Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where E. Levy is active.

Publication


Featured researches published by E. Levy.


CardioVascular and Interventional Radiology | 2012

Multimodality image fusion-guided procedures: technique, accuracy, and applications.

Nadine Abi-Jaoudeh; Jochen Kruecker; Samuel Kadoury; Hicham Kobeiter; Aradhana M. Venkatesan; E. Levy; Bradford J. Wood

Personalized therapies play an increasingly critical role in cancer care: Image guidance with multimodality image fusion facilitates the targeting of specific tissue for tissue characterization and plays a role in drug discovery and optimization of tailored therapies. Positron-emission tomography (PET), magnetic resonance imaging (MRI), and contrast-enhanced computed tomography (CT) may offer additional information not otherwise available to the operator during minimally invasive image-guided procedures, such as biopsy and ablation. With use of multimodality image fusion for image-guided interventions, navigation with advanced modalities does not require the physical presence of the PET, MRI, or CT imaging system. Several commercially available methods of image-fusion and device navigation are reviewed along with an explanation of common tracking hardware and software. An overview of current clinical applications for multimodality navigation is provided.


Radiology | 2011

Real-time FDG PET Guidance during Biopsies and Radiofrequency Ablation Using Multimodality Fusion with Electromagnetic Navigation

Aradhana M. Venkatesan; Samuel Kadoury; Nadine Abi-Jaoudeh; E. Levy; Roberto Maass-Moreno; Jochen Krücker; Sandeep Dalal; Sheng Xu; Neil Glossop; Bradford J. Wood

PURPOSE To assess the feasibility of combined electromagnetic device tracking and computed tomography (CT)/ultrasonography (US)/fluorine 18 fluorodeoxyglucose (FDG) positron emission tomography (PET) fusion for real-time feedback during percutaneous and intraoperative biopsies and hepatic radiofrequency (RF) ablation. MATERIALS AND METHODS In this HIPAA-compliant, institutional review board-approved prospective study with written informed consent, 25 patients (17 men, eight women) underwent 33 percutaneous and three intraoperative biopsies of 36 FDG-avid targets between November 2007 and August 2010. One patient underwent biopsy and RF ablation of an FDG-avid hepatic focus. Targets demonstrated heterogeneous FDG uptake or were not well seen or were totally inapparent at conventional imaging. Preprocedural FDG PET scans were rigidly registered through a semiautomatic method to intraprocedural CT scans. Coaxial biopsy needle introducer tips and RF ablation electrode guider needle tips containing electromagnetic sensor coils were spatially tracked through an electromagnetic field generator. Real-time US scans were registered through a fiducial-based method, allowing US scans to be fused with intraprocedural CT and preacquired FDG PET scans. A visual display of US/CT image fusion with overlaid coregistered FDG PET targets was used for guidance; navigation software enabled real-time biopsy needle and needle electrode navigation and feedback. RESULTS Successful fusion of real-time US to coregistered CT and FDG PET scans was achieved in all patients. Thirty-one of 36 biopsies were diagnostic (malignancy in 18 cases, benign processes in 13 cases). RF ablation resulted in resolution of targeted FDG avidity, with no local treatment failure during short follow-up (56 days). CONCLUSION Combined electromagnetic device tracking and image fusion with real-time feedback may facilitate biopsies and ablations of focal FDG PET abnormalities that would be challenging with conventional image guidance.


Journal of Hepatology | 2017

Tremelimumab in combination with ablation in patients with advanced hepatocellular carcinoma

Austin Duffy; Susanna Varkey Ulahannan; Oxana Makorova-Rusher; Osama E. Rahma; Heiner Wedemeyer; Drew Pratt; Jeremy L. Davis; Marybeth S. Hughes; Theo Heller; Mei ElGindi; Ashish Uppala; Firouzeh Korangy; David E. Kleiner; William D. Figg; David Venzon; Seth M. Steinberg; Aradhana M. Venkatesan; Venkatesh Krishnasamy; Nadine Abi-Jaoudeh; E. Levy; Brad J. Wood; Tim F. Greten

