Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Hernia ; 26(4): 1069-1075, 2022 08.
Article in English | MEDLINE | ID: mdl-34743254

ABSTRACT

PURPOSE: In 2003, randomized trials demonstrated potentially improved outcomes when local instead of general anesthesia is used for inguinal hernia repair. Our study aimed to evaluate how the use of local anesthesia for this procedure changed over time following the publication of the trials' level 1 evidence. METHODS: We used the 1998-2018 Veterans Affairs Surgical Quality Improvement Program database to identify adults who underwent open, unilateral inguinal hernia repair under local or general anesthesia. Our primary outcome was the percentage of cases performed under local anesthesia. We used a time-series design to examine the trend and rate of change of the use of local anesthesia. RESULTS: We included 97,437 veterans, of which 22,333 (22.9%) had hernia surgery under local anesthesia. The median age of veterans receiving local anesthesia remained stable at 64-67 years over time. The use of local anesthesia decreased steadily, from 38.2% at the beginning year to 15.1% in the final year (P < 0.0001). The publication of results from randomized trials (in 2003) did not appear to increase the overall use or change the rate of decline in the use of local anesthesia. Overall, we found that the use of local anesthesia decreased by about 1.5% per year. CONCLUSION: The utilization of local anesthesia for inguinal hernia repair in the VA has steadily declined over the last 20 + years, despite data showing equivalence or superiority to general anesthesia. Future studies should explore barriers to the use of local anesthesia for hernia repair.


Subject(s)
Hernia, Inguinal , Adult , Aged , Anesthesia, General , Anesthesia, Local/methods , Hernia, Inguinal/surgery , Herniorrhaphy/adverse effects , Herniorrhaphy/methods , Humans , Middle Aged , Time Factors
2.
Langenbecks Arch Surg ; 406(3): 597-605, 2021 May.
Article in English | MEDLINE | ID: mdl-33301071

ABSTRACT

PURPOSE: The reported conversion rates for minimally invasive distal pancreatectomy (MIDP) range widely from 2 to 38%. The identification of risk factors for conversion may help surgeons during preoperative planning and patient counseling. Moreover, the impact of conversion on outcomes of MIDP is unknown. METHODS: A systematic review was conducted as part of the 2019 Miami International Evidence-Based Guidelines on Minimally Invasive Pancreas Resection (IG-MIPR). The PubMed, Cochrane, and Embase databases were searched for studies concerning conversion to open surgery in MIDP. RESULTS: Of the 828 studies screened, eight met the eligibility criteria, resulting in a combined dataset including 2592 patients after MIDP. The overall conversion rate was 17.1% (range 13.0-32.7%) with heterogeneity between studies associated with the definition of conversion adopted. Only one study divided conversion into elective and emergency conversion. The main indications for conversion were vascular involvement (23.7%), concern for oncological radicality (21.9%), and bleeding (18.9%). The reported risk factors for conversion included a malignancy as an indication for surgery, the proximity of the tumor to vascular structures in preoperative imaging, higher BMI or visceral fat, and multi-organ resection or extended resection. Contrasting results were seen in terms of blood loss and length of stay in comparing converted MIDP and completed MIDP patients. CONCLUSION: The identified risk factors for conversion from this study can be used for patient selection and counseling. Surgeon experience should be considered when contemplating MIDP for a complex patient. Future studies should divide conversion into elective and emergency conversion.


Subject(s)
Laparoscopy , Pancreatic Neoplasms , Robotic Surgical Procedures , Humans , Pancreatectomy , Pancreatic Neoplasms/surgery , Risk Factors , Treatment Outcome
3.
Eur J Surg Oncol ; 44(9): 1378-1383, 2018 09.
Article in English | MEDLINE | ID: mdl-30131104

