Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Obes Surg ; 34(5): 1552-1560, 2024 May.
Article in English | MEDLINE | ID: mdl-38564172

ABSTRACT

OBJECTIVE: To investigate usage and utility of routine upper gastrointestinal (UGI) series in the immediate post-operative period to evaluate for leak and other complications. METHODS: Single institution IRB-approved retrospective review of patients who underwent bariatric procedure between 01/08 and 12/12 with at least 6-month follow-up. RESULTS: Out of 135 patients (23%) who underwent routine UGI imaging, 32% of patients were post-gastric bypass (127) versus 4% of sleeve gastrectomy (8). In patients post-gastric bypass, 22 were found with delayed contrast passage, 3 possible obstruction, 4 possible leak, and only 1 definite leak. In patients post-sleeve gastrectomy, 2 had delayed passage of contrast without evidence of a leak. No leak was identified in 443 patients (77%) who did not undergo imaging. The sensitivity and specificity of UGI series for the detection of leak in gastric bypass patients were 100% and 97%, respectively, and the positive and negative predictive values were 20% and 100%, respectively. On univariate and multivariate analysis, sleeve gastrectomy patients (OR 0.4 sleeve vs bypass; P < 0.01) and male patients (OR 0.4 M vs F; P 0.02) were less likely to undergo routine UGI series (OR 0.4 M vs F; P 0.02). CONCLUSION: Routine UGI series may be of limited value for the detection of anastomotic leaks after gastric bypass or sleeve gastrectomy and patients should undergo routine imaging based on clinical parameters. Gastric bypass procedure and female gender were factors increasing the likelihood of routine post-operative UGI. Further larger scale analysis of this important topic is warranted.


Subject(s)
Bariatric Surgery , Gastric Bypass , Laparoscopy , Obesity, Morbid , Humans , Male , Female , Obesity, Morbid/surgery , Postoperative Complications/diagnosis , Postoperative Complications/surgery , Contrast Media , Laparoscopy/methods , Gastric Bypass/adverse effects , Gastric Bypass/methods , Anastomotic Leak/diagnostic imaging , Anastomotic Leak/surgery , Retrospective Studies , Gastrectomy/adverse effects , Gastrectomy/methods
2.
NPJ Breast Cancer ; 8(1): 101, 2022 Sep 02.
Article in English | MEDLINE | ID: mdl-36056005

ABSTRACT

Metastatic dissemination in breast cancer is regulated by specialized intravasation sites called "tumor microenvironment of metastasis" (TMEM) doorways, composed of a tumor cell expressing the actin-regulatory protein Mena, a perivascular macrophage, and an endothelial cell, all in stable physical contact. High TMEM doorway number is associated with an increased risk of distant metastasis in human breast cancer and mouse models of breast carcinoma. Here, we developed a novel magnetic resonance imaging (MRI) methodology, called TMEM Activity-MRI, to detect TMEM-associated vascular openings that serve as the portal of entry for cancer cell intravasation and metastatic dissemination. We demonstrate that TMEM Activity-MRI correlates with primary tumor TMEM doorway counts in both breast cancer patients and mouse models, including MMTV-PyMT and patient-derived xenograft models. In addition, TMEM Activity-MRI is reduced in mouse models upon treatment with rebastinib, a specific and potent TMEM doorway inhibitor. TMEM Activity-MRI is an assay that specifically measures TMEM-associated vascular opening (TAVO) events in the tumor microenvironment, and as such, can be utilized in mechanistic studies investigating molecular pathways of cancer cell dissemination and metastasis. Finally, we demonstrate that TMEM Activity-MRI increases upon treatment with paclitaxel in mouse models, consistent with prior observations that chemotherapy enhances TMEM doorway assembly and activity in human breast cancer. Our findings suggest that TMEM Activity-MRI is a promising precision medicine tool for localized breast cancer that could be used as a non-invasive test to determine metastatic risk and serve as an intermediate pharmacodynamic biomarker to monitor therapeutic response to agents that block TMEM doorway-mediated dissemination.

3.
Cureus ; 13(6): e15502, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34268033

ABSTRACT

Paroxysmal supraventricular tachycardia (PSVT) is a common tachyarrhythmia, and an electrocardiogram is the best tool for making a diagnosis. If Valsalva maneuvers and carotid sinus massage do not give positive results, then the next choice is either adenosine or calcium channel blockers. At this time, adenosine is the drug of choice of treatment. Verapamil and diltiazem are the most commonly used calcium channel blockers (CCBs). This review aimed to compare the efficacy of both drugs in the treatment of PSVT. We utilized the databases PubMed Central and Medline by using keywords: "calcium channel blockers OR adenosine AND supraventricular tachycardia." In the end, we finalized 32 studies, including observational studies, literature reviews, systematic reviews/metanalysis, and randomized control trials. We included articles only in the English language and related to humans. Two authors completed the quality assessment and evaluation of bias according to specific guidelines. Only high-quality studies were included in this systematic review based on the cut-off score of seven or above. Calcium channel blockers have a longer half-life than adenosine and were previously used as the drug of choice in the treatment of PSVT. Calcium channel blockers are safe if given slowly; however, adenosine is safer and useful when an electrocardiogram is uncertain. We compared both drugs in certain aspects and found equal efficacy. Though safer, adenosine was found to have a higher cost and a higher probability of re-initiation arrhythmia compared to calcium channel blockers.

