Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Gland Surg ; 7(2): 207-215, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29770314

RESUMO

BACKGROUND: Resection is the only option for potential cure in pancreatic cancer. Patients admitted for resection may have the procedure deferred during their hospitalization. We aim to identify factors that lead pancreatic cancer patients to undergo resection. METHODS: An analysis utilizing the Nationwide Inpatient Sample (NIS) database, 2003-2009. Study population included adults (≥18 years) with pancreatic cancer who underwent either pancreatic resection or other interventions. Surgeon volume classified based on the median into low and high-volume surgeon. RESULTS: Eleven thousand three hundred and sixty-five patients were included; 68.0% underwent pancreatic resection, while 32.0% had other interventions. The majority of patients resected were <60 years old, female, with higher annual household income (P<0.05 for all). Patients with Medicaid coverage and comorbidity scores ≥2 were least likely to undergo pancreatic resection. Resection was more likely for high-volume surgeons, high-volume hospitals and teaching hospitals (P<0.05 for all). Those managed by high-volume surgeons were at a lower risk of postoperative complications, lower mortality, shorter hospital stay, and lower healthcare costs (P<0.05 for all). CONCLUSIONS: Patients' insurance type and economic status are significantly associated with their ability to achieve pancreatic resection. Surgeon experience and hospital volumes were also significantly associated with pancreatic resection, clinical and economic outcomes.

2.
Anticancer Res ; 38(5): 2897-2901, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29715114

RESUMO

BACKGROUND/AIM: Immunotherapy combined with surgery is associated with better survival than surgery alone in patients with advanced melanoma. This study examined the utilization of immunotherapy in relation to population characteristics and the associated survival benefit. MATERIALS AND METHODS: This was a retrospective cohort study utilizing the US National Cancer Database. The study population included 6,165 adult patients (≥18 years) with stage III cutaneous melanoma (median follow-up=32 months). RESULTS: A total of 1,854 patients underwent immunotherapy in addition to surgery, which was associated with a survival benefit over surgery alone (hazard ratio(HR)=0.66, 95% confidence interval(CI)=0.56-0.77, p<0.001). Older age, presence of comorbidities, Medicaid/Medicare insurance, and living in a community with lower average education level were associated with less immunotherapy utilization (all p<0.05). No statistically significant racial disparity in immunotherapy usage was found (p=0.07). CONCLUSION: Compared to other demographic factors, insurance status was associated with the greatest disparities in immunotherapy utilization and mortality for patients who underwent surgery for advanced melanoma.


Assuntos
Imunoterapia/estatística & dados numéricos , Melanoma/terapia , Neoplasias Cutâneas/terapia , Adulto , Idoso , Estudos de Coortes , Feminino , Humanos , Estimativa de Kaplan-Meier , Masculino , Melanoma/mortalidade , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Modelos de Riscos Proporcionais , Estudos Retrospectivos , Neoplasias Cutâneas/mortalidade , Resultado do Tratamento , Estados Unidos , Melanoma Maligno Cutâneo
3.
Am Surg ; 83(6): 628-632, 2017 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-28637566

RESUMO

Liver resection for noncolorectal, nonneuroendocrine metastases remains controversial. Here, we evaluate a single institutional experience with hepatic resection for metastatic urologic malignancies. A single-institution review of patients who underwent hepatic resection for metastatic urologic tumors between the years of 2000 and 2013 was performed. Patient charts were analyzed for pathologic data and perioperative outcomes including short- and long-term morbidity, mortality, and overall and disease-free survival. Eleven patients were identified who underwent hepatic resection for metastatic urologic malignancy. The mean age was 63.5 years. All patients had an R0 resection. There were three major complications. Mean length of stay was 6.5 days and there was no 90-day mortality. Three patients have died of recurrent disease at an average of 11.2 months from resection to death. The remaining patients are still alive during a mean follow-up of 31.5 months. Five-year overall and disease-free survival was 50 and 21 months, respectively. Hepatic resection for metastatic urologic tumors is safe with low morbidity and mortality and durable long-term survival can be achieved. Liver resection for isolated hepatic disease should be considered for this rare metastatic disease to the liver.