ABSTRACT

BACKGROUND: Peritoneal metastasis from biliary carcinoma (PMC) is associated with poor prognosis when treated with chemotherapy. OBJECTIVE: To evaluate the impact on survival of cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC), and compare with conventional palliative chemotherapy for patients with PMC. MATERIAL AND METHODS: A prospective multicenter international database was retrospectively searched to identify all patients with PMC treated with a potentially curative CRS/HIPEC (CRS/HIPEC group). The overall survival (OS) was compared to patients with PMC treated with palliative chemotherapy (systemic chemotherapy group). Survival was analyzed using Kaplan-Meier method and compared with Log-Rank test. RESULTS: Between 1995 and 2015, 34 patients were included in the surgical group, and compared to 21 in the systemic chemotherapy group. In the surgical group, median peritoneal cancer index was 9 (range 3-26), macroscopically complete resection was obtained for 25 patients (73%). There was more gallbladder localization in the surgical group compared to the chemotherapy group (35% vs. 18%, p = 0.001). Median OS was 21.4 and 9.3 months for surgical and chemotherapy group, respectively (p=0.007). Three-year overall survival was 30% and 10% for surgical and chemotherapy group, respectively. CONCLUSION: Treatment with CRS and HIPEC for biliary carcinoma with peritoneal metastasis is feasible and may provide survival benefit when compared to palliative chemotherapy.


Subject(s)
Bile Duct Neoplasms/therapy , Cytoreduction Surgical Procedures/methods , Hyperthermia, Induced/methods , Peritoneal Neoplasms/therapy , Registries , Adult , Aged , Aged, 80 and over , Bile Duct Neoplasms/mortality , Bile Duct Neoplasms/secondary , Female , Follow-Up Studies , France/epidemiology , Humans , Male , Middle Aged , Peritoneal Neoplasms/mortality , Peritoneal Neoplasms/secondary , Prognosis , Prospective Studies , Survival Rate/trends
4.
J Pancreat Cancer ; 3(1): 1-4, 2017.
Article in English | MEDLINE | ID: mdl-29082362

ABSTRACT

BACKGROUND: Pancreatic panniculitis is a rare cause of subcutaneous fat necrosis secondary to elevated serum levels of pancreatic enzymes. It is most often associated with pancreatic acinar cell carcinoma, but has also been seen in patients with pancreatitis. CASE REPORT: We present a case of a 64 year old Caucasian man without symptoms of pancreatitis who presents with pancreatic panniculitis manifesting in multiple subcutaneous ulcerating nodules of the bilateral lower extremities, discovered to have a previously unreported etiology for this condition. He had no evidence of pancreatitis or malignancy, but instead a pancreatic-portal fistula resulting in panniculitis. CONCLUSION: Peripancreatic vascular lesions must also be considered in the differential diagnosis of pancreatic panniculitis. The diagnosis, pathology, and treatment of pancreatic panniculitis are reviewed herein.

5.
Cancer Gene Ther ; 22(6): 326-34, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25908451

ABSTRACT

Neutrophil extracellular traps (NETs) are formed when neutrophils expel their DNA, histones and intracellular proteins into the extracellular space or circulation. NET formation is dependent on autophagy and is mediated by citrullination of histones to allow for the unwinding and subsequent expulsion of DNA. NETs have an important role in the pathogenesis of several sterile inflammatory diseases, including malignancy, therefore we investigated the role of NETs in the setting of pancreatic ductal adenocarcinoma (PDA). Neutrophils isolated from two distinct animal models of PDA had an increased propensity to form NETs following stimulation with platelet activating factor (PAF). Serum DNA, a marker of circulating NET formation, was elevated in tumor bearing animals as well as in patients with PDA. Citrullinated histone H3 expression, a marker of NET formation, was observed in pancreatic tumors obtained from murine models and patients with PDA. Inhibition of autophagy with chloroquine or genetic ablation of receptor for advanced glycation end products (RAGE) resulted in decreased propensity for NET formation, decreased serum DNA and decreased citrullinated histone H3 expression in the pancreatic tumor microenvironment. We conclude that NETs are upregulated in pancreatic cancer through RAGE-dependent/autophagy mediated pathways.