4.
Cureus ; 13(2): e13552, 2021 Feb 25.
Article in English | MEDLINE | ID: mdl-33815972

ABSTRACT

Immunotherapy is the upcoming trend in cancer treatment. Traditional cancer treatment methods include surgical resection, radiotherapy, chemotherapy, small molecule targeted drugs, monoclonal antibodies, and hematopoietic stem cell transplantation (HSCT). Surgical resection is useful for early-stage patients but not for metastatic cancer cells; radiotherapy and chemotherapy are more common but produce substantial damage to normal tissues and have poor selectivity. Targeted drugs, including monoclonal antibodies, have better comprehensive efficacy but can also encourage gene mutation of tumor cells and drug tolerance. HSCT is effective, but choosing a donor is often difficult, and the graft is also prone to rejection. Thus, chimeric antigen receptor (CAR)-T cell therapy, a form of cellular/adoptive immunotherapy, is at the forefront of cancer therapy treatments due to its sustained remission, fewer side effects, and a better quality of life. CAR-T cell therapy involves genetically modifying the T cells and multiplying their numbers to kill cancer cells. This review article gives an insight into how the CAR-T cells have evolved from simple T cells with modest immune function to genetically engineered robust counterparts that brought great hope in the treatment of hematological malignancies. Much research has been undertaken during the past decade to design and deliver CAR-T cells. This has led to successful outcomes in leukemias, lymphomas, and multiple myeloma, paving the way for expanding CAR therapy. Despite tremendous progress, CAR-T cell therapies are faced with many challenges. Areas for improvement include limited T cell persistence, tumor escape, immunosuppressive components in the tumor microenvironment, cancer relapse rate, manufacturing time, and production cost. In this manuscript, we summarize the innovations in the design and delivery of CAR technologies, their applications in hematological malignancies, limitations to its widespread application, latest developments, and the future scope of research to counter the challenges and improve its effectiveness and persistence.

5.
Cureus ; 13(3): e14010, 2021 Mar 20.
Article in English | MEDLINE | ID: mdl-33884251

ABSTRACT

Inflammatory bowel disease (IBD) is a chronic condition of the bowel that can be further categorized into ulcerative colitis and Crohn's disease. Rarely, this condition can be associated with pericarditis, which can be an extraintestinal manifestation of the disease or drug-induced. This review aims to determine the pathogenesis and management of pericarditis in IBD. In this review, the goal is to elucidate the pathogenesis of pericarditis in IBD and determine if pericarditis is an extraintestinal manifestation of IBD or a complication of current drug therapy used to manage IBD. Additionally, this review intends to explain the first-line management of pericarditis in IBD and explore the role of biologicals in attenuating pericarditis. An electronic search was conducted to identify relevant reports of pericarditis in IBD, and a quality assessment was conducted to identify high-quality articles according to the inclusion criteria. Full-text articles from inception to November 2020 were included, while non-English articles, gray literature, and animal studies were excluded. The majority of studies suggest that pericarditis arises as a complication of drug therapy by 5-aminosalicylic acid derivatives such as sulfasalazine, mesalamine, and balsalazide, and it occurs due to IgE-mediated allergic reactions, direct cardiac toxicity, cell-mediated hypersensitivity reactions, and humoral antibody response to therapy. Drug cessation or the initiation of a corticosteroid regimen seems to be the most effective means of managing pericarditis in IBD due to drug therapy or an extraintestinal manifestation.