BACKGROUND & AIMS Tremelimumab is a fully human monoclonal antibody that binds to cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) on the surface of activated T lymphocytes. Ablative therapies induce a peripheral immune response which may enhance the effect of anti-CTLA4 treatment in patients with advanced hepatocellular carcinoma (HCC). This study aimed to demonstrate whether tremelimumab could be combined safely and feasibly with ablation. METHODS Thirty-two patients with HCC were enrolled: male:female: 28:4; median age: 62 (range 36-76). Patients were given tremelimumab at two dose levels (3.5 and 10mg/kg i.v.) every 4weeks for 6 doses, followed by 3-monthly infusions until off-treatment criteria were met. On day 36, patients underwent subtotal radiofrequency ablation or chemoablation. Staging was performed by contrast-enhanced CT or MRI scan every 8weeks. RESULTS No dose-limiting toxicities were encountered. The most common toxicity was pruritus. Of the 19 evaluable patients, five (26.3%; 95% CI: 9.1-51.2%) achieved a confirmed partial response. Twelve of 14 patients with quantifiable HCV experienced a marked reduction in viral load. Six-week tumor biopsies showed a clear increase in CD8+ T cells in patients showing a clinical benefit only. Six and 12-month probabilities of tumor progression free survival for this refractory HCC population were 57.1% and 33.1% respectively, with median time to tumor progression of 7.4months (95% CI 4.7 to 19.4months). Median overall survival was 12.3months (95% CI 9.3 to 15.4months). CONCLUSIONS Tremelimumab in combination with tumor ablation is a potential new treatment for patients with advanced HCC, and leads to the accumulation of intratumoral CD8+ T cells. Positive clinical activity was seen, with a possible surrogate reduction in HCV viral load. LAY SUMMARY Studies have shown that the killing of tumors by direct methods (known as ablation) can result in the immune system being activated or switched on. The immune system could potentially also recognize and kill the cancer that is left behind. There are new drugs available known as immune checkpoint inhibitors which could enhance this effect. Here, we test one of these drugs (tremelimumab) together with ablation. CLINICAL TRIAL NUMBER ClinicalTrials.gov: NCT01853618.


Journal of Vascular and Interventional Radiology | 2010

Navigation Systems for Ablation

Bradford J. Wood; Jochen Kruecker; Nadine Abi-Jaoudeh; Julia K. Locklin; E. Levy; Sheng Xu; Luigi Solbiati; Ankur Kapoor; Hayet Amalou; Aradhana M. Venkatesan

Navigation systems, devices, and intraprocedural software are changing the way interventional oncology is practiced. Before the development of precision navigation tools integrated with imaging systems, thermal ablation of hard-to-image lesions was highly dependent on operator experience, spatial skills, and estimation of positron emission tomography-avid or arterial-phase targets. Numerous navigation systems for ablation bring the opportunity for standardization and accuracy that extends the operators ability to use imaging feedback during procedures. In this report, existing systems and techniques are reviewed and specific clinical applications for ablation are discussed to better define how these novel technologies address specific clinical needs and fit into clinical practice.


Trials | 2011

Regional Chemotherapy in Locally Advanced Pancreatic Cancer: RECLAP Trial

Jeremy L. Davis; Prakash Pandalai; R. Taylor Ripley; Russell C. Langan; Seth M. Steinberg; Melissa Walker; Mary Ann Toomey; E. Levy; Itzhak Avital