Assuntos
Hepatectomia , Neoplasias Hepáticas/secundário , Neoplasias Hepáticas/cirurgia , Recidiva Local de Neoplasia/secundário , Recidiva Local de Neoplasia/cirurgia , Neoplasias Urológicas/patologia , Neoplasias Urológicas/cirurgia , Idoso , Intervalo Livre de Doença , Feminino , Seguimentos , Hepatectomia/métodos , Humanos , Neoplasias Hepáticas/mortalidade , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/mortalidade , Estudos Retrospectivos , Fatores de Risco , Resultado do Tratamento , Neoplasias Urológicas/mortalidade
4.
Case Rep Surg ; 2016: 5792980, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27200204

RESUMO

It is essential to identify any variant anatomy prior to surgery as this could have a drastic effect on surgical planning. We describe a case in which two vascular irregularities, an Arc of Buhler and celiac stenosis, were identified on angiogram after completion of a pancreaticoduodenectomy. While there could have been catastrophic results from his surgery in the setting of celiac stenosis, the presence of the aberrant Arc of Buhler allowed this patient to emerge without any permanent morbidity.

5.
World J Surg Oncol ; 13: 290, 2015 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-26419361

RESUMO

Pancreatic adenocarcinoma frequently recurs in patients even after resection with curative intent. The majority of these are early recurrences and are associated with metastatic disease, thus not amenable to repeat resection. Here we report a patient who underwent completion pancreatectomy for a metachronous pancreatic adenocarcinoma. This patient initially presented with painless jaundice and computed tomography (CT) revealed a mass in the head of the pancreas. Brushings obtained at endoscopic retrograde cholangiopancreatography (ERCP) were positive for adenocarcinoma. This patient then underwent a Whipple procedure and final pathology demonstrated stage III pancreatic ductal adenocarcinoma. Adjuvant therapy included gemcitabine and erlotinib. This patient was followed with physical examinations and serial laboratory and imaging studies. There was no evidence of disease for four years at which time and sharp elevation in CA-19-9 was found. Subsequent imaging revealed a mass in the remnant pancreas. Curative intent completion pancreatectomy was then performed which confirmed the presence of pancreatic adenocarcinoma. This was followed by adjuvant Gemcitabine based chemotherapy and chemoradiation. One year later the patient is alive with no evidence of disease. Thus, in highly selected patients with recurrent or metachronous pancreatic cancer, repeat pancreatectomy can be considered, but the course of treatment should be considered in a multidisciplinary setting.


Assuntos
Adenocarcinoma/cirurgia , Carcinoma Ductal Pancreático/cirurgia , Recidiva Local de Neoplasia/cirurgia , Segunda Neoplasia Primária , Neoplasias Pancreáticas/cirurgia , Pancreaticoduodenectomia , Adenocarcinoma/patologia , Adenocarcinoma/terapia , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma Ductal Pancreático/patologia , Carcinoma Ductal Pancreático/terapia , Terapia Combinada , Desoxicitidina/administração & dosagem , Desoxicitidina/análogos & derivados , Cloridrato de Erlotinib/administração & dosagem , Feminino , Humanos , Recidiva Local de Neoplasia/patologia , Recidiva Local de Neoplasia/terapia , Estadiamento de Neoplasias , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/terapia , Prognóstico , Gencitabina , Neoplasias Pancreáticas
6.
J Gastrointest Surg ; 19(9): 1699-703, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26001370

