Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
J Vasc Surg ; 78(4): 1030-1040.e2, 2023 10.
Article in English | MEDLINE | ID: mdl-37318431

ABSTRACT

OBJECTIVE: Prior research has shown that socioeconomic status (SES) is associated with higher rates of diabetes, peripheral vascular disease, and amputation. We sought to determine whether SES or insurance type increases the risk of mortality, major adverse limb events (MALE), or hospital length of stay (LOS) after open lower extremity revascularization. METHODS: We conducted a retrospective analysis of patients who underwent open lower extremity revascularization at a single tertiary care center from January 2011 to March 2017 (n = 542). SES was determined using state Area Deprivation Index (ADI), a validated metric determined by income, education, employment, and housing quality by census block group. Patients undergoing amputation in this same time period (n = 243) were included to compare rates of revascularization to amputation by ADI and insurance status. For patients undergoing revascularization or amputation procedures on both limbs, each limb was treated individually for this analysis. We performed a multivariate analysis of the association between ADI and insurance type with mortality, MALE, and LOS using Cox proportional hazard models, including confounding variables such as age, gender, smoking status, body mass index, hyperlipidemia, hypertension, and diabetes. The cohort with an ADI quintile of 1, meaning least deprived, and the Medicare cohort were used for reference. P values of <.05 were considered statistically significant. RESULTS: We included 246 patients undergoing open lower extremity revascularization and 168 patients undergoing amputation. Controlling for age, gender, smoking status, body mass index, hyperlipidemia, hypertension, and diabetes, ADI was not an independent predictor of mortality (P = .838), MALE (P = .094), or hospital LOS (P = .912). Controlling for the same confounders, uninsured status was independently predictive of mortality (P = .033), but not MALE (P = .088) or hospital LOS (P = .125). There was no difference in the distribution of revascularizations or amputations by ADI (P = .628), but there was higher proportion of uninsured patients undergoing amputation compared with revascularization (P < .001). CONCLUSIONS: This study suggests that ADI is not associated with an increased risk of mortality or MALE in patients undergoing open lower extremity revascularization, but that uninsured patients are at higher risk of mortality after revascularization. These findings indicate that individuals undergoing open lower extremity revascularization at this single tertiary care teaching hospital received similar care, regardless of their ADI. Further study is warranted to understand the specific barriers that uninsured patients face.


Subject(s)
Diabetes Mellitus , Endovascular Procedures , Hypertension , Peripheral Arterial Disease , Humans , Aged , United States/epidemiology , Risk Factors , Endovascular Procedures/adverse effects , Limb Salvage/methods , Retrospective Studies , Treatment Outcome , Peripheral Arterial Disease/diagnosis , Peripheral Arterial Disease/surgery , Medicare , Lower Extremity/blood supply , Hypertension/etiology , Ischemia
2.
Cancer Gene Ther ; 22(12): 591-6, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26584530

ABSTRACT

Although early stage cholangiocarcinoma (CC) can be cured by surgical extirpation, the options for treatment of advanced stage CC are very few and suboptimal. Oncolytic virotherapy using replication-competent vaccinia virus (VACV) is a promising new strategy to treat human cancers. The ability of oncolytic VACV GLV-1h68 to infect, replicate in, and lyse three human CC cell lines was assayed in vitro and in subcutaneous flank xenografts in athymic nude mice. In this study, we have demonstrated that GLV-1h68 effectively infects and lyses three CC cell lines (KMC-1, KMBC, and KMCH-1) in vitro. Expression of the viral marker gene ruc-gfp facilitated real-time monitoring of infection and replication. Furthermore in athymic nude mice, a single dose of GLV-1h68 significantly suppressed tumor growth. The treatment was well tolerated in all animals. Recombinant VACV GLV-1h68 has significant oncolytic ability against CC both in vitro and in vivo. GLV-1h68 has the potential to be used clinically as a therapeutic agent against CC.


Subject(s)
Bile Duct Neoplasms/therapy , Cholangiocarcinoma/therapy , Genetic Vectors , Oncolytic Virotherapy , Vaccinia virus/genetics , Animals , Bile Duct Neoplasms/virology , Cell Line, Tumor , Chlorocebus aethiops , Cholangiocarcinoma/virology , Humans , Mice , Mice, Nude , Xenograft Model Antitumor Assays
3.
Surgery ; 158(2): 331-8, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26049609

