cascade in this process. Robust activation of caspase 8 in combination-treated cells was completely suppressed either by Bcl-2 overexpression or by blocking of the activity of the mitochondria-regulated caspase 9, thus identifying the amplification feedback loop as the source of elevated caspase 8 activity. Finally, mitochondria-mediated amplification of caspase 8 activity was indispensable for complete caspase activation and full execution of apoptosis, because suppression of its activity using the selective caspase 6 inhibitor (located downstream of the caspase 3 but upstream of the caspase 8 in the feedback loop) resulted in profound suppression of not only caspase 8 activity but also those of caspases 9 and 3, as well as complete protection of cancer cells from combination-induced cytotoxicity. CONCLUSION: Cisplatin or paclitaxel synergistically interacts with Apo2L/TRAIL to mediate profound induction of apoptosis. The mitochondria-dependent caspase activation cascade and the amplification feedback loop are essential for the complete execution of the cell death program. Furthermore, our data identify mitochondria as the direct target for the development of more refined strategies to enhance the therapeutic effect of Apo2L/TRAIL as an anticancer agent.


Subject(s)
Apoptosis/drug effects , Caspase 8/metabolism , Signal Transduction/drug effects , TNF-Related Apoptosis-Inducing Ligand/metabolism , Thoracic Neoplasms , Antineoplastic Agents/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Cisplatin/pharmacology , Enzyme Activation/drug effects , Humans , Mitochondria/metabolism , Paclitaxel/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , TNF-Related Apoptosis-Inducing Ligand/genetics , Thoracic Neoplasms/drug therapy , Thoracic Neoplasms/metabolism , Thoracic Neoplasms/pathology
5.
Neoplasia ; 8(6): 446-57, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16820090

ABSTRACT

Inhibitors of histone deacetylases have been shown to enhance the sensitivity of cancer cells to tumor necrosis factor-related apoptosis-inducing ligand TRAIL-mediated cytotoxicity. Valproic acid (VA), a commonly used antiepileptic agent whose pharmacokinetics and toxicity profiles are well described, is a histone deacetylase inhibitor. This project aims to evaluate if VA can potentiate Apo2L/TRAIL-mediated cytotoxicity in cultured thoracic cancer cells and to elucidate the underlying molecular mechanism responsible for this effect. VA sensitized cultured thoracic cancer cells to Apo2L/TRAIL, as indicated by a 4-fold to a >20-fold reduction of Apo2L/TRAIL IC50 values in combination-treated cells. Although VA (0.5-5 mM) or Apo2L/TRAIL (20 ng/ml) induced less than 20% cell death, VA + Apo2L/TRAIL combinations caused 60% to 90% apoptosis of cancer cells. Moreover, substantial activation of caspases 8, 9, and 3, which was observed only in cells treated with the drug combination, was completely suppressed by Bcl2 overexpression or by the caspase 9 inhibitor. Both the caspase 9 inhibitor and Bcl2 completely abrogated the substantial cytotoxicity and apoptosis induced by this combination, thus highlighting the pivotal role of the type II pathway in this process. These findings provide a rationale for the development of VA and Apo2L/TRAIL combination as a novel molecular therapeutic for thoracic cancers.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Caspases/metabolism , Enzyme Inhibitors/pharmacology , Epilepsy/drug therapy , Histone Deacetylase Inhibitors , Lung Neoplasms/drug therapy , Mitochondria/enzymology , Thoracic Neoplasms/drug therapy , Valproic Acid/pharmacology , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Enzyme Activation , Humans , Membrane Glycoproteins/metabolism , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/metabolism
6.
J Thorac Cardiovasc Surg ; 128(6): 883-91, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15573073

ABSTRACT

BACKGROUND: Cancer cells frequently exhibit resistance to the cytotoxic effect of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Pretreatment of TRAIL-resistant cells with cisplatin sensitizes them to this ligand. Cisplatin also has been shown to enhance adenoviral transgene expression. OBJECTIVE: This study aims to evaluate the ability of cisplatin to enhance the expression and the cytotoxic effect of the tumor-specific adenoviral vector Ad/gTRAIL, which expresses a green fluorescent protein-TRAIL fusion protein. METHODS: Cultured cancer cells and normal human cells were infected with Ad/gTRAIL with or without cisplatin pretreatment. Adenoviral transgene expression was determined by using flow cytometry to measure green fluorescent protein fluorescence. Cytotoxicity was measured by using thiazolyl blue tetrazolium bromide assays and an apoptosis enzyme-linked immunosorbent assay kit. RESULTS: Green fluorescent protein-TRAIL fusion protein expression was significantly enhanced by cisplatin pretreatment in cancer cells. Cisplatin treatment before Ad/gTRAIL infection resulted in a 2- to 12-fold increase in green fluorescent protein fluorescence intensity across cancer lines. Although Ad/gTRAIL induced mild cytotoxicity in all cancer lines (inhibitory concentration of 50% values of >500 pfu/cell), pretreatment with cisplatin resulted in a dose-dependent enhancement of Ad/gTRAIL-mediated cytotoxicity, as indicated by the drastic reduction of inhibitory concentration of 50% values to 4 to 42 pfu/cell in all cell lines. There was no cytotoxicity noted in normal cells treated with both cisplatin and Ad/gTRAIL. CONCLUSION: Cisplatin pretreatment enhances Ad/gTRAIL cytotoxicity in malignant cells while not affecting normal cells. The mechanisms underlying this effect might include both enhancement of the susceptibility of cisplatin-treated cells to TRAIL and cisplatin-mediated enhancement of TRAIL expression in Ad/gTRAIL infected cells. These findings provide a rationale for development of Ad/gTRAIL-based therapy for thoracic malignancies.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cisplatin/pharmacology , Membrane Glycoproteins/drug effects , Tumor Necrosis Factor-alpha/drug effects , Adenoviridae , Apoptosis Regulatory Proteins , Carcinoma, Non-Small-Cell Lung/metabolism , Dose-Response Relationship, Drug , Flow Cytometry , Genetic Vectors , Green Fluorescent Proteins , Humans , Ligands , Lung Neoplasms/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor/metabolism , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/biosynthesis
7.
J Vasc Surg ; 35(3): 573-5, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11877709

ABSTRACT

We report the case of a patient with congenital absence of the external carotid artery in whom we performed a carotid endarterectomy. The radiographic features and operative findings are presented. Four similar cases previously reported in the literature are reviewed. A comment on the pathophysiology of atherosclerosis at the carotid bulb in the absence of a bifurcation and a brief discussion on the possible embryologic explanation of this anomaly are discussed.


Subject(s)
Carotid Artery, External/abnormalities , Aged , Arteriosclerosis/complications , Arteriosclerosis/diagnosis , Carotid Stenosis/complications , Carotid Stenosis/diagnosis , Endarterectomy, Carotid , Humans , Male , Ultrasonography, Doppler, Duplex
SELECTION OF CITATIONS
SEARCH DETAIL
...