6.
Front Oncol ; 10: 595820, 2020.
Article in English | MEDLINE | ID: mdl-33614481

ABSTRACT

PURPOSE: Breast MRI background parenchymal enhancement (BPE) can potentially serve as a prognostic marker, by possible correlation with molecular subtype. Oncotype Dx, a gene assay, is a prognostic and predictive surrogate for tumor aggressiveness and treatment response. The purpose of this study was to investigate the association between contralateral non-tumor breast magnetic resonance imaging (MRI) background parenchymal enhancement and tumor oncotype score. METHODS: In this retrospective study, patients with ER+ and HER2- early stage invasive ductal carcinoma who underwent preoperative breast MRI, oncotype risk scoring, and breast conservation surgery from 2008-2010 were identified. After registration, BPE from the pre and three post-contrast phases was automatically extracted using a k-means clustering algorithm. Four metrics were calculated: initial enhancement (IE) relative to the pre-contrast signal, late enhancement, overall enhancement (OE), and area under the enhancement curve (AUC). Histogram analysis was performed to determine first order metrics which were compared to oncotype risk score groups using Mann-Whitney tests and Spearman rank correlation analysis. RESULTS: This study included 80 women (mean age = 51.1 ± 10.3 years); 46 women were categorized as low risk (≤17) and 34 women were categorized as intermediate/high risk (≥18) according to Oncotype Dx. For the mean of the top 10% pixels, significant differences were noted for IE (p = 0.032), OE (p = 0.049), and AUC (p = 0.044). Using the risk score as a continuous variable, correlation analysis revealed a weak but significant correlation with the mean of the top 10% pixels for IE (r = 0.26, p = 0.02), OE (r = 0.25, p = 0.02), and AUC (r = 0.27, p = 0.02). CONCLUSION: BPE metrics of enhancement in the non-tumor breast are associated with tumor Oncotype Dx recurrence score, suggesting that the breast microenvironment may relate to likelihood of recurrence and magnitude of chemotherapy benefit.

7.
Surg Obes Relat Dis ; 13(8): 1327-1336, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28606718

ABSTRACT

PURPOSE: A lack of well-defined postoperative imaging guidelines for bariatric patients may lead to false-positive findings, radiation exposure, additional cost, and patient anxiety. We investigated our institutional usage and utility of nonroutine postoperative abdominal imaging. METHODS AND MATERIALS: Laparoscopic gastric bypass and sleeve gastrectomy patients over a 5-year period were retrospectively identified. All bariatric-related nonroutine initial and all subsequent prompted abdominal and pelvic imaging was included. RESULTS: A total of 578 patients were included (399 gastric bypass, 179 sleeve gastrectomy); 907 nonroutine studies in 69% of patients were performed, and 36% patients underwent computed tomography (CT). Only 20.3% of findings were symptom-related, 26% had benign incidental findings, and 50% were negative. Incidental findings prompted 71 additional studies. Bypass procedure (sleeve versus bypass, odds ratio [OR] .3), older age (median 43 versus 48 years), and lower initial body mass index (BMI) (median 43 versus 45) increased the likelihood of imaging. History of prior abdominal surgery and dyspepsia increased the probability of undergoing CT by an odds ratio of 1.8 and 2.0, respectively (P<.05). History of ulcer (OR .6) or reflux on routine upper gastrointestinal imaging (OR .3) decreased probability (P<.05). Patients who underwent CT were more likely to undergo other abdominal imaging (3 versus 1 study per patient, P<.01). CONCLUSIONS: Postoperative abdominal imaging in the bariatric population is common, with almost 70% of patients undergoing imaging and 70% of findings not related to patient symptoms. Bypass procedure, older age, and lower initial BMI were associated with a higher likelihood of patients undergoing imaging. Heightened understanding of this important subject is necessary to help streamline cost-effective imaging protocols for these patients.


Subject(s)
Diagnostic Imaging/statistics & numerical data , Gastrectomy/statistics & numerical data , Gastric Bypass/statistics & numerical data , Laparoscopy/statistics & numerical data , Abdominal Cavity , Adult , Body Mass Index , Female , Fluoroscopy/statistics & numerical data , Humans , Incidental Findings , Length of Stay/statistics & numerical data , Magnetic Resonance Imaging/statistics & numerical data , Male , Middle Aged , Postoperative Complications/diagnosis , Retrospective Studies , Tomography, X-Ray Computed/statistics & numerical data
8.
Front Oncol ; 7: 96, 2017.
Article in English | MEDLINE | ID: mdl-28589082

ABSTRACT

Despite advances in technology, the formidable challenge of treating cancer, especially if advanced, still remains with no significant improvement in survival rates, even with the most common forms of cancer. Oncolytic viral therapies have shown great promise for the treatment of various cancers, with the possible advantages of stronger treatment efficacy compared to conventional therapy due to higher tumor selectivity, and less toxicity. They are able to preferentially and selectively propagate in cancer cells, consequently destroying tumor tissue mainly via cell lysis, while leaving non-cancerous tissues unharmed. Several wild-type and genetically engineered vaccinia virus (VACV) strains have been tested in both preclinical and clinical trials with promising results. Greater understanding and advancements in molecular biology have enabled the generation of genetically engineered oncolytic viruses for safer and more efficacious treatment, including arming VACVs with cytokines and immunostimulatory molecules, anti-angiogenic agents, and enzyme prodrug therapy, in addition to combining VACVs with conventional external and systemic radiotherapy, chemotherapy, immunotherapy, and other virus strains. Furthermore, novel oncolytic vaccinia virus strains have been generated that express reporter genes for the tracking and imaging of viral therapy and monitoring of therapeutic response. Further study is needed to unlock VACVs' full potential as part of the future of cancer therapy.