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) commonly presents at a late stage when surgery is no longer a curative option. As such, novel therapies for advanced HCC are needed. Oncolytic viruses are a viable option for cancer therapy owing to their ability to specifically infect, replicate within, and kill cancer cells. In this study, we have investigated the ability of GLV-2b372, a novel light-emitting recombinant vaccinia virus derived from a wild-type Lister strain, to kill HCC. METHODS: Four human HCC cell lines were assayed in vitro for infectivity and cytotoxicity. Viral replication was quantified via standard viral plaque assays. Flank HCC xenografts generated in athymic nude mice were treated with intratumoral GLV-2b372 to assess for tumor growth inhibition and viral biodistribution. RESULTS: Infectivity occurred in a time- and concentration-dependent manner with 70% cell death in all cell lines by day 5. All cell lines supported efficient viral replication. At 25 days after infection, flank tumor volumes decreased by 50% whereas controls increased by 400%. Tumor tissue demonstrated substantial GLV-2b372 infection at 24 hours, 48 hours, and 2 weeks. CONCLUSION: We demonstrate that GLV-2b372 efficiently kills human HCC in vitro and in vivo and is a viable treatment option for patients with HCC.


Subject(s)
Carcinoma, Hepatocellular/therapy , Genetic Therapy , Liver Neoplasms/therapy , Oncolytic Virotherapy , Oncolytic Viruses/genetics , Vaccinia virus/genetics , Animals , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Treatment Outcome , Xenograft Model Antitumor Assays
4.
Surgery ; 157(2): 331-7, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25616946

ABSTRACT

BACKGROUND: Peritoneal carcinomatosis (PC) is a terminal progression of colorectal cancer (CRC). Poor response to cytoreductive operation and chemotherapy coupled with the inability to reliably track disease progression by the use of established diagnostic methods, make this a deadly disease. We examined the effectiveness of the oncolytic vaccinia virus GLV-1h153 as a therapeutic and diagnostic vehicle. We believe that viral expression of the human sodium iodide transporter (hNIS) provides both real-time monitoring of viral therapy and effective treatment of colorectal peritoneal carcinomatosis (CRPC). METHODS: Infectivity and cytotoxic effect of GLV-1h153 on CRC cell lines was assayed in vitro. Viral replication was examined by standard viral plaque assays. Orthotopic CRPC xenografts were generated in athymic nude mice and subsequently administered GLV-1h153 intraperitoneally. A decrease in tumor burden was assessed by mass. Orthotopic tumors were visualized by single-photon emission computed tomography/computed tomography after Iodine ((131)I) administration and by fluorescence optical imaging. RESULTS: GLV-1h153 infected and killed CRC cells in a time- and concentration-dependent manner. Viral replication demonstrated greater than a 2.35 log increase in titer over 4 days. Intraperitoneal treatment of orthotopic CRPC xenografts resulted in a substantial decrease in tumor burden. Infection of orthotopic xenografts was therapeutic and facilitated monitoring by (131)I-single-photon emission computed tomography/computed tomography via expression of hNIS in infected tissue. CONCLUSION: GLV-1h153 kills CRC in vitro effectively and decreases tumor burden in vivo. We demonstrate that GLV-1h153 can be used as an agent to provide accurate delineation of tumor burden in vivo. These findings indicate that GLV-1h153 has potential for use as a therapeutic and diagnostic agent in the treatment of CRPC.


Subject(s)
Colorectal Neoplasms/therapy , Oncolytic Virotherapy/methods , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/therapy , Vaccinia virus/genetics , Animals , Cell Line, Tumor , Colorectal Neoplasms/pathology , Disease Progression , Gene Expression , Green Fluorescent Proteins/genetics , Humans , Iodine Radioisotopes , Mice , Mice, Nude , Neoplasm Seeding , Oncolytic Viruses/genetics , Peritoneal Neoplasms/pathology , Radiopharmaceuticals , Recombinant Proteins/genetics , Symporters/genetics , Tomography, Emission-Computed, Single-Photon , Xenograft Model Antitumor Assays
5.
Front Physiol ; 5: 400, 2014.
Article in English | MEDLINE | ID: mdl-25400582

ABSTRACT

Malignant pleural mesothelioma is a particularly aggressive and locally invasive malignancy with a poor prognosis despite advances in understanding of cancer cell biology and development of new therapies. At the cellular level, cultured mesothelioma cells present a mesenchymal appearance and a strong capacity for local cellular invasion. One important but underexplored area of mesothelioma cell biology is intercellular communication. Our group has previously characterized in multiple histological subtypes of mesothelioma a unique cellular protrusion known as tunneling nanotubes (TnTs). TnTs are long, actin filament-based, narrow cytoplasmic extensions that are non-adherent when cultured in vitro and are capable of shuttling cellular cargo between connected cells. Our prior work confirmed the presence of nanotube structures in tumors resected from patients with human mesothelioma. In our current study, we quantified the number of TnTs/cell among various mesothelioma subtypes and normal mesothelial cells using confocal microscopic techniques. We also examined changes in TnT length over time in comparison to cell proliferation. We further examined potential approaches to the in vivo study of TnTs in animal models of cancer. We have developed novel approaches to study TnTs in aggressive solid tumor malignancies and define fundamental characteristics of TnTs in malignant mesothelioma. There is mounting evidence that TnTs play an important role in intercellular communication in mesothelioma and thus merit further investigation of their role in vivo.