Subject(s)
Autophagy , Carcinoma, Pancreatic Ductal/physiopathology , Extracellular Traps/physiology , Neutrophils/physiology , Pancreatic Neoplasms/physiopathology , Receptor for Advanced Glycation End Products/physiology , Animals , Carcinoma, Pancreatic Ductal/immunology , Female , Humans , Mice , Mice, Knockout , Pancreatic Neoplasms/immunology , Receptor for Advanced Glycation End Products/genetics
6.
Cell Death Dis ; 5: e1480, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25341034

ABSTRACT

A hypoxic tumor microenvironment is characteristic of many cancer types, including one of the most lethal, pancreatic cancer. We recently demonstrated that the receptor for advanced glycation end products (RAGE) has an important role in promoting the development of pancreatic cancer and attenuating the response to chemotherapy. We now demonstrate that binding of RAGE to oncogenic KRAS facilitates hypoxia-inducible factor 1 (HIF1)α activation and promotes pancreatic tumor growth under hypoxic conditions. Hypoxia induces NF-κB-dependent and HIF1α-independent RAGE expression in pancreatic tumor cells. Moreover, the interaction between RAGE and mutant KRAS increases under hypoxia, which in turn sustains KRAS signaling pathways (RAF-MEK-ERK and PI3K-AKT), facilitating stabilization and transcriptional activity of HIF1α. Knock down of RAGE in vitro inhibits KRAS signaling, promotes HIF1α degradation, and increases hypoxia-induced pancreatic tumor cell death. RAGE-deficient mice have impaired oncogenic KRAS-driven pancreatic tumor growth with significant downregulation of the HIF1α signaling pathway. Our results provide a novel mechanistic link between NF-κB, KRAS, and HIF1α, three potent molecular pathways in the cellular response to hypoxia during pancreatic tumor development and suggest alternatives for preventive and therapeutic strategies.


Subject(s)
Oncogenes , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Immunologic/metabolism , Signal Transduction , ras Proteins/metabolism , Animals , Autophagy , Cell Hypoxia , Cell Line, Tumor , Cell Survival , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , NF-kappa B/metabolism , Protein Stability , Receptor for Advanced Glycation End Products , Transcriptional Activation
7.
Oncogene ; 33(5): 567-77, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-23318458

ABSTRACT

Tumor cells require increased adenosine triphosphate (ATP) to support anabolism and proliferation. The precise mechanisms regulating this process in tumor cells are unknown. Here, we show that the receptor for advanced glycation endproducts (RAGE) and one of its primary ligands, high-mobility group box 1 (HMGB1), are required for optimal mitochondrial function within tumors. We found that RAGE is present in the mitochondria of cultured tumor cells as well as primary tumors. RAGE and HMGB1 coordinately enhanced tumor cell mitochondrial complex I activity, ATP production, tumor cell proliferation and migration. Lack of RAGE or inhibition of HMGB1 release diminished ATP production and slowed tumor growth in vitro and in vivo. These findings link, for the first time, the HMGB1-RAGE pathway with changes in bioenergetics. Moreover, our observations provide a novel mechanism within the tumor microenvironment by which necrosis and inflammation promote tumor progression.


Subject(s)
Electron Transport Complex I/metabolism , HMGB1 Protein/metabolism , Pancreatic Neoplasms/pathology , Receptor for Advanced Glycation End Products/metabolism , Adenosine Triphosphate/biosynthesis , Adenosine Triphosphate/metabolism , Animals , Butadienes/pharmacology , CD24 Antigen/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cycloheximide/pharmacology , Electron Transport Complex I/antagonists & inhibitors , Energy Metabolism , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , HMGB1 Protein/drug effects , Humans , Inflammation/metabolism , MAP Kinase Kinase 2/genetics , MAP Kinase Kinase 2/metabolism , Mice , Mitochondria/drug effects , Mitochondria/metabolism , NF-kappa B/drug effects , NF-kappa B/metabolism , Nitriles/pharmacology , Pancreatic Neoplasms/metabolism , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Synthesis Inhibitors/pharmacology , RNA Interference , RNA, Small Interfering/genetics , Receptor for Advanced Glycation End Products/genetics , Rotenone/pharmacology , Signal Transduction , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics , Tumor Microenvironment , Uncoupling Agents
8.
Ann Surg Oncol ; 20(7): 2180-7, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23575907