9.
J Radiol Case Rep ; 11(9): 28-34, 2017 Sep.
Article in English | MEDLINE | ID: mdl-29299107

ABSTRACT

We report a case of absent pituitary infundibulum and ectopic neurohypophysis in a 4-year-old patient presenting clinically with hypopituitarism as well as heterotaxy syndrome complicated by global developmental delay and growth retardation. The clinical and laboratory workup of our patient suggested underlying hypopituitarism related to either congenital or acquired pathology, necessitating MRI to distinguish between them. We explain the various structural causes of hypopituitarism and detail how to predict the MRI findings and treatment, based on a fundamental understanding of the anatomy and pathophysiology of the hypothalamic pituitary axis and distinguishing anterior versus posterior pituitary hormone derangements. We also discuss two important theories widely acknowledged in the literature to explain congenital hypopituitarism: 1. Head trauma typically during birth resulting in a stretch injury to the infundibulum. 2. Congenital fetal maldevelopment of midline structures.


Subject(s)
Choristoma/diagnostic imaging , Empty Sella Syndrome/diagnostic imaging , Heterotaxy Syndrome/complications , Hypopituitarism/diagnostic imaging , Median Eminence/diagnostic imaging , Pituitary Gland/diagnostic imaging , Child, Preschool , Empty Sella Syndrome/congenital , Humans , Hypopituitarism/complications , Hypopituitarism/congenital , Magnetic Resonance Imaging , Male , Pituitary Gland/abnormalities , Pituitary Gland, Posterior/abnormalities , Pituitary Gland, Posterior/diagnostic imaging
10.
Mol Ther Oncolytics ; 3: 16013, 2016.
Article in English | MEDLINE | ID: mdl-27347556

ABSTRACT

Stem-like tumor-initiating cells (TICs) are implicated in cancer progression and recurrence, and can be identified by sphere-formation and tumorigenicity assays. Oncolytic viruses infect, replicate in, and kill a variety of cancer cells. In this study, we seek proof of principle that TICs are susceptible to viral infection. HCT8 human colon cancer cells were subjected to serum-free culture to generate TIC tumorspheres. Parent cells and TICs were infected with HSV-1 subtype NV1066. Cytotoxicity, viral replication, and Akt1 expression were assessed. TIC tumorigenicity was confirmed and NV1066 efficacy was assessed in vivo. NV1066 infection was highly cytotoxic to both parent HCT8 cells and TICs. In both populations, cell-kill of >80% was achieved within 3 days of infection at a multiplicity of infection (MOI) of 1.0. However, the parent cells required 2-log greater viral replication to achieve the same cytotoxicity. TICs overexpressed Akt1 in vitro and formed flank tumors from as little as 100 cells, growing earlier, faster, larger, and with greater histologic atypia than tumors from parent cells. Treatment of TIC-induced tumors with NV1066 yielded tumor regression and slowed tumor growth. We conclude that colon TICs are selected for by serum-free culture, overexpress Akt1, and are susceptible to oncolytic viral infection.

11.
Mol Ther Oncolytics ; 3: 16008, 2016.
Article in English | MEDLINE | ID: mdl-27119120

ABSTRACT

BACKGROUND: Pancreatic cancer is a fatal disease associated with resistance to conventional therapies. This study aimed to determine changes in gene expression patterns associated with infection and susceptibility of pancreatic cancer cells to an oncolyticvaccinia virus, GLV-1h153, carrying the human sodium iodide symporter for deep tissue imaging of virotherapy. METHODS: Replication and susceptibility of pancreatic adenocarcinoma PANC-1 cells to GLV-1h153 was confirmed with replication and cytotoxicity assays. PANC-1 cells were then infected with GLV-1h153 and near-synchronous infection confirmed via flow cytometry of viral-induced green fluorescent protein (GFP) expression. Six and 24 hours after infection, three samples of each time point were harvested, and gene expression patterns assessed using HG-U133A cDNA microarray chips as compared to uninfected control. Differentially expressed genes were identified using Bioconductor LIMMA statistical analysis package. A fold change of 2.0 or above was used as a cutoff, with a P value of 0.01. The gene list was then analyzed using Ingenuity Pathways Analysis software. RESULTS: Differential gene analysis revealed a total of 12,412 up- and 11,065 downregulated genes at 6 and 24 hours postinfection with GLV-1h153 as compared to control. At 6 hours postinfection. A total of 139 genes were either up or downregulated >twofold (false discovery rate < 0.05), of which 124 were mapped by Ingenuity Pathway Analysis (IPA). By 24 hours postinfection, a total of 5,698 genes were identified and 5,563 mapped by IPA. Microarray revealed gene expression changes, with gene networks demonstrating downregulation of processes such as cell death, cell cycle, and DNA repair, and upregulation of infection mechanisms (P < 0.01). Six hours after infection, gene changes involved pathways such as HMGB-1, interleukin (IL)-2, IL-6, IL-8, janus kinase/signal tranducer and activator of transcription (JAK/STAT), interferon, and ERK 5 signaling (P < 0.01). By 24 hours, prominent pathways included P53- and Myc-induced apoptotic processes, pancreatic adenocarcinoma signaling, and phosphoinositide 3-kinase/v-akt murine thymoma vial oncogene homolog 1 (PI3/AKT) pathways. CONCLUSIONS: Our study reveals the ability to assess time-dependent changes in gene expression patterns in pancreatic cancer cells associated with infection and susceptibility to vaccinia viruses. This suggests that molecular assays may be useful to develop safer and more efficacious oncolyticvirotherapies and support the idea that these treatments may target pathways implicated in pancreatic cancer resistance to conventional therapies.