6.
Surgery ; 156(2): 263-9, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24957667

ABSTRACT

BACKGROUND: Sorafenib is the standard systemic therapy for unresectable or recurrent hepatocellular carcinoma (HCC) but adds minimal increase in survival. Therefore, there is a great need to develop novel therapies for advanced or recurrent HCC. One emerging field of cancer treatment involves oncolytic viruses that specifically infect, replicate within, and kill cancer cells. In this study, we examined the ability of GLV-1h68, a recombinant vaccinia virus derived from the vaccine strain that was used to eradicate smallpox, to kill sorafenib-resistant (SR) HCC cell lines. METHODS: Four SR HCC cell lines were generated by repeated passage in the presence of sorafenib. Median inhibitory concentration was determined for all cell lines. The infectivity, viral replication, and cytotoxicity of GLV-1h68 were assayed for both parental and SR HCC cells. RESULTS: Infectivity increased in a time and concentration-dependent manner in all cell lines. All cell lines supported efficient replication of virus. No difference between the rates of cell death between the parental and SR cell lines was observed. CONCLUSION: Our results demonstrate that the oncolytic vaccinia virus GLV-1h68 kills both parental and SR HCC cell lines efficiently. This study indicates that patients who have failed treatment with sorafenib remain viable candidates for oncolytic therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/therapy , Liver Neoplasms/therapy , Niacinamide/analogs & derivatives , Oncolytic Virotherapy/methods , Phenylurea Compounds/pharmacology , Vaccinia virus/genetics , Vaccinia virus/immunology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Neoplasm , Genetic Engineering , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Niacinamide/pharmacology , Sorafenib , Vaccinia virus/physiology , Viral Plaque Assay , Virus Replication
7.
Oncolytic Virother ; 3: 35-46, 2014.
Article in English | MEDLINE | ID: mdl-27512661

ABSTRACT

The development of targeted agents and chemotherapies for pancreatic cancer has only modestly affected clinical outcome and not changed 5-year survival. Fortunately the genetic and molecular mechanisms underlying pancreatic cancer are being rapidly uncovered and are providing opportunities for novel targeted therapies. Oncolytic viral therapy is one of the most promising targeted agents for pancreatic cancer. This review will look at the current state of the development of these self-replicating nanoparticles in the treatment of pancreatic cancer.

8.
Surgery ; 154(3): 486-95, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23890748

ABSTRACT

BACKGROUND: The purpose of this work was to examine the ability of an oncolytic vaccinia virus expressing the human sodium iodine transporter (hNIS) to provide real time monitoring of viral therapy and effective treatment of malignant pleural mesothelioma (MPM). METHODS: Infectivity and cytotoxic effects of GLV-1h153 on mesothelioma cell lines of all histologic subtypes were assayed in vitro. Viral replication was examined by standard viral plaque assay. Orthotopic MPM xenografts were generated in athymic nude mice, treated with intrapleural GLV-1h153, and assessed for effect on tumor burden and survival. Orthotopic tumors were also imaged on single photon emission computed tomography (SPECT)/computed tomography (CT) after (131)I administration. RESULTS: GLV-1h153-infected and killed all cell lines in a time- and concentration-dependent manner. Viral replication demonstrated a >2.5-log increase in titer over 4 days. Intrapleural treatment of orthotopic MPM xenografts resulted in a significant decrease in tumor burden 1 week after treatment and an improvement in survival. Infection of orthotopic xenografts was both therapeutic and facilitated monitoring by (131)I-SPECT/CT via expression of hNIS in infected tissue. CONCLUSION: Our results suggest that GLV-1h153 may be a promising therapeutic agent for MPM and warrants further investigation.


Subject(s)
Mesothelioma/therapy , Multimodal Imaging/methods , Oncolytic Viruses/genetics , Pleural Neoplasms/therapy , Positron-Emission Tomography , Symporters/genetics , Tomography, X-Ray Computed , Vaccinia virus/genetics , Animals , Cell Line, Tumor , Female , Humans , Luminescent Measurements , Mesothelioma/diagnostic imaging , Mesothelioma/mortality , Mice , Pleural Neoplasms/diagnostic imaging , Pleural Neoplasms/mortality , Tumor Burden , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...