RESUMO

BACKGROUND: Hospital cancer registries are only required to report gastrointestinal stromal tumors (GISTs) if labeled malignant or metastatic, leading to potential loss of cases in national cancer registries. Our objective was to determine whether GISTs are underreported in the US. METHODS: Retrospective review of pathology reports between 2010 and 2013 with diagnosis of GIST was performed at two academic medical centers. Recurrent GISTs were excluded. Pathology reports were cross-referenced to cases reported by each cancer registry. Risk for metastasis/death was determined according to National Comprehensive Cancer Network (NCCN) guidelines. RESULTS: Forty-nine cases of non-recurrent GIST were identified. Only 19/49 (38.8%) cases were reported. None of the 30 non-reported cases were labeled malignant/metastatic on final pathology. To illustrate malignant potential, these tumors were risk stratified. Most (60%) of the non-reported cases were low risk, but there were 4 (13.3%) cases each in the intermediate, high, and unknown risk groups. Additionally, 7/30 (23.0%) cases were treated with tyrosine kinase inhibitors, highlighting clinical concern of malignant GIST. CONCLUSIONS: Our results show that nearly two thirds of GIST cases have been underreported, suggesting that current reporting practices underestimate its true incidence. Revision of reporting guidelines may result in a more accurate estimation of the US disease burden of GIST.


Assuntos
Neoplasias Gastrointestinais/diagnóstico , Neoplasias Gastrointestinais/epidemiologia , Tumores do Estroma Gastrointestinal/diagnóstico , Tumores do Estroma Gastrointestinal/epidemiologia , Neoplasias Gastrointestinais/terapia , Tumores do Estroma Gastrointestinal/terapia , Humanos , Incidência , Estudos Retrospectivos , Estados Unidos/epidemiologia
7.
J Gastrointest Surg ; 19(2): 251-7, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25421357

RESUMO

INTRODUCTION: In prior analyses, conditional survival (CS) estimates for gastric cancer have weighed clinical and pathologic factors to predict prognosis at time intervals after surgery. Since racial disparities in gastric cancer outcomes were not considered, our objective was to determine whether race influences CS estimates. METHODS: Data from the Surveillance, Epidemiology, and End Results cancer registry were used to identify gastric adenocarcinoma patients who underwent curative surgical intervention between 1988 and 2005. Five-year relative conditional survival (RCS) was computed for patients who survived at least 1 to 5 years after surgery. RCS was calculated by assessing observed and expected survival in an age- and race-matched standard population. Results were compared across time and racial groups (white, black, and Asian) using z test statistics. RESULTS: Of 14,067 patients, 63.8 % were white, 15.5 % black, and 20.7 % Asian. Racial disparities among groups were observed with improved survival of Asians at time point zero and improved RCS at 1 year. At 5 years following curative surgery, each racial group had increased RCS and the greatest magnitude of relative increase was observed in white and black patients (121 and 118 %, respectively). Comparison of RCS at the 5-year time point revealed a reduction of racial disparities in survival among the three groups. CONCLUSIONS: Our investigation shows that racial disparities in gastric cancer outcomes are pronounced at the time of curative surgery but diminish after years of survival, thus suggesting race has less influence over outcomes the longer patients survive. The reasons for reduction of racial disparities remain unclear and warrant future study.


Assuntos
Adenocarcinoma/etnologia , Adenocarcinoma/mortalidade , Disparidades nos Níveis de Saúde , Grupos Raciais/estatística & dados numéricos , Neoplasias Gástricas/etnologia , Neoplasias Gástricas/mortalidade , Adenocarcinoma/cirurgia , Adulto , Idoso , Povo Asiático/estatística & dados numéricos , População Negra/estatística & dados numéricos , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Programa de SEER , Neoplasias Gástricas/cirurgia , Taxa de Sobrevida , Fatores de Tempo , Estados Unidos/epidemiologia , População Branca/estatística & dados numéricos , Adulto Jovem
8.
Ann Surg Oncol ; 15(11): 3259-67, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18726131