12.
Ann Surg Oncol ; 22(4): 1366-70, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25472650

ABSTRACT

BACKGROUND: Routine cross-sectional imaging for staging of early-stage cutaneous melanoma is not recommended. This study sought to investigate the use of imaging for staging of cutaneous melanoma in the United States. METHODS: Patients with nonmetastatic cutaneous melanoma newly diagnosed between 2000 and 2007 were identified from the Surveillance Epidemiology End Results-Medicare registry. Any imaging study performed within 90 days after diagnosis was considered a staging study. RESULTS: The study identified 25,643 patients, 3,116 (12.2 %) of whom underwent cross-sectional imaging: positron emission tomography (PET) (7.2 %), computed tomography (CT) (5.9 %), and magnetic resonance imaging (MRI) (0.6 %). From 2000 to 2007, the use of cross-sectional imaging increased from 8.7 to 16.1 % (p < 0.001), driven predominantly by increased usage of PET (4.2-12.1 %). Stratification by T and N classification showed that cross-sectional imaging was used for 8.6 % of T1, 14.3 % of T2, 18.6 % of T3, and 26.7 % of T4 tumors (p < 0.001) and for 33.3 % of node-positive patients versus 11.1 % of node-negative patients (p < 0.001). Factors predictive of cross-sectional imaging included T classification [odds ratio (OR) for T4 vs T1, 2.66; 95 % confidence interval (CI) 2.33-3.03], node positivity (OR 2.70; 95 % CI 2.36-3.10), more recent year of diagnosis (OR 2.05 for 2007 vs 2000; 95 % CI 1.74-2.42), atypical histology, and non-Caucasian race (OR 1.32; 95 % CI 1.02-1.73). CONCLUSIONS: The use of cross-sectional imaging for staging of early-stage cutaneous melanoma is increasing in the Medicare population. Better dissemination of guidelines and judicious use of imaging should be encouraged.


Subject(s)
Lymph Nodes/pathology , Melanoma/diagnosis , Melanoma/epidemiology , Neoplasm Staging/methods , Aged , Aged, 80 and over , Cohort Studies , Cross-Sectional Studies , Female , Fluorodeoxyglucose F18/pharmacokinetics , Follow-Up Studies , Humans , Lymph Nodes/diagnostic imaging , Magnetic Resonance Imaging , Male , Positron-Emission Tomography , Prognosis , Radiopharmaceuticals/pharmacokinetics , SEER Program , Skin Neoplasms , Tissue Distribution , Tomography, X-Ray Computed , United States/epidemiology , Melanoma, Cutaneous Malignant
13.
Mol Ther Oncolytics ; 1: 14007, 2015.
Article in English | MEDLINE | ID: mdl-27119098

ABSTRACT

Oncolytic viruses have made their mark on the cancer world as a potential therapeutic option, with the possible advantages of reduced side effects and strengthened treatment efficacy due to higher tumor selectivity. Results have been so promising, that oncolytic viral treatments have now been approved for clinical trials in several countries. However, clinical studies may benefit from the ability to noninvasively and serially identify sites of viral targeting via molecular imaging in order to provide safety, efficacy, and toxicity information. Furthermore, molecular imaging of oncolytic viral therapy may provide a more sensitive and specific diagnostic technique to detect tumor origin and, more importantly, presence of metastases. Several strategies have been investigated for molecular imaging of viral replication broadly categorized into optical and deep tissue imaging, utilizing several reporter genes encoding for fluorescence proteins, conditional enzymes, and membrane protein and transporters. Various imaging methods facilitate molecular imaging, including computer tomography, magnetic resonance imaging, positron emission tomography, single photon emission CT, gamma-scintigraphy, and photoacoustic imaging. In addition, several molecular probes are used for medical imaging, which act as targeting moieties or signaling agents. This review will explore the preclinical and clinical use of in vivo molecular imaging of replication-competent oncolytic viral therapy.