ABSTRACT

BACKGROUND: Isolated hepatic perfusion (IHP) with melphalan is an established approach for patients with unresectable metastatic liver lesions. This study determined the safety and maximum tolerated dose (MTD) of 5-FU with oxaliplatin via IHP. METHODS: Standard 3 × 3 Phase I design. Subjects with unresectable isolated CRC liver metastases scheduled for HAI pump were eligible. IHP used fixed-dose oxaliplatin with escalating 5-FU doses. Toxicity (CTCAE v 4.0) and response (RECIST), progression-free survival, and overall survival (OS) were assessed. Systemic and IHP plasma PK of 5-FU, anabolites, and platinum were determined. RESULTS: All 12 patients had received ≥ 1 line of pre-IHP chemotherapy. There were 4 grade 3 serious adverse events (33.3 %) and 1 grade 4 event (8.3 %). Also, 2 dose-limiting toxicities occurred at DL2 at 300 mg/m(2), resulting in expansion of DL1 at 200 mg/m(2) 5-FU, the eventual MTD. At 6-month follow-up, 9 patients (82 %) demonstrated partial response, while 2 (18 %) exhibited stable disease. Also, 64 % of patients demonstrated a >50 % decrease in CEA. The 1- and 2-year OS probabilities were 90.9 and 71.6 %, respectively, with median follow-up of 24 months. IHP exposures (AUC0-60 min) were 10.9 ± 4.5 µgPt h/mL, 49.3 ± 30.7 µg h/mL 5-FU (DL1), and 70.5 ± 35.5 µg h/mL 5-FU (DL2). Systemic exposure (AUC0-inf) relative to IHP exposure was negligible for both platinum (1.1 ± 1.5 %) and 5-FU (0.09 ± 0.10 %). CONCLUSIONS: The MTD for IHP was 200 mg/m(2) 5-FU with 40 mg/m(2) oxaliplatin. Systemic exposure to the agents was minimal during IHP. The response and survival observed warrants assessment in a larger phase II trial.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Chemotherapy, Cancer, Regional Perfusion , Colorectal Neoplasms/pathology , Liver Neoplasms/drug therapy , Maximum Tolerated Dose , Adult , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Area Under Curve , Carcinoembryonic Antigen/blood , Chemotherapy, Cancer, Regional Perfusion/adverse effects , Colorectal Neoplasms/blood , Disease-Free Survival , Female , Fluorouracil/administration & dosage , Fluorouracil/pharmacokinetics , Humans , Kaplan-Meier Estimate , Liver Neoplasms/blood , Liver Neoplasms/secondary , Male , Middle Aged , Organoplatinum Compounds/administration & dosage , Organoplatinum Compounds/pharmacokinetics , Oxaliplatin
9.
Adv Surg ; 45: 323-40, 2011.
Article in English | MEDLINE | ID: mdl-21954697

ABSTRACT

Robotic-assisted major pancreatic resections allow recreation of time-tested open surgical procedures on a minimally invasive platform. Early outcomes from robotic-assisted major pancreatic resections are comparable with those of laparoscopic and open approaches. Robotic assistance has the potential to bring the well-recognized advantages of minimally invasive surgery to major pancreatic resections. Technological innovations and increased surgeon familiarity with this approach will improve, likely leading to greater adoption and acceptance.


Subject(s)
Pancreatectomy/methods , Pancreatic Diseases/surgery , Pancreaticoduodenectomy/methods , Robotics , Adenocarcinoma/surgery , Dissection , Humans , Laparoscopy , Neuroendocrine Tumors/surgery , Pancreatic Neoplasms/surgery , Portal Vein/surgery , Robotics/methods , Treatment Outcome
10.
Cell Death Differ ; 18(4): 571-80, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21311563