RESUMO

BACKGROUND: We have shown that continuous systemic delivery of interferon beta (IFN-beta) remodels dysfunctional tumor vasculature, thereby improving tumor perfusion and enhancing delivery and efficacy of chemotherapeutic drugs. We hypothesized that because of their inherent tumor tropism, neural progenitor cells (NPCs) engineered to express IFN-beta could also effect maturation of tumor vasculature without generating high systemic levels of IFN-beta. METHODS: Mice with luciferase-expressing disseminated human neuroblastoma were divided into four groups of equal tumor burden by bioluminescence imaging: (1) untreated controls; (2) NPC-IFN-beta only; (3) cyclophosphamide (CTX) only; and (4) NPC-IFN-beta in combination with CTX. Two million NPC-IFN-beta cells were administered twice, 7 days apart, starting 21 days after tail vein administration of tumor cells. CTX was administered every 6 days for three doses. Mice were killed at 6 weeks, livers and kidneys weighed, and tumor removed for immunohistochemical staining for endothelial cells (CD34), pericytes (alpha-SMA), apoptosis (TUNEL [terminal deoxynucleotidyl transferase dUTP nick-end labeling]), and diI-labeled NPCs. RESULTS: Fluorescent-labeled NPCs confirmed localization of these cells to tumors. The alpha-SMA/CD34 ratio, a marker for vascular maturation, greatly increased in NPC-IFN-beta-treated tumors compared with controls. Bioluminescent signal from luciferase-expressing tumor cells, reflecting tumor burden, was lower with combination therapy than control or either monotherapy, and combination therapy resulted in far less tumor burden by weight in the kidneys and liver. CONCLUSIONS: Targeted delivery of IFN-beta with NPCs produced low circulating levels of IFN-beta, yet the maturing effect on the tumor vasculature and the enhanced efficacy of adjuvant therapy was maintained. Thus, combination therapy of NPC-IFN-beta with CTX warrants further investigation for the treatment of high-risk neuroblastoma patients.


Assuntos
Antineoplásicos Alquilantes/uso terapêutico , Antivirais/administração & dosagem , Ciclofosfamida/uso terapêutico , Interferon beta/administração & dosagem , Neuroblastoma/terapia , Neurônios/fisiologia , Células-Tronco/fisiologia , Adenoviridae/genética , Animais , Células Cultivadas , Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos/métodos , Terapia Genética , Vetores Genéticos/administração & dosagem , Humanos , Injeções Intravenosas , Masculino , Camundongos , Camundongos SCID , Neuroblastoma/genética , Neuroblastoma/patologia , Carga Tumoral
9.
J Pediatr Surg ; 43(1): 177-82; discussion 182-3, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18206478

RESUMO

PURPOSE: Trichostatin A (TSA) is a potent histone deacetylase inhibitor and has demonstrated significant antitumor activity against a variety of cancer cell lines. Type I interferons have also shown significant antitumor as well as antiangiogenic activity. In this study, we examined the effectiveness of combination therapy of TSA and interferon beta (IFN-beta) on human neuroblastoma cells in vitro and in vivo using a murine model of retroperitoneal neuroblastoma. MATERIALS AND METHODS: For in vitro experiments, plated human neuroblastoma cells (NB-1643 and NB-1691) were treated with vehicle or with IFN-beta, TSA, or both for 24 hours. Cytotoxicity was assessed by counting cells and expressing the results as a percentage of controls. Expression of the tumor suppressor p21(Waf1) was assessed by Western blot. For in vivo experiments, retroperitoneal neuroblastomas were established in severe combined immune deficiency (SCID) mice. Interferon beta was given using a gene therapy approach, administering 1.5 x 10(10) particles of an adeno-associated virus vector encoding human IFN-beta (AAV hIFN-beta) via tail vein as a single dose per mouse. Trichostatin A was given at a dose of 5 mg/kg every 48 hours subcutaneously. Treatment groups included controls, AAV hIFN-beta alone, TSA alone, and AAV hIFN-beta together with TSA. Tumor volume was assessed 2 weeks after the treatment began. RESULTS: After 24 hours, treatment with IFN-beta, TSA, and a combination of both resulted in a 45.3%, 68.1%, and 75% reduction in cell count relative to controls in the NB-1691 cell line. In the NB-1643 line, cell counts were reduced by 23%, 58%, and 62.3% respectively. In addition, NB-1691 cells treated with TSA showed increased expression of p21(Waf1) on Western blot. For in vivo experiments, control-, AAV hIFN-beta-, TSA-, and combination-treated tumors had the following final volumes: 1577.7 +/- 264.2 mm(3) (n = 3); 128.5 +/- 74.4 mm(3) (n = 4; P = .0001); 1248.7 +/- 673.9 mm(3) (n = 4; P = .48); and 127.5 +/- 36.8 mm(3) (n = 4; P = .0007), respectively. CONCLUSION: Neuroblastoma, because of its unique biology, continues to be a challenging tumor to treat, and many times these tumors are refractory to standard chemotherapeutic regimens. These data show that both TSA and IFN-beta inhibit neuroblastoma growth and that the combination may potentially provide a unique way to treat this difficult disease.