14.
J Exp Clin Cancer Res ; 33: 2, 2014 Jan 02.
Article in English | MEDLINE | ID: mdl-24383569

ABSTRACT

BACKGROUND: Gastric cancers have poor overall survival despite recent advancements in early detection methods, endoscopic resection techniques, and chemotherapy treatments. Vaccinia viral therapy has had promising therapeutic potential for various cancers and has a great safety profile. We investigated the therapeutic efficacy of a novel genetically-engineered vaccinia virus carrying the human sodium iodide symporter (hNIS) gene, GLV-1 h153, on gastric cancers and its potential utility for imaging with (99m)Tc pertechnetate scintigraphy and ¹²4I positron emission tomography (PET). METHODS: GLV-1 h153 was tested against five human gastric cancer cell lines using cytotoxicity and standard viral plaque assays. In vivo, subcutaneous flank tumors were generated in nude mice with human gastric cancer cells, MKN-74. Tumors were subsequently injected with either GLV-1 h153 or PBS and followed for tumor growth. (99m)Tc pertechnetate scintigraphy and ¹²4I microPET imaging were performed. RESULTS: GFP expression, a surrogate for viral infectivity, confirmed viral infection by 24 hours. At a multiplicity of infection (MOI) of 1, GLV-1 h153 achieved > 90% cytotoxicity in MNK-74, OCUM-2MD3, and AGS over 9 days, and >70% cytotoxicity in MNK- 45 and TMK-1. In vivo, GLV-1 h153 was effective in treating xenografts (p < 0.001) after 2 weeks of treatment. GLV-1 h153-infected tumors were readily imaged by (99m)Tc pertechnetate scintigraphy and ¹²4I microPET imaging 2 days after treatment. CONCLUSIONS: GLV-1 h153 is an effective oncolytic virus expressing the hNIS protein that can efficiently regress gastric tumors and allow deep-tissue imaging. These data encourages its continued investigation in clinical settings.


Subject(s)
Oncolytic Viruses/genetics , Stomach Neoplasms/therapy , Symporters/genetics , Vaccinia virus/genetics , Animals , Cell Line, Tumor , Female , Genetic Engineering , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Oncolytic Virotherapy , Radionuclide Imaging , Radiopharmaceuticals , Sodium Pertechnetate Tc 99m , Stomach Neoplasms/diagnostic imaging , Stomach Neoplasms/pathology , Tumor Burden , Virus Replication
15.
Am J Surg ; 206(6): 979-85; discussion 985-6, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24124660

ABSTRACT

BACKGROUND: Preoperative imaging for early-stage cutaneous melanoma is not recommended by current guidelines. Our goal was to investigate our institutional usage and utility. METHODS: Patients with clinically node-negative cutaneous melanoma undergoing surgery with sentinel lymph node biopsy were identified retrospectively. Any melanoma-related imaging after diagnosis and before surgery was considered a staging study. RESULTS: Five hundred fifteen studies were performed in 409 of 546 (75%) patients. Chest x-rays was performed in 70% and advanced imaging in 14% (computed tomography imaging, magnetic resonance imaging, ultrasound, and positron-emission computed tomography imaging). No metastatic lesions were identified. A Breslow thickness greater than 4 mm (odds ratio = 6.46 vs <1 mm; 95% confidence interval, 2.07 to 20.15) and male sex (odds ratio = 2.62 vs female; 95% confidence interval, 1.26 to 5.46) were associated with an increased likelihood of advanced imaging. CONCLUSIONS: Preoperative imaging was performed in the majority of patients with node-negative melanoma, with 14% undergoing advanced studies. No metastatic lesions were identified, confirming the limited utility in this setting.


Subject(s)
Magnetic Resonance Imaging/statistics & numerical data , Melanoma/diagnosis , Neoplasm Staging/methods , Positron-Emission Tomography/statistics & numerical data , Preoperative Care/statistics & numerical data , Tomography, Emission-Computed/statistics & numerical data , Tomography, X-Ray Computed/statistics & numerical data , Adult , Aged , Female , Follow-Up Studies , Humans , Male , Middle Aged , Predictive Value of Tests , Preoperative Care/methods , Retrospective Studies , Skin Neoplasms , Melanoma, Cutaneous Malignant
16.
Surgery ; 154(3): 496-503, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23972655

ABSTRACT

BACKGROUND: Electroporation uses an electric field to induce pores in the cell membrane that can transfer macromolecules into target cells. Modulation of electrical parameters leads to irreversible electroporation (IRE), which is being developed for tissue ablation. We sought to evaluate whether the application of IRE may induce a lesser electric field in the periphery where reversible electroporation may occur, facilitating gene transfer of a granulocyte macrophage colony-stimulating factor (GM-CSF) plasmid to produce its biologic response. METHODS: Yorkshire pigs underwent laparotomy, and IRE of the liver was performed during hepatic arterial infusion of 1 or 7 mg of a naked human GM-CSF plasmid. The serum, liver, lymph nodes, and bone marrow were harvested for analysis. RESULTS: Human GM-CSF level rose from undetectable to 131 pg/mL in the serum at 24 hours after IRE and plasmid infusion. The liver demonstrated an ablation zone surrounded by an immune infiltrate that had greater macrophage intensity than when treated with IRE or plasmid infusion alone. This dominance of macrophages was dose dependent. Distant effects of GM-CSF were found in the bone marrow, where proliferating myeloid cells increased from 14% to 25%. CONCLUSION: IRE facilitated gene transfer of the GM-CSF plasmid and brought about a local and systemic biologic response. This technique holds potential for tumor eradication and immunotherapy of residual cancer.