ABSTRACT

Beclin 1, the mammalian orthologue of yeast Atg6, has a central role in autophagy, a process of programmed cell survival, which is increased during periods of cell stress and extinguished during the cell cycle. It interacts with several cofactors (Atg14L, UVRAG, Bif-1, Rubicon, Ambra1, HMGB1, nPIST, VMP1, SLAM, IP(3)R, PINK and survivin) to regulate the lipid kinase Vps-34 protein and promote formation of Beclin 1-Vps34-Vps15 core complexes, thereby inducing autophagy. In contrast, the BH3 domain of Beclin 1 is bound to, and inhibited by Bcl-2 or Bcl-XL. This interaction can be disrupted by phosphorylation of Bcl-2 and Beclin 1, or ubiquitination of Beclin 1. Interestingly, caspase-mediated cleavage of Beclin 1 promotes crosstalk between apoptosis and autophagy. Beclin 1 dysfunction has been implicated in many disorders, including cancer and neurodegeneration. Here, we summarize new findings regarding the organization and function of the Beclin 1 network in cellular homeostasis, focusing on the cross-regulation between apoptosis and autophagy.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Apoptosis , Autophagy , Membrane Proteins/physiology , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Class III Phosphatidylinositol 3-Kinases/metabolism , Humans , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Vacuolar Sorting Protein VPS15/metabolism , bcl-X Protein/metabolism
11.
Oncogene ; 30(16): 1851-4, 2011 Apr 21.
Article in English | MEDLINE | ID: mdl-21151175

ABSTRACT

One hallmark of cancer is intrinsic or acquired resistance to apoptosis. Surprisingly, recent studies demonstrate that CD95/Fas/Apo1 and p53 upregulated mediator of apoptosis/PUMA (potent inducers of the death receptor and the mitochondrial apoptotic pathways, respectively) promote tumorigenesis. These findings provide important insights into the multifaceted roles of apoptosis in tumorigenesis.


Subject(s)
Apoptosis , Cell Transformation, Neoplastic , Humans
12.
Oncogene ; 29(38): 5299-310, 2010 Sep 23.
Article in English | MEDLINE | ID: mdl-20622903

ABSTRACT

The functional relationship and cross-regulation between autophagy and apoptosis is complex. In this study we show that the high-mobility group box 1 protein (HMGB1) is a redox-sensitive regulator of the balance between autophagy and apoptosis. In cancer cells, anticancer agents enhanced autophagy and apoptosis, as well as HMGB1 release. HMGB1 release may be a prosurvival signal for residual cells after various cytotoxic cancer treatments. Diminished HMGB1 by short hairpin RNA transfection or inhibition of HMGB1 release by ethyl pyruvate or other small molecules led predominantly to apoptosis and decreased autophagy in stressed cancer cells. In this setting, reducible HMGB1 binds to the receptor for advanced glycation end products (RAGEs), but not to Toll-like receptor 4, induces Beclin1-dependent autophagy and promotes tumor resistance to alkylators (melphalan), tubulin disrupting agents (paclitaxel), DNA crosslinkers (ultraviolet light) and DNA intercalators (oxaliplatin or adriamycin). On the contrary, oxidized HMGB1 increases the cytotoxicity of these agents and induces apoptosis mediated by the caspase-9/-3 intrinsic pathway. HMGB1 release, as well as its redox state, thus links autophagy and apoptosis, representing a suitable target when coupled with conventional tumor treatments.


Subject(s)
Apoptosis/physiology , Autophagy/physiology , HMGB1 Protein/physiology , Neoplasms/pathology , Antineoplastic Agents/pharmacology , HMGB1 Protein/metabolism , Oxidation-Reduction
13.
Cell Death Differ ; 17(4): 666-76, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19834494

ABSTRACT

Activation of the induced receptor for advanced glycation end products (RAGE) leads to initiation of NF-kappaB and MAP kinase signaling pathways, resulting in propagation and perpetuation of inflammation. RAGE-knockout animals are less susceptible to acute inflammation and carcinogen-induced tumor development. We have reported that most forms of tumor cell death result in release of the RAGE ligand, high-mobility group protein 1 (HMGB1). We now report a novel role for RAGE in the tumor cell response to stress. Targeted knockdown of RAGE in the tumor cell, leads to increased apoptosis, diminished autophagy and decreased tumor cell survival . In contrast, overexpression of RAGE is associated with enhanced autophagy, diminished apoptosis and greater tumor cell viability. RAGE limits apoptosis through a p53-dependent mitochondrial pathway. Moreover, RAGE-sustained autophagy is associated with decreased phosphorylation of mammalian target of rapamycin (mTOR) and increased Beclin-1/VPS34 autophagosome formation. These findings show that the inflammatory receptor, RAGE, has a heretofore unrecognized role in the tumor cell response to stress. Furthermore, these studies establish a direct link between inflammatory mediators in the tumor microenvironment and resistance to programmed cell death. Our data suggest that targeted inhibition of RAGE or its ligands may serve as novel targets to enhance current cancer therapies.


Subject(s)
Apoptosis/physiology , Autophagy/physiology , Carcinoma/metabolism , Pancreatic Neoplasms/metabolism , Receptors, Immunologic/metabolism , Stress, Physiological/physiology , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Carcinoma/physiopathology , Cell Line, Tumor , Cell Survival/physiology , HMGB1 Protein/metabolism , Humans , Inflammation/immunology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Pancreatic Neoplasms/physiopathology , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Receptor for Advanced Glycation End Products , Receptors, Immunologic/genetics , TOR Serine-Threonine Kinases , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
14.
J Gastrointest Surg ; 12(12): 2141-8, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18841423

ABSTRACT

INTRODUCTION: Leiomyosarcoma of the inferior vena cava (IVC) is a rare tumor for which en bloc resection offers the only chance of cure. Due to its rarity, however, optimal strategies for the management of the primary tumor and subsequent recurrences are not well defined. METHODS: We performed a retrospective review of patients who underwent surgical resection of IVC leiomyosarcoma. We evaluated clinical presentations, operative techniques, patterns of recurrence and survival. RESULTS: From 1990 to 2008, nine patients (four females) were identified. Median age was 55 years (40-76). Presentations included abdominal pain (n = 5), back pain (n = 2), leg swelling (n = 4) and abdominal mass (n = 2). Pre-operative imaging studies showed tumor location to be from the right atrium to renal veins (n = 1), retrohepatic (n = 5), and from hepatic veins to the iliac bifurcations (n = 3). En bloc resection included right nephrectomy (n = 5), right adrenalectomy (n = 4), pancreaticoduodenectomy (n = 1), right hepatic trisectionectomy (n = 1) and right hemicolectomy (n = 1). The IVC was ligated in six patients, and a prosthetic graft was used for IVC reconstruction in three patients. Resection margins were negative in seven cases. Median length of stay was 12 days (range, 6-22 days). Major morbidity included renal failure (n = 1) and there was one post-operative mortality. Five patients had leg edema post-operatively, four of whom had IVC ligation. Median survival was 47 months (range, 1-181 months). Four patients had recurrence and the median time to recurrence was 14 months (range, 3-25 months). Two patients underwent successful resection of recurrence. CONCLUSIONS: Curative resection of IVC leiomyosarcoma can lead to long-term survival. However, recurrence is common, and effective adjuvant treatments are needed. In selected cases, aggressive surgical treatment of recurrence should be considered.


Subject(s)
Leiomyosarcoma/surgery , Vascular Neoplasms/surgery , Adult , Aged , Contrast Media , Female , Humans , Leiomyosarcoma/diagnostic imaging , Leiomyosarcoma/pathology , Male , Middle Aged , Retrospective Studies , Survival Rate , Tomography, X-Ray Computed , Treatment Outcome , Vascular Neoplasms/diagnostic imaging , Vascular Neoplasms/pathology , Vena Cava, Inferior/pathology , Vena Cava, Inferior/surgery
16.
Cancer Gene Ther ; 15(2): 115-25, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18084242

ABSTRACT

In this study, we assessed the ability of a highly tumor-selective oncolytic vaccinia virus armed with a yeast cytosine deaminase gene to infect and lyse human and murine ovarian tumors both in vitro and in vivo. The virus vvDD-CD could infect, replicate in and effectively lyse both human and mouse ovarian cancer cells in vitro. In two different treatment schedules involving either murine MOSEC or human A2780 ovarian carcinomatosis models, regional delivery of vvDD-CD selectively targeted tumor cells and ovarian tissue, effectively delaying the development of either tumor or ascites and leading to significant survival advantages. Oncolytic virotherapy using vvDD-CD in combination with the prodrug 5-fluorocytosine conferred an additional long-term survival advantage upon tumor-bearing immunocompetent mice. These findings demonstrate that a tumor-selective oncolytic vaccinia combined with gene-directed enzyme prodrug therapy is a highly effective strategy for treating advanced ovarian cancers in both syngeneic mouse and human xenograft models. Given the biological safety, tumor selectivity and oncolytic potency of this armed oncolytic virus, this dual therapy merits further investigation as a promising new treatment for metastatic ovarian cancer.


Subject(s)
Carcinoma/therapy , Cytosine Deaminase/genetics , Oncolytic Virotherapy , Ovarian Neoplasms/therapy , Saccharomyces cerevisiae/genetics , Vaccinia virus/genetics , Virus Replication , Animals , Antimetabolites/administration & dosage , Antimetabolites/therapeutic use , Carcinoma/drug therapy , Cell Line, Tumor , Combined Modality Therapy , Cytosine Deaminase/administration & dosage , Cytosine Deaminase/therapeutic use , Female , Flucytosine/administration & dosage , Flucytosine/therapeutic use , Humans , Mice , Mice, Inbred C57BL , Mice, Nude , Ovarian Neoplasms/drug therapy , Saccharomyces cerevisiae/enzymology , Vaccinia virus/physiology , Virus Replication/genetics
17.
Gene Ther ; 14(8): 638-47, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17268533

ABSTRACT

To enhance further the safety and efficacy of oncolytic vaccinia virus, we have developed a new virus with targeted deletions of three viral genes encoding thymidine kinase and antiapoptotic/host range proteins SPI-1 and SPI-2 (vSPT). Infection of human and murine tumor cell lines yielded nearly equivalent or a log lower virus recovery in comparison to parental viruses. Viral infection activated multiple caspases in cancer cells but not in normal cells, suggesting infected cells may die via different pathways. In tumor-bearing mice, vSPT recovery from MC38 tumor was slightly reduced in comparison to two parental viruses. However, no virus was recovered from the brains and livers of mice injected with vSPT in contrast to control viruses. vSPT demonstrated significantly lower pathogenicity in nude mice. Systemic delivery of vSPT showed significant tumor inhibition in subcutaneous MC38 tumor, human ovarian A2780 and murine ovarian MOSEC carcinomatosis models; however, the tumor inhibition by vSPT was reduced compared with parental viruses. These results demonstrated that although deletion of these three viral genes further enhanced tumor selectivity, it also weakened the oncolytic potency. This study illustrates the complexity of creating a tumor-selective oncolytic virus by deleting multiple viral genes involved in multiple cellular pathways.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/genetics , Neoplasms/therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Vaccinia virus/genetics , Animals , Cell Line, Tumor , Female , Gene Deletion , Gene Targeting/methods , Genetic Engineering , Humans , Mice , Mice, Inbred C57BL , Mice, Nude , Models, Animal , Neoplasm Transplantation , Protein Kinases/genetics , Recombinant Fusion Proteins/genetics , Safety , Sp2 Transcription Factor/genetics , Thymidine Kinase/genetics , Transplantation, Heterologous , Virus Replication
18.
Cancer Biomark ; 1(6): 259-69, 2005.
Article in English | MEDLINE | ID: mdl-17192050

ABSTRACT

Early detection of pancreatic cancer might improve clinical outcome. Significant alterations in the levels of individual serum cytokines have been reported in pancreatic cancer. We hypothesized that a multicytokine panel could serve as biomarkers for pancreatic cancer. To evaluate the diagnostic utility of such a panel, we have utilized a novel multianalyte LabMAP profiling technology that allows simultaneous measurement of multiple markers. In this study, a panel of 31 serological markers including cytokines, chemokines, growth and angiogenic factors in combination with CA 19-9 was analyzed in sera of pancreatic cancer patients, patients with chronic pancreatitis, and matched control healthy subjects. Statistical analysis identified a multicytokine panel that was able to distinguish pancreatic cancer from healthy controls with a sensitivity of 85.7% and specificity of 92.3%, which was superior to performance of CA 19-9 alone. Importantly, a multicytokine panel allowed the discrimination of pancreatic cancer from chronic pancreatitis with high sensitivity of 98% and specificity of 96.4%. In conclusion, we demonstrated that analysis of multiple serum cytokines using a novel LabMAP technology is a promising approach for development of a diagnostic assay for pancreatic cancer.


Subject(s)
Biomarkers, Tumor/blood , Cytokines/blood , Pancreatic Neoplasms/diagnosis , Protein Array Analysis/methods , CA-19-9 Antigen/blood , Case-Control Studies , Control Groups , Data Interpretation, Statistical , Diagnosis, Differential , Female , Humans , Male , Pancreatic Neoplasms/blood , Pancreatitis, Chronic/blood , Pancreatitis, Chronic/diagnosis , Sensitivity and Specificity
19.
Trends Mol Med ; 7(7): 307-13, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11425639

ABSTRACT

Despite recent advances in the treatment of colorectal cancer, the overall survival rate for those patients with advanced locoregional disease remains less than 50%. Although adjuvant systemic chemotherapy has improved survival of these patients, more effective therapies are needed. Immunotherapy is an approach that could have a particular role in the adjuvant therapy of colorectal cancer. There is now convincing evidence that the immune system can specifically recognize and destroy malignant cells. Although both antibody- and T-cell-mediated anti-tumor responses have been documented, the cellular immune response with its direct cytotoxic mechanisms is felt to be the principal anti-tumor arm of the immune system. Analysis of the T cells that recognize tumors has led to the identification and characterization of many tumor-associated antigens including several colorectal antigens. Current approaches to developing a vaccine for colorectal cancer use our expanded understanding of these tumor-associated antigens and the conditions that allow development of an effective cellular immune response to them.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines , Colorectal Neoplasms/immunology , Colorectal Neoplasms/therapy , BCG Vaccine , Dendritic Cells/immunology , Humans , Immunotherapy/methods , Vaccines, Synthetic
20.
J Comput Assist Tomogr ; 23(5): 664-9, 1999.
Article in English | MEDLINE | ID: mdl-10524842

ABSTRACT

PURPOSE: Isolated hepatic perfusion (IHP) is a new treatment for patients with isolated unresectable liver metastases, which can result in a partial or complete response in approximately 75% of patients. Preoperative knowledge of hepatic arterial anatomy is important to adequately perfuse the liver. Digital subtraction angiography (DSA) is currently used to identify the hepatic arterial anatomy. The purpose of this study was to determine if MR angiography (MRA) could replace DSA prior to IHP. METHOD: Twenty-seven patients scheduled to undergo IHP underwent MRA with a contrast-enhanced 3D time-of-flight gradient echo sequence. Both maximal intensity projections (MIPs) and source coronal images were used to evaluate the images. The results of the MRA were interpreted by two readers who were blinded to the surgical results. The first 17 patients also underwent DSA, and a separate comparison was made with those results. Anatomy was characterized as either normal hepatic arteries (NHAs), normal vasculature with an accessory left hepatic artery (aLHA), or a replaced right hepatic artery (rRHA). RESULTS: MRA correctly detected all 22 patients with NHAs but also identified 6 aLHAs, of which only 2 were confirmed surgically. MRA correctly detected all five rRHAs. MIP images alone accurately depicted the hepatic arterial anatomy in only 9 of 27 (33%), usually because significant vessels were not visualized or their origin could not be determined. Source coronal images were required to accurately determine the anatomy in all patients. Among the 17 patients who underwent DSA, MRA detected 14 of 14 with NHA and 3 of 3 with rRHA. Six aLHAs were identified by MRA and five were confirmed by DSA. CONCLUSION: Enhanced 3D MRA is an accurate method of depicting the hepatic arterial supply. In comparison to surgery, MRA overestimates the number of aLHAs, but this may be because these small vessels are not detected at surgery. Based on the results of this study, DSA has been replaced by MRA in the planning of IHP at our institution. A better display of MRA images is needed as MIP images were usually insensitive for the small caliber arteries supplying the liver.


Subject(s)
Chemotherapy, Cancer, Regional Perfusion/methods , Contrast Media , Gadolinium , Heterocyclic Compounds , Liver Neoplasms/diagnosis , Liver Neoplasms/secondary , Magnetic Resonance Angiography/methods , Organometallic Compounds , Angiography, Digital Subtraction , Celiac Artery/diagnostic imaging , Celiac Artery/pathology , Hepatic Artery/diagnostic imaging , Hepatic Artery/pathology , Humans , Liver Neoplasms/blood supply , Liver Neoplasms/drug therapy , Liver Neoplasms/surgery , Magnetic Resonance Angiography/instrumentation , Preoperative Care/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...