BackgroundPancreatic cancer is the fourth leading cause of cancer death in the United States. Surgery offers the only chance for cure. However, less than twenty percent of patients are considered operative candidates at the time of diagnosis. A common reason for being classified as unresectable is advanced loco-regional disease.A review of the literature indicates that almost nine hundred patients with pancreatic cancer have received regional chemotherapy in the last 15 years. Phase I studies have shown regional administration of chemotherapy to be safe. The average reported response rate was approximately 26%. The average 1-year survival was 39%, with an average median survival of 9 months. Of the patients that experienced a radiographic response to therapy, 78 (78/277, 28%) patients underwent exploratory surgery following regional chemotherapy administration; thirty-two (41%) of those patients were amenable to pancreatectomy. None of the studies performed analyses to identify factors predicting response to regional chemotherapy.Progressive surgical techniques combined with current neoadjuvant chemoradiotherapy strategies have already yielded emerging support for a multimodality approach to treatment of advanced pancreatic cancer.Intravenous gemcitabine is the current standard treatment of pancreatic cancer. However, >90% of the drug is secreted unchanged affecting toxicity but not the cancer per se. Gemcitabine is converted inside the cell into its active drug form in a rate limiting reaction. We hypothesize that neoadjuvant regional chemotherapy with continuous infusion of gemcitabine will be well tolerated and may improve resectability rates in cases of locally advanced pancreatic cancer.DesignThis is a phase I study designed to evaluate the feasibility and toxicity of super-selective intra-arterial administration of gemcitabine in patients with locally advanced, unresectable pancreatic adenocarcinoma. Patients considered unresectable due to locally advanced pancreatic cancer will receive super-selective arterial infusion of gemcitabine over 24 hours via subcutaneous indwelling port. Three to six patients will be enrolled per dose cohort, with seven cohorts, plus an additional six patients at the maximum tolerated dose; accrual is expected to last 36 months. Secondary objectives will include the determination of progression free and overall survival, as well as the conversion rate from unresectable to potentially resectable pancreatic cancer.Trial RegistrationClinicalTrials.gov ID: NCT01294358


Journal of Controlled Release | 2017

Characterization of a novel intrinsically radiopaque Drug-eluting Bead for image-guided therapy: DC Bead LUMI™

Koorosh Ashrafi; Yiqing Tang; Hugh Britton; Orianne Domenge; Delphine Blino; A. J. Bushby; Kseniya Shuturminska; Mark den Hartog; Alessandro Radaelli; Ayele H. Negussie; Andrew S. Mikhail; David L. Woods; Venkatesh Krishnasamy; E. Levy; Bradford J. Wood; Sean Willis; Matthew R. Dreher; Andrew L. Lewis

ABSTRACT We have developed a straightforward and efficient method of introducing radiopacity into Polyvinyl alcohol (PVA)‐2‐Acrylamido‐2‐methylpropane sulfonic acid (AMPS) hydrogel beads (DC Bead™) that are currently used in the clinic to treat liver malignancies. Coupling of 2,3,5‐triiodobenzaldehyde to the PVA backbone of pre‐formed beads yields a uniformly distributed level of iodine attached throughout the bead structure (˜ 150 mg/mL) which is sufficient to be imaged under standard fluoroscopy and computed tomography (CT) imaging modalities used in treatment procedures (DC Bead LUMI™). Despite the chemical modification increasing the density of the beads to ˜ 1.3 g/cm3 and the compressive modulus by two orders of magnitude, they remain easily suspended, handled and administered through standard microcatheters. As the core chemistry of DC Bead LUMI™ is the same as DC Bead™, it interacts with drugs using ion‐exchange between sulfonic acid groups on the polymer and the positively charged amine groups of the drugs. Both doxorubicin (Dox) and irinotecan (Iri) elution kinetics for all bead sizes evaluated were within the parameters already investigated within the clinic for DC Bead™. Drug loading did not affect the radiopacity and there was a direct relationship between bead attenuation and Dox concentration. The ability (Dox)‐loaded DC Bead LUMI™ to be visualized in vivo was demonstrated by the administration of into hepatic arteries of a VX2 tumor‐bearing rabbit under fluoroscopy, followed by subsequent CT imaging.


Journal of Vascular and Interventional Radiology | 2016

Preparation of Radiopaque Drug-Eluting Beads for Transcatheter Chemoembolization.