Subject(s)
Electroporation/methods , Gene Transfer Techniques , Genetic Therapy , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Neoplasms/therapy , Animals , Humans , Plasmids , Swine
17.
Surgery ; 153(6): 787-93, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23489942

ABSTRACT

BACKGROUND: Irreversible electroporation (IRE) is a novel ablation technique that induces permanent membrane permeability and cell death. We are interested in ultrasound B-mode and elastography to monitor IRE ablation in the liver. METHODS: Yorkshire pigs underwent IRE ablation of the liver and were imaged with ultrasound B-mode and elastography. Histologic evaluation of cell death by triphenyltetrazolium chloride and hematoxylin and eosin staining was performed. RESULTS: Elastography showed that liver ablated by IRE exhibited increased tissue stiffness with a peak strain ratio of 2.22. The IRE lesion had a discrete border without bubble artifact, and the lesion size significantly correlated with area of cell death on histology. IRE ablation was unaffected by presence of large blood vessels or bile ducts. CONCLUSION: IRE ablation led to increased tissue stiffness that was detectable by elastography and indicative of cell death. Elastography may complement B-mode ultrasonography to monitor IRE ablation of the liver.


Subject(s)
Ablation Techniques/methods , Elasticity Imaging Techniques , Electrochemotherapy/methods , Liver/diagnostic imaging , Liver/surgery , Animals , Cell Membrane Permeability , Humans , Liver/metabolism , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/metabolism , Liver Neoplasms/surgery , Models, Animal , Sus scrofa
18.
J Nucl Med ; 53(12): 1933-42, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23139088

ABSTRACT

UNLABELLED: To assess therapeutic response and potential toxicity of oncolytic virotherapy, a noninvasive, deep-tissue imaging modality is needed. This study aimed to assess the feasibility, parameters, and determining factors of serial imaging and long-term monitoring of virotherapy and the therapeutic response of pancreatic cancer xenografts treated with a vaccinia virus carrying the human sodium iodide symporter GLV-1h153. METHODS: Pancreatic cancer xenografts (PANC-1) in nude mice were treated systemically or intratumorally with GLV-1h153 and serially imaged using (124)I PET at 1, 2, 3, and 5 wk after viral injection. Signal intensity was compared with tumor therapeutic response and optical imaging, and tumors were histologically analyzed for morphology and the presence of virus. Autoradiography was performed using technetium-pertechnetate and γ-scintigraphy to assess determining factors for radiouptake in tumors. Finally, the enhanced therapeutic effect of combination therapy with GLV-1h153 and systemic radioiodine was assessed. RESULTS: GLV-1h153 successfully facilitated serial long-term imaging of virotherapy, with PET signal intensity correlating to tumor response. GLV-1h153 colonization of tumors mediated radioiodine uptake at potentially therapeutic doses. Successful radiouptake required the presence of virus, adequate blood flow, and viable tissue, whereas loss of signal intensity was linked to tumor death and necrosis. Finally, combining systemically administered GLV-1h153 and (131)I led to enhanced tumor kill when compared with virus or (131)I alone (P < 0.01). CONCLUSION: GLV-1h153 is a promising oncolytic agent for the treatment, long-term imaging, and monitoring of therapeutic response in a xenograft model of pancreatic cancer. GLV-1h153 provided insight into tumor biologic activity and facilitated enhanced tumor kill when combined with systemic targeted radiotherapy. These results warrant further investigation into parameters and potential synergistic effects of combination therapy.


Subject(s)
Molecular Imaging/methods , Oncolytic Virotherapy/methods , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/therapy , Radiotherapy, Image-Guided/methods , Symporters/genetics , Vaccinia virus/genetics , Animals , Cell Line, Tumor , Feasibility Studies , Humans , Iodine Radioisotopes , Mice , Optical Imaging , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/radiotherapy , Positron-Emission Tomography , Time Factors , Treatment Outcome , Tumor Burden
19.
PLoS One ; 7(8): e41647, 2012.
Article in English | MEDLINE | ID: mdl-22912675

ABSTRACT

INTRODUCTION: Oncolytic viruses show promise for treating cancer. However, to assess therapy and potential toxicity, a noninvasive imaging modality is needed. This study aims to determine the in vivo biodistribution, and imaging and timing characteristics of a vaccinia virus, GLV-1h153, encoding the human sodium iodide symporter (hNIS. METHODS: GLV-1h153 was modified from GLV-1h68 to encode the hNIS gene. Timing of cellular uptake of radioiodide (131)I in human pancreatic carcinoma cells PANC-1 was assessed using radiouptake assays. Viral biodistribution was determined in nude mice bearing PANC-1 xenografts, and infection in tumors confirmed histologically and optically via Green Fluorescent Protein (GFP) and bioluminescence. Timing characteristics of enhanced radiouptake in xenografts were assessed via (124)I-positron emission tomography (PET). Detection of systemic administration of virus was investigated with both (124)I-PET and 99m-technecium gamma-scintigraphy. RESULTS: GLV-1h153 successfully facilitated time-dependent intracellular uptake of (131)I in PANC-1 cells with a maximum uptake at 24 hours postinfection (P<0.05). In vivo, biodistribution profiles revealed persistence of virus in tumors 5 weeks postinjection at 10(9) plaque-forming unit (PFU)/gm tissue, with the virus mainly cleared from all other major organs. Tumor infection by GLV-1h153 was confirmed via optical imaging and histology. GLV-1h153 facilitated imaging virus replication in tumors via PET even at 8 hours post radiotracer injection, with a mean %ID/gm of 3.82 ± 0.46 (P<0.05) 2 days after intratumoral administration of virus, confirmed via tissue radiouptake assays. One week post systemic administration, GLV-1h153-infected tumors were detected via (124)I-PET and 99m-technecium-scintigraphy. CONCLUSION: GLV-1h153 is a promising oncolytic agent against pancreatic cancer with a promising biosafety profile. GLV-1h153 facilitated time-dependent hNIS-specific radiouptake in pancreatic cancer cells, facilitating detection by PET with both intratumoral and systemic administration. Therefore, GLV-1h153 is a promising candidate for the noninvasive imaging of virotherapy and warrants further study into longterm monitoring of virotherapy and potential radiocombination therapies with this treatment and imaging modality.


Subject(s)
Molecular Imaging , Oncolytic Viruses/genetics , Oncolytic Viruses/physiology , Symporters/genetics , Vaccinia virus/genetics , Vaccinia virus/physiology , Virus Replication , Animals , Biological Transport , Cell Line, Tumor , Cell Transformation, Neoplastic , Humans , Iodine Radioisotopes/metabolism , Male , Mice , Optical Imaging , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/virology , Positron-Emission Tomography , Time Factors , Tissue Distribution
20.
Ann Surg Oncol ; 19 Suppl 3: S665-74, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22258815

ABSTRACT

BACKGROUND: Replication-competent oncolytic viruses have shown great promise as a potential cancer treatment. This study aimed to determine whether a novel vaccinia virus, GLV-1h151, with genetic modifications enhancing cancer specificity and enabling virus detection, is effective against a range of human cancers and is safe when administered in preclinical models. METHODS: GLV-1h151 was modified with deletion of thymidine kinase enhancing specificity and insertion of the green fluorescent protein (GFP) gene. The virus was tested in several human cancer cell lines for cytotoxicity including breast, lung, pancreatic, and colorectal. Virus replication was assessed via visualization of GFP expression and bioluminescence, and viral plaque assays. Finally, GLV-1h151 was administered systemically or intratumorally in mice with pancreatic cancer xenografts (PANC-1) to assess virus biodistribution, toxicity, and effect on tumor growth. RESULTS: GLV-1h151 effectively infected, replicated in, and killed several cancer cell types. Detection and visualization of virus replication was successful via fluorescence imaging of GFP expression, which was dose dependent. When administered intravenously or intratumorally in vivo, GLV-1h151 regressed tumor growth (P < 0.001) and displayed a good biosafety profile. GLV-1h151 infection and replication in tumors was successfully visualized via GFP and bioluminescence, with virus presence in tumors confirmed histologically. CONCLUSIONS: GLV-1h151 is effective as an oncolytic agent against a wide range of cancers in cell culture and is effective against pancreatic human xenografts displaying a good biosafety profile and ability to be detected via optical imaging. GLV-1h151 thus adds another potential medium for the killing of cancer and detection of virus in infected tissue.


Subject(s)
Oncolytic Virotherapy , Oncolytic Viruses/physiology , Pancreatic Neoplasms/therapy , Vaccinia virus/physiology , Animals , Cell Line, Tumor , Cell Survival , Flow Cytometry , Genetic Engineering , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Injections, Intralesional , Injections, Intravenous , Male , Mice , Mice, Nude , Neoplasms/therapy , Oncolytic Viruses/genetics , Pancreatic Neoplasms/virology , Vaccinia virus/genetics , Virus Replication , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...