Assuntos
Terapia Genética/métodos , Ácidos Hidroxâmicos/farmacologia , Interferon beta/farmacologia , Neuroblastoma/terapia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Adenoviridae , Animais , Western Blotting , Linhagem Celular Tumoral/citologia , Linhagem Celular Tumoral/efeitos dos fármacos , Terapia Combinada , Modelos Animais de Doenças , Humanos , Técnicas In Vitro , Masculino , Camundongos , Camundongos SCID , Neuroblastoma/patologia , Probabilidade , Distribuição Aleatória , Sensibilidade e Especificidade
10.
Surgery ; 142(2): 185-91, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17689684

RESUMO

BACKGROUND: Bortezomib is a proteasome inhibitor with pleiotropic antitumor activity. Here we investigate the antiangiogenic and antitumor efficacy of bortezomib against neuroblastoma both in vitro and in a murine model of localized and disseminated disease. METHODS: In vitro activity of bortezomib was assessed by evaluating its effect on cell proliferation and cell cycle status. Localized tumor burden was followed with caliper measurements and total-body bioluminescence in mice with disseminated disease. The antiangiogenic activity was evaluated with immunohistochemistry and human vascular endothelial growth factor (VEGF) enzyme-linked immunosorbent assay on tumor protein extracts. RESULTS: Bortezomib treatment resulted in dose and time-dependent decreases in cell proliferation and resulted in cell cycle arrest. In vivo, bortezomib restricted tumor growth in a model of localized disease and decreased bioluminescence in mice with disseminated disease. That decreased bioluminescence reflected decreased tumor burden was confirmed at necropsy by assessing disease in specific organs. In addition, treatment resulted in a decrease in intratumoral vessel counts and reduced tumor VEGF expression. CONCLUSION: Bortezomib shows significant activity against neuroblastoma in vitro, and it inhibits tumor growth and angiogenesis in vivo. These results suggest that clinical studies of bortezomib are warranted for the treatment of this difficult disease.


Assuntos
Antineoplásicos/farmacologia , Ácidos Borônicos/farmacologia , Neovascularização Patológica/tratamento farmacológico , Neuroblastoma/tratamento farmacológico , Pirazinas/farmacologia , Neoplasias de Tecidos Moles/tratamento farmacológico , Animais , Bortezomib , Linhagem Celular Tumoral , Humanos , Técnicas In Vitro , Masculino , Camundongos , Camundongos SCID , Neovascularização Patológica/patologia , Neuroblastoma/patologia , Neoplasias de Tecidos Moles/patologia , Tela Subcutânea , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Clin Cancer Res ; 13(13): 3942-50, 2007 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-17606728

RESUMO

PURPOSE: Dysfunctional tumor vessels can be a significant barrier to effective cancer therapy. However, increasing evidence suggests that vascular endothelial growth factor (VEGF) inhibition can effect transient "normalization" of the tumor vasculature, thereby improving tumor perfusion and, consequently, delivery of systemic chemotherapy. We sought to examine temporal changes in tumor vascular function in response to the anti-VEGF antibody, bevacizumab. EXPERIMENTAL DESIGN: Established orthotopic neuroblastoma xenografts treated with bevacizumab were evaluated at serial time points for treatment-associated changes in intratumoral vascular physiology, penetration of systemically administered chemotherapy, and efficacy of combination therapy. RESULTS: After a single bevacizumab dose, a progressive decrease in tumor microvessel density to <30% of control was observed within 7 days. Assessment of the tumor microenvironment revealed a rapid, sustained decrease in both tumor vessel permeability and tumor interstitial fluid pressure, whereas intratumoral perfusion, as assessed by contrast-enhanced ultrasonography, was improved, although this latter change abated by 1 week. Intratumoral drug delivery mirrored these changes; penetration of chemotherapy was improved by as much as 81% when given 1 to 3 days after bevacizumab, compared with when both drugs were given concomitantly, or 7 days apart. Finally, administering topotecan to tumor-bearing mice 3 days after bevacizumab resulted in greater tumor growth inhibition (36% of control size) than with monotherapy (88% bevacizumab, 54% topotecan) or concomitant administration of the two drugs (44%). CONCLUSIONS: Bevacizumab-mediated VEGF blockade effects alterations in tumor vessel physiology that allow improved delivery and efficacy of chemotherapy, although careful consideration of drug scheduling is required to optimize antitumor activity.


Assuntos
Inibidores da Angiogênese/administração & dosagem , Inibidores da Angiogênese/farmacologia , Anticorpos Monoclonais/farmacologia , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Neuroblastoma/tratamento farmacológico , Animais , Anticorpos Monoclonais Humanizados , Bevacizumab , Linhagem Celular Tumoral , Humanos , Masculino , Camundongos , Microcirculação , Transplante de Neoplasias , Neovascularização Patológica , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Fenótipo , Pressão , Fator A de Crescimento do Endotélio Vascular/química , Fator A de Crescimento do Endotélio Vascular/metabolismo
12.
Mol Cancer Res ; 5(6): 531-42, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17579115

RESUMO

IFNs have pleiotropic antitumor mechanisms of action. The purpose of this study was to further investigate the effects of IFN-beta on the vasculature of human xenografts in immunodeficient mice. We found that continuous, systemic IFN-beta delivery, established with liver-targeted adeno-associated virus vectors, led to sustained morphologic and functional changes of the tumor vasculature that were consistent with vessel maturation. These changes included increased smooth muscle cell coverage of tumor vessels, improved intratumoral blood flow, and decreased vessel permeability, tumor interstitial pressure, and intratumoral hypoxia. Although these changes in the tumor vasculature resulted in more efficient tumor perfusion, further tumor growth was restricted, as the mature vasculature seemed to be unable to expand to support further tumor growth. In addition, maturation of the intratumoral vasculature resulted in increased intratumoral penetration of systemically administered chemotherapy. Finally, molecular analysis revealed increased expression by treated tumors of angiopoietin-1, a cytokine known to promote vessel stabilization. Induction of angiopoietin-1 expression in response to IFN-beta was broadly observed in different tumor lines but not in those with defects in IFN signaling. In addition, IFN-beta-mediated vascular changes were prevented when angiopoietin signaling was blocked with a decoy receptor. Thus, we have identified an alternative approach for achieving sustained vascular remodeling-continuous delivery of IFN-beta. In addition to restricting tumor growth by inhibiting further angiogenesis, maturation of the tumor vasculature also improved the efficiency of delivery of adjuvant therapy. These results have significant implications for the planning of combination anticancer therapy.


Assuntos
Interferon beta/administração & dosagem , Neoplasias/irrigação sanguínea , Neoplasias/tratamento farmacológico , Neovascularização Patológica , Angiopoietina-1/metabolismo , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Citocinas/metabolismo , Dependovirus/metabolismo , Humanos , Hipóxia , Interferon beta/uso terapêutico , Masculino , Camundongos , Camundongos SCID , Transplante de Neoplasias
13.
Ann Surg Oncol ; 14(6): 1904-8, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17342565

RESUMO

BACKGROUND: Despite recent advances in breast-conserving surgery, upper-extremity lymphedema remains a problem for patients after the treatment of breast cancer. This study examines the results of a protocol of therapy for lymphedema in breast cancer patients. METHODS: A total of 135 patients with lymphedema after breast cancer treatment were provided a protocol of complete decongestive therapy (CDT). This involved manual lymphatic drainage, compression garments, skin care, and range-of-motion exercises. Therapy was divided into an induction phase involving twice-weekly therapy for 8 weeks and maintenance therapy individualized to patient needs. Absolute volume and percentage of volume of lymphedema was compared before and after treatment. Also assessed was the degree of chronic pain and the need for pain medication. RESULTS: Mean initial lymphedema volume was 709 mL, and the percentage of lymphedema was 31%. The induction phase of CDT reduced this to 473 mL and 18%, respectively. Before therapy, 76 patients had chronic pain and 41 required oral pain medication. CDT reduced this to 20 and 11, respectively. The degree of pain was also assessed on a numerical scale from 0 to 10. Those patients with chronic pain initially rated their pain at an average of 6.9. After treatment, this was reduced to 1.1. CONCLUSIONS: Lymphedema continues to be a problem for patients with breast cancer. A program of lymphedema therapy can reduce the volume of edema and reduce pain in this population.


Assuntos
Neoplasias da Mama/cirurgia , Linfedema/terapia , Dor Pós-Operatória/terapia , Complicações Pós-Operatórias/terapia , Adulto , Idoso , Idoso de 80 Anos ou mais , Analgésicos/uso terapêutico , Doença Crônica , Protocolos Clínicos , Terapia Combinada , Drenagem , Terapia por Exercício , Feminino , Seguimentos , Humanos , Pessoa de Meia-Idade , Medição da Dor , Dor Pós-Operatória/tratamento farmacológico , Estudos Retrospectivos , Higiene da Pele , Meias de Compressão
14.
J Pediatr Surg ; 42(1): 48-53, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17208540

RESUMO

BACKGROUND: Interferon-beta (IFN-beta) has potent antitumor activity; however, systemic toxicity has limited its clinical use. We investigated the potential of targeted delivery using tumor-tropic neural progenitor cells (NPCs) transduced to express human IFN-beta (hIFN-beta). METHODS: Disseminated neuroblastoma was established in SCID mice by tail vein injection of tumor cells. Fourteen days after tumor cell inoculation, systemic disease was confirmed with bioluminescence imaging (BLI). Mice were then treated by intravenous injection of human F3.C1 NPCs that had been transduced with a replication deficient adenovirus to overexpress hIFN-beta (F3-IFN-beta). Two injections were given: the first at 14 days and the second at 28 days following tumor cell injection. Control mice received NPCs transduced with empty vector adenovirus at the same time points. Progression of disease was monitored using BLI. At sacrifice, organ weights and histology further evaluated tumor burden. RESULTS: After initiation of therapy, BLI demonstrated a significant decrease in the rate of disease progression in mice receiving F3-IFN-beta. At necropsy, control mice had bulky tumor replacing the liver and kidneys, as well as extensive retroperitoneal and mediastinal adenopathy. Impressively, these sites within mice receiving F3-IFN-beta therapy appeared grossly normal with the exception of small nodules within the kidneys of some of the F3-IFN-beta-treated mice. The accumulation of F3.C1 cells within sites of tumor growth was confirmed by fluorescence imaging. Importantly, systemic levels of hIFN-beta in the treated mice remained below detectable levels. CONCLUSIONS: These data indicate that in this model of disseminated neuroblastoma, the tumor-tropic property of F3.C1 NPCs was exploited to target delivery of IFN-beta to disseminated tissue foci, resulting in significant tumor growth delay. The described novel approach for effective IFN-beta therapy may circumvent limitations associated with the systemic toxicity of IFN-beta.


Assuntos
Antineoplásicos/administração & dosagem , Terapia Genética/métodos , Interferon beta/administração & dosagem , Células-Tronco/fisiologia , Adenoviridae , Animais , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular , Modelos Animais de Doenças , Progressão da Doença , Sistemas de Liberação de Medicamentos/métodos , Vetores Genéticos , Humanos , Injeções Intravenosas , Camundongos , Camundongos SCID , Neuroblastoma , Transdução Genética , Carga Tumoral
15.
J Pediatr Surg ; 42(1): 160-5; discussion 165, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17208558

RESUMO

BACKGROUND: We have recently demonstrated that continuous delivery of interferon beta (IFN-beta) stabilizes solid tumor vasculature and improves tumor perfusion. In this study, we have further investigated the functional consequences of this effect by assessing delivery and efficacy of conventional chemotherapy against neuroblastoma xenografts when used in combination with IFN-beta. METHODS: Mice with established retroperitoneal tumors received adeno-associated virus vector encoding IFN-beta (AAV IFN-beta) or control vector. One week later, at 1 hour before sacrifice, a 1 mg/kg i.v. bolus of topotecan (TPT) was given. Intratumoral levels of TPT were measured by high-performance liquid chromatography and then standardized to plasma levels to determine tumor TPT penetration. Subsequent experiments evaluated the antitumor efficacy of topotecan alone or in combination with AAV IFN-beta. RESULTS: As observed in prior experiments, AAV IFN-beta resulted in a marked increase in tumor vessel association with stabilizing perivascular smooth muscle cells. These more "matured" vessels facilitated improved tumor TPT penetration (51.2% +/- 4.2%) compared with controls (30.8% +/- 4.7%, P = .004). In additional cohorts of mice, this resulted in an improved antitumor effect. Mice with established tumors (301.8 +/- 18.1 mm3) were treated with TPT (1 mg/kg daily for 5 days for 2 consecutive weeks) either alone or in combination with AAV IFN-beta (5 x 10(10) vector particles per mouse). Topotecan monotherapy resulted in a reduction in mean tumor volume of 12% (264.2 +/- 65.8 mm3, P = .66). However, when the same regimen was administered to mice receiving continuous IFN-beta therapy, a 61% (118.9 +/- 42.3 mm3, P = .004) reduction in mean tumor volume was achieved. CONCLUSION: Interferon beta-mediated vessel stabilization resulted in improved intratumoral delivery of systemically administered TPT, enhancing its antitumor efficacy. This approach of altering the tumor vasculature provides a strategy to help overcome solid tumor resistance to traditional cytotoxic agents.


Assuntos
Antineoplásicos/farmacologia , Vasos Sanguíneos/efeitos dos fármacos , Interferon beta/farmacologia , Neuroblastoma/metabolismo , Topotecan/farmacologia , Adenoviridae , Animais , Modelos Animais de Doenças , Vetores Genéticos , Camundongos , Transplante de Neoplasias
16.
Surgery ; 140(2): 227-35, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16904974

RESUMO

BACKGROUND: We investigated the antitumor and antiosteoclastic effects of zoledronate against human neuroblastoma in vitro and in a murine model of bone metastasis. METHODS: Antitumor activity of zoledronate against neuroblastoma cell lines was assessed by evaluating proliferation, apoptosis, and cell-cycle progression. A murine model of bone invasion was used to assess antiosteoclastic and antitumor activity in vivo. Mice were followed by radiographic and bioluminescence imaging. RESULTS: Treatment of human neuroblastoma cells resulted in a decrease in cell count, increase in apoptosis, and arrest of cell-cycle progression. In the model of bone invasion, mice were treated weekly with zoledronate or vehicle control 10 days after tumor cell inoculation. Five weeks later, radiographs revealed a large degree of osteolytic disease in control mice. Impressively, mice treated with zoledronate demonstrated minimal radiographic changes at this time. Bioluminescence imaging of these mice revealed a significant restriction of tumor growth during the course of therapy. CONCLUSIONS: Zoledronate exhibits significant antitumor activity against human neuroblastoma cells in vitro, prevents development of osteolytic lesions, and restricts tumor growth in a murine model of bone metastasis. These results suggest that clinical investigation of zoledronate or similar bisphosphonates as adjuvant therapy in neuroblastoma patients is warranted.


Assuntos
Conservadores da Densidade Óssea/uso terapêutico , Neoplasias Ósseas/tratamento farmacológico , Difosfonatos/uso terapêutico , Imidazóis/uso terapêutico , Neuroblastoma/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Conservadores da Densidade Óssea/administração & dosagem , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Difosfonatos/administração & dosagem , Relação Dose-Resposta a Droga , Humanos , Imidazóis/administração & dosagem , Camundongos , Camundongos SCID , Transplante de Neoplasias , Neuroblastoma/patologia , Ácido Zoledrônico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...