Carmen Gacchina Johnson; Yiqing Tang; A. Beck; Matthew R. Dreher; David L. Woods; Ayele H. Negussie; Danielle Donahue; E. Levy; Sean Willis; Andrew L. Lewis; Bradford J. Wood; Karun V. Sharma

PURPOSE To develop a simple method to produce radiopaque drug-eluting microspheres (drug-eluting beads [DEBs]) that could be incorporated into the current clinical transcatheter arterial chemoembolization workflow and evaluate their performance in vitro and in vivo. MATERIALS AND METHODS An ethiodized oil (Lipiodol; Guerbet, Villepinte, France) and ethanol solution was added to a lyophilized 100-300 µm bead before loading with doxorubicin. These radiopaque drug-eluting beads (DEBs; Biocompatibles UK Ltd, Farnham, United Kingdom) were evaluated in vitro for x-ray attenuation, composition, size, drug loading and elution, and correlation between attenuation and doxorubicin concentration. In vivo conspicuity was evaluated in a VX2 tumor model. RESULTS Lipiodol was loaded into lyophilized beads using two glass syringes and a three-way stopcock. Maximum bead attenuation was achieved within 30 minutes. X-ray attenuation of radiopaque beads increased linearly (21-867 HU) with the amount of beads (0.4-12.5 vol%; R(2) = 0.9989). Doxorubicin loading efficiency and total amount eluted were similar to DC Bead (Biocompatibles UK Ltd); however, the elution rate was slower for radiopaque DEBs (P < .05). Doxorubicin concentration linearly correlated with x-ray attenuation of radiopaque DEBs (R(2) = 0. 99). Radiopaque DEBs were seen in tumor feeding arteries after administration by fluoroscopy, computed tomography, and micro-computed tomography, and their location was confirmed by histology. CONCLUSIONS A simple, rapid method to produce radiopaque DEBs was developed. These radiopaque DEBs provided sufficient conspicuity to be visualized with x-ray imaging techniques.


International Journal of Cancer | 2016

Modulation of tumor eIF4E by antisense inhibition: A phase I/II translational clinical trial of ISIS 183750 – an antisense oligonucleotide against eIF4E – in combination with irinotecan in solid tumors and irinotecan‐refractory colorectal cancer

Austin Duffy; Oxana V. Makarova-Rusher; Susanna Varkey Ulahannan; Osama E. Rahma; Suzanne Fioravanti; Melissa Walker; S. Abdullah; Mark Raffeld; Victoria L. Anderson; Nadine Abi-Jaoudeh; E. Levy; Bradford J. Wood; Su Jae Lee; Yasushi Tomita; Jane B. Trepel; Seth M. Steinberg; A.S. Revenko; A.R. MacLeod; Cody J. Peer; William D. Figg; Tim F. Greten

The eukaryotic translation initiation factor 4E (eIF4E) is a potent oncogene that is found to be dysregulated in 30% of human cancer, including colorectal carcinogenesis (CRC). ISIS 183750 is a second‐generation antisense oligonucleotide (ASO) designed to inhibit the production of the eIF4E protein. In preclinical studies we found that EIF4e ASOs reduced expression of EIF4e mRNA and inhibited proliferation of colorectal carcinoma cells. An additive antiproliferative effect was observed in combination with irinotecan. We then performed a clinical trial evaluating this combination in patients with refractory cancer. No dose‐limiting toxicities were seen but based on pharmacokinetic data and tolerability the dose of irinotecan was reduced to 160 mg/m2 biweekly. Efficacy was evaluated in 15 patients with irinotecan‐refractory colorectal cancer. The median time of disease control was 22.1 weeks. After ISIS 183750 treatment, peripheral blood levels of eIF4E mRNA were decreased in 13 of 19 patients. Matched pre‐ and posttreatment tumor biopsies showed decreased eIF4E mRNA levels in five of nine patients. In tumor tissue, the intracellular and stromal presence of ISIS 183750 was detected by IHC in all biopsied patients. Although there were no objective responses stable disease was seen in seven of 15 (47%) patients who were progressing before study entry, six of whom were stable at the time of the week 16 CT scan. We were also able to confirm through mandatory pre‐ and posttherapy tumor biopsies penetration of the ASO into the site of metastasis.


Journal of Vascular and Interventional Radiology | 2016

Microvascular Perfusion Changes following Transarterial Hepatic Tumor Embolization.

Carmen Gacchina Johnson; Karun Sharma; E. Levy; David L. Woods; Aaron H. Morris; John Bacher; Andrew L. Lewis; Bradford J. Wood; Matthew R. Dreher

PURPOSE To quantify changes in tumor microvascular (< 1 mm) perfusion relative to commonly used angiographic endpoints. MATERIALS AND METHODS Rabbit Vx2 liver tumors were embolized with 100-300-μm LC Bead particles to endpoints of substasis or complete stasis (controls were not embolized). Microvascular perfusion was evaluated by delivering two different fluorophore-conjugated perfusion markers (ie, lectins) through the catheter before embolization and 5 min after reaching the desired angiographic endpoint. Tumor microvasculature was labeled with an anti-CD31 antibody and analyzed with fluorescence microscopy for perfusion marker overlap/mismatch. Data were analyzed by analysis of variance and post hoc test (n = 3-5 per group; 18 total). RESULTS Mean microvascular density was 70 vessels/mm(2) ± 17 (standard error of the mean), and 81% ± 1 of microvasculature (ie, CD31(+) structures) was functionally perfused within viable Vx2 tumor regions. Embolization to the extent of substasis eliminated perfusion in 37% ± 9 of perfused microvessels (P > .05 vs baseline), whereas embolization to the extent of angiographic stasis eliminated perfusion in 56% ± 8 of perfused microvessels. Persistent microvascular perfusion following embolization was predominantly found in the tumor periphery, adjacent to normal tissue. Newly perfused microvasculature was evident following embolization to substasis but not when embolization was performed to complete angiographic stasis. CONCLUSIONS Nearly half of tumor microvasculature remained patent despite embolization to complete angiographic stasis. The observed preservation of tumor microvasculature perfusion with angiographic endpoints of substasis and stasis may have implications for tumor response to embolotherapy.


Journal of Vascular and Interventional Radiology | 2015

Direct Quantification and Comparison of Intratumoral Hypoxia following Transcatheter Arterial Embolization of VX2 Liver Tumors with Different Diameter Microspheres

E. Levy; Carmen Gacchina Johnson; Genevieve Jacobs; David L. Woods; Karun V. Sharma; John Bacher; Andrew L. Lewis; Matthew R. Dreher; Bradford J. Wood

PURPOSE To evaluate the effect of embolic diameter on achievement of hypoxia after embolization in an animal model of liver tumors. MATERIALS AND METHODS Inoculation of VX2 tumors in the left liver lobe was performed successfully in 12 New Zealand white rabbits weighing 3.7 kg ± 0.5 (mean ± SD). Tumors were deemed eligible for oxygen measurements when the maximum transverse diameter measured 15 mm or more by ultrasound examination. Direct monitoring of oxygenation of implanted rabbit hepatic VX2 tumors was performed with a fiberoptic electrode during and after transarterial embolization of the proper hepatic artery to angiographic flow stasis with microspheres measuring 70-150 μm, 100-300 μm, or 300-500 μm in diameter. RESULTS Failure to achieve tumor hypoxia as defined despite angiographic flow stasis was observed in 10 of 11 animals. Embolization microsphere size effect failed to demonstrate a significant trend on hypoxia outcome among the diameters tested, and pair-wise comparisons of different embolic diameter treatment groups showed no difference in hypoxia outcome. All microsphere diameters tested resulted in similar absolute reduction (24.3 mm Hg ± 18.3, 29.1 mm Hg ± 1.8, and 19.9 mm Hg ± 9.3, P = .66) and percentage decrease in oxygen (56.0 mm Hg ± 23.9, 56.0 mm Hg ± 6.4, and 35.8 mm Hg ± 20.6, P = .65). Pair-wise comparisons for percent tumor area occupied by embolic agents showed a significantly reduced fraction for 300-500 μm diameters compared with 70-150 μm diameters (P < .05). CONCLUSIONS In the rabbit VX2 liver tumor model, three tested microsphere diameters failed to cause tumor hypoxia as measured by a fiberoptic probe sensor according to the adopted hypoxia definitions.

Collaboration


Dive into the E. Levy's collaboration.

Top Co-Authors

Avatar

Bradford J. Wood

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Venkatesh Krishnasamy

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

David L. Woods

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Matthew R. Dreher

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Nadine Abi-Jaoudeh

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew S. Mikhail

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Aradhana M. Venkatesan

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ayele H. Negussie

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

John W. Karanian

Center for Devices and Radiological Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge