Network


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

Hotspot


Dive into the research topics where Jeane Chen is active.

Publication


Featured researches published by Jeane Chen.


Journal of Controlled Release | 2014

Poly(lactide-co-glycolide) microspheres for MRI-monitored transcatheter delivery of sorafenib to liver tumors.

Jeane Chen; A. Sheu; Weiguo Li; Zhuoli Zhang; Dong Hyun Kim; Robert J. Lewandowski; Reed A. Omary; Lonnie D. Shea; Andrew C. Larson

The multi-kinase inhibitor (MKI) sorafenib can be an effective palliative therapy for patients with hepatocellular carcinoma (HCC). However, patient tolerance is often poor due to common systemic side effects following oral administration. Local transcatheter delivery of sorafenib to liver tumors has the potential to reduce systemic toxicities while increasing the dose delivered to targeted tumors. We developed sorafenib-eluting PLG microspheres for delivery by intra-hepatic transcatheter infusion in an orthotropic rodent HCC model. The particles also encapsulated iron-oxide nanoparticles permitting magnetic resonance imaging (MRI) of intra-hepatic biodistributions. The PLG microspheres (diameter≈1μm) were loaded with 18.6% (w/w) sorafenib and 0.54% (w/w) ferrofluid and 65.2% of the sorafenib was released within 72h of media exposure. In vitro studies demonstrated significant reductions in HCC cell proliferation with increasing doses of the sorafenib-eluting microspheres, where the estimated IC50 was a 29μg/mL dose of microspheres. During in vivo studies, MRI permitted intra-procedural visualization of intra-hepatic microsphere delivery. At 72h after microsphere infusion, microvessel density was significantly reduced in tumors treated with the sorafenib-eluting microspheres compared to both sham control tumors (by 35%) and controls (by 30%). These PLG microspheres offer the potential to increase the efficacy of molecularly targeted MKI therapies while reducing systemic exposures via selective catheter-directed delivery to HCC.


Theranostics | 2015

MRI visible drug eluting magnetic microspheres for transcatheter intra-arterial delivery to liver tumors

Dong Hyun Kim; Jeane Chen; Reed A. Omary; Andrew C. Larson

Magnetic resonance imaging (MRI)-visible amonafide-eluting alginate microspheres were developed for targeted arterial-infusion chemotherapy. These alginate microspheres were synthesized using a highly efficient microfluidic gelation process. The microspheres included magnetic clusters formed by USPIO nanoparticles to permit MRI and a sustained drug-release profile. The biocompatibility, MR imaging properties and amonafide release kinetics of these microspheres were investigated during in vitro studies. A xenograft rodent model was used to demonstrate the feasibility to deliver these microspheres to liver tumors using hepatic transcatheter intra-arterial infusions and potential to visualize the intra-hepatic delivery of these microspheres to both liver tumor and normal tissues with MRI immediately after infusion. This approach offer the potential for catheter-directed drug delivery to liver tumors for reduced systemic toxicity and superior therapeutic outcomes.


Nanomedicine: Nanotechnology, Biology and Medicine | 2017

An antigen-encapsulating nanoparticle platform for TH1/17 immune tolerance therapy

Derrick P. McCarthy; Jonathan Woon Teck Yap; Christopher T. Harp; W. Kelsey Song; Jeane Chen; Ryan M. Pearson; Stephen D. Miller; Lonnie D. Shea

Tolerogenic nanoparticles (NPs) are rapidly being developed as specific immunotherapies to treat autoimmune disease. However, many NP-based therapies conjugate antigen (Ag) directly to the NP posing safety concerns due to antibody binding or require the co-delivery of immunosuppressants to induce tolerance. Here, we developed Ag encapsulated NPs comprised of poly(lactide-co-glycolide) [PLG(Ag)] and investigated the mechanism of action for Ag-specific tolerance induction in an autoimmune model of T helper type 1/17 dysfunction - relapse-remitting experimental autoimmune encephalomyelitis (R-EAE). PLG(Ag) completely abrogated disease induction in an organ specific manner, where the spleen was dispensable for tolerance induction. PLG(Ag) delivered intravenously distributed to the liver, associated with macrophages, and recruited Ag-specific T cells. Furthermore, programmed death ligand 1 (PD-L1) was increased on Ag presenting cells and PD-1 blockade lessened tolerance induction. The robust promotion of tolerance by PLG(Ag) without co-delivery of immunosuppressive drugs, suggests that these NPs effectively deliver antigen to endogenous tolerogenic pathways.


ACS Applied Materials & Interfaces | 2016

Acidic pH-Triggered Drug-Eluting Nanocomposites for Magnetic Resonance Imaging-Monitored Intra-arterial Drug Delivery to Hepatocellular Carcinoma

Wooram Park; Jeane Chen; Soojeong Cho; Sin Jung Park; Andrew C. Larson; Kun Na; Dong Hyun Kim

Transcatheter hepatic intra-arterial (IA) injection has been considered as an effective targeted delivery technique for hepatocellular carcinoma (HCC). Recently, drug-eluting beads (DEB) were developed for transcatheter IA delivery to HCC. However, the conventional DEB has offered relatively modest survival benefits. It can be difficult to control drug loading/release from DEB and to monitor selective delivery to the targeted tumors. Embolized DEBs in hepatic arteries frequently induce hypoxic and low pH conditions, promoting cancer cell growth. In this study, an acidic pH-triggered drug-eluting nanocomposite (pH-DEN) including superparamagnetic iron oxide nanocubes and pH-responsive synthetic peptides with lipid tails [octadecylamine-p(API-l-Asp)10] was developed for magnetic resonance imaging (MRI)-monitored transcatheter delivery of sorafenib (the only FDA-approved systemic therapy for liver cancer) to HCC. The synthesized sorafenib-loaded pH-DENs exhibited distinct pH-triggered drug release behavior at acidic pH levels and highly sensitive MR contrast effects. In an orthotopic HCC rat model, successful hepatic IA delivery and distribution of sorafenib-loaded pH-DEN was confirmed with MRI. IA-delivered sorafenib-loaded pH-DENs elicited significant tumor growth inhibition in a rodent HCC model. These results indicate that the sorafenib-pH-DENs platform has the potential to be used as an advanced tool for liver-directed IA treatment of unresectable HCC.


Scientific Reports | 2016

Multimodal Imaging of Nanocomposite Microspheres for Transcatheter Intra-Arterial Drug Delivery to Liver Tumors

Dong Hyun Kim; Weiguo Li; Jeane Chen; Zhuoli Zhang; Richard M. Green; Sui Huang; Andrew C. Larson

A modern multi-functional drug carrier is critically needed to improve the efficacy of image-guided catheter-directed approaches for the treatment of hepatic malignancies. For this purpose, a nanocomposite microsphere platform was developed for selective intra-arterial transcatheter drug delivery to liver tumors. In our study, continuous microfluidic methods were used to fabricate drug-loaded multimodal MRI/CT visible microspheres that included both gold nanorods and magnetic clusters. The resulting hydrophilic, deformable, and non-aggregated microspheres were mono-disperse and roughly 25 um in size. Sustained drug release and strong MRI T2 and CT contrast effects were achieved with the embedded magnetic nano-clusters and radiopaque gold nanorods. The microspheres were successfully infused through catheters selectively placed within the hepatic artery in rodent models and subsequent distribution in the targeted liver tissues and hepatic tumors confirmed with MRI and CT imaging. These multimodal nanocomposite drug carriers should be ideal for selective intra-arterial catheter-directed administration to liver tumors while permitting MRI/CT visualization for patient-specific confirmation of tumor-targeted delivery.


Radiology | 2016

SPIO-labeled Yttrium Microspheres for MR Imaging Quantification of Transcatheter Intrahepatic Delivery in a Rodent Model.

Weiguo Li; Zhuoli Zhang; Andrew C. Gordon; Jeane Chen; Jodi Nicolai; Robert J. Lewandowski; Reed A. Omary; Andrew C. Larson

PURPOSE To investigate the qualitative and quantitative impacts of labeling yttrium microspheres with increasing amounts of superparamagnetic iron oxide (SPIO) material for magnetic resonance (MR) imaging in phantom and rodent models. MATERIALS AND METHODS Animal model studies were approved by the institutional Animal Care and Use Committee. The r2* relaxivity for each of four microsphere SPIO compositions was determined from 32 phantoms constructed with agarose gel and in eight concentrations from each of the four compositions. Intrahepatic transcatheter infusion procedures were performed in rats by using each of the four compositions before MR imaging to visualize distributions within the liver. For quantitative studies, doses of 5, 10, 15, or 20 mg 2% SPIO-labeled yttrium microspheres were infused into 24 rats (six rats per group). MR imaging R2* measurements were used to quantify the dose delivered to each liver. Pearson correlation, analysis of variance, and intraclass correlation analyses were performed to compare MR imaging measurements in phantoms and animal models. RESULTS Increased r2* relaxivity was observed with incremental increases of SPIO microsphere content. R2* measurements of the 2% SPIO-labeled yttrium microsphere concentration were well correlated with known phantom concentrations (R(2) = 1.00, P < .001) over a broader linear range than observed for the other three compositions. Microspheres were heterogeneously distributed within each liver; increasing microsphere SPIO content produced marked signal voids. R2*-based measurements of 2% SPIO-labeled yttrium microsphere delivery were well correlated with infused dose (intraclass correlation coefficient, 0.98; P < .001). CONCLUSION MR imaging R2* measurements of yttrium microspheres labeled with 2% SPIO can quantitatively depict in vivo intrahepatic biodistribution in a rat model.


Radiology | 2017

Biofunctionalized Hybrid Magnetic Gold Nanoparticles as Catalysts for Photothermal Ablation of Colorectal Liver Metastases

Sarah B. White; Dong Hyun Kim; Yang Guo; Weiguo Li; Yihe Yang; Jeane Chen; Venkateswara Rao Gogineni; Andrew C. Larson

Purpose To demonstrate that anti-MG1 conjugated hybrid magnetic gold nanoparticles (HNPs) act as a catalyst during photothermal ablation (PTA) of colorectal liver metastases, and thus increase ablation zones. Materials and Methods All experiments were performed with approval of the institutional animal care and use committee. Therapeutic and diagnostic multifunctional HNPs conjugated with anti-MG1 monoclonal antibodies were synthesized, and the coupling efficiency was determined. Livers of 19 Wistar rats were implanted with 5 × 106 rat colorectal liver metastasis cell line cells. The rats were divided into three groups according to injection: anti-MG1-coupled HNPs (n = 6), HNPs only (n = 6), and cells only (control group, n = 7). Voxel-wise R2 and R2* magnetic resonance (MR) imaging measurements were obtained before, immediately after, and 24 hours after injection. PTA was then performed with a fiber-coupled near-infrared (808 nm) diode laser with laser power of 0.56 W/cm2 for 3 minutes, while temperature changes were measured. Tumors were assessed for necrosis with hematoxylin-eosin staining. Organs were analyzed with inductively coupled plasma mass spectrometry to assess biodistribution. Therapeutic efficacy and tumor necrosis area were compared by using a one-way analysis of variance with post hoc analysis for statistically significant differences. Results The coupling efficiency was 22 μg/mg (55%). Significant differences were found between preinfusion and 24-hour postinfusion measurements of both T2 (repeated measures analysis of variance, P = .025) and T2* (P < .001). Significant differences also existed for T2* measurements between the anti-MG1 HNP and HNP-only groups (P = .034). Mean temperature ± standard deviation with PTA in the anti-MG1-coated HNP, HNP, and control groups was 50.2°C ± 7.8, 51°C ± 4.4, and 39.5°C ± 2.0, respectively. Inductively coupled plasma mass spectrometry revealed significant tumor targeting and splenic sequestration. Mean percentages of tumor necrosis in the anti-MG1-coated HNP, HNP, and control groups were 38% ± 29, 14% ± 17, and 7% ± 8, respectively (P = .043). Conclusion Targeted monoclonal antibody-conjugated HNPs can serve as a catalyst for photothermal ablation of colorectal liver metastases by increasing ablation zones.


PLOS ONE | 2015

Percutaneous ultrasound guided implantation of VX2 for creation of a rabbit hepatic tumor model

Sarah B. White; Jeane Chen; Andrew C. Gordon; Kathleen R. Harris; Jodi Nicolai; Derek L. West; Andrew C. Larson

Creation of a VX2 tumor model has traditionally required a laparotomy and surgical implantation of tumor fragments. Open surgical procedures are invasive and require long procedure times and recovery that can result in post-operative morbidity and mortality. The purpose of this study is to report the results of a percutaneous ultrasound guided method for creation of a VX2 model in rabbit livers. A total of 27 New Zealand white rabbits underwent a percutaneous ultrasound guided approach, where a VX2 tumor fragment was implanted in the liver. Magnetic resonance imaging was used to assess for tumor growth and necropsy was performed to determine rates of tract seeding and metastatic disease. Ultrasound guided tumor implantation was successful in all 27 rabbits. One rabbit died 2 days following the implantation procedure. Two rabbits had no tumors seen on follow-up imaging. Therefore, tumor development was seen in 24/26 (92%) rabbits. During the follow-up period, tract seeding was seen in 8% of rabbits and 38% had extra-hepatic metastatic disease. Therefore, percutaneous ultrasound guided tumor implantation safely provides reliable tumor growth for establishing hepatic VX2 tumors in a rabbit model with decreased rates of tract seeding, compared to previously reported methods.


PLOS ONE | 2016

Characterization of CC-531 as a Rat Model of Colorectal Liver Metastases

Sarah B. White; Daniele Procissi; Jeane Chen; Venkateswara Rao Gogineni; Patrick D. Tyler; Yihe Yang; Reed A. Omary; Andrew C. Larson

Purpose Surgical resection of colorectal liver metastases is not achievable in more than 70% of the cases. Although the liver directed therapies have become a part of the stand of care, lack of a preclinical model impedes the assessment of toxicity and therapeutic benefits attributed several candidate drugs or treatment regimens that can be designed. In the present study we aim develop and characterize a rat colorectal liver metastasis model. Materials and Methods Growth characteristics of CC-531 cells were determined in vitro followed by subcapsular liver implantation in syngeneic WAG/Rij rats. Tumor growth progression was followed over 3 weeks by ultrasound (US) and magnetic resonance imaging (MRI). Growth characteristics were also assessed by histopathology and immunohistochemistry in harvested tumor tissues. Results The doubling time of CC-531 cells was found be under 24hrs and all the implanted rats grew tumors. US imaging showed hypoechoic masses and MRI showed contrast enhancement representing complex tumor microenvironments. Hematoxylin and Eosin staining confirmed tumor growth and uniform CD31 staining in tumor confirmed even vessel density. Conclusion CC-531 can be used as a metastatic rat tumor colorectal liver metastases model with well-defined characteristics that can be readily followed by imaging whilst having a therapeutic window for interventions.


Cancer Research | 2012

Abstract 5695: PLGA microspheres for MRI-guided localized transcatheter delivery of sorafenib: development and preclinical feasibility studies

Jeane Chen; Alex Sheu; Weiguo Li; Reed A. Omary; Lonnie D. Shea; Andrew C. Larson

Purpose: Sorafenib is a multi-kinase inhibitor with efficacy for the palliative treatment of hepatocellular carcinoma (HCC). However, oral administration limits the dose that ultimately reaches tumors and many patients cannot tolerate associated systemic toxicities such as hand-foot syndrome and cardiac ischemia. Local delivery of sorafenib-eluting microspheres via intra-arterial transcather infusion should increase the dose supplied to tumors while limiting the harsh side-effects. The purpose of this study was to design and characterize sorafenib-eluting microspheres that also permit MRI of the intra-hepatic biodistribution post-infusion. Methods: Polylactide-co-glycolide (PLGA) microspheres encapsulating sorafenib and an iron-oxide ferrofluid were synthesized via a double emulsion solvent evaporation method. Microsphere encapsulation properties were characterized using high performance liquid chromatography (HPLC) for sorafenib encapsulation, and inductively coupled plasma mass spectrometry (ICP-MS) for ferrofluid encapsulation. A cell viability study was performed to investigate the therapeutic efficacy of the microspheres (cell proliferation in McA-RH7777 HCC cell line). An in vitro imaging study was then performed where the microspheres were embedded in agar gels and imaged with MRI. Lastly, six Sprague Dawley rats were catheterized and infused with 4-6 mgs of the microspheres via the proper hepatic artery and imaged with MRI to visualize the biodistribution of the microspheres. Prussian blue staining was used to identify microsphere positions within hepatic tissue specimens at necropsy. The agar phantom and rat MRI studies were performed using a 7T Bruker Clinscan system. Results: The characterization studies indicated that the Sorafenib-eluting PLGA microspheres (roughly 10 microns in diameter) included 16.4% (w/w) sorafenib and 0.81% (w/w) ferrofluid. Cell viability studies showed an 84% decrease in cell number compared to the control (50% compared to the initial amount plated) after treatment of the McA-RH7777 cells with the microspheres at a concentration of 36.6 μg/mL for 72 hours. MRI phantom studies demonstrated that T2* relaxation times decreased from 6.9 ms −1 to 2.8 ms −1 when microsphere concentrations increased from 0 to 2 mg/mL. For rat studies, the intra-hepatic distribution of the microspheres was clearly depicted in follow-up T2-weighted MRI images. Prussian blue slides confirmed hepatic microsphere delivery. Conclusion: The designed PLGA microspheres with sorafenib and ferrofluid permitted transcatheter delivery to rodent livers and MRI visualization of the resulting intra-hepatic biodistribution. This design allows for local delivery that may ultimately reduce systemic toxicities and increase therapeutic efficacy of sorafenib and other similar MKIs. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 5695. doi:1538-7445.AM2012-5695

Collaboration


Dive into the Jeane Chen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sarah B. White

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Weiguo Li

Northwestern University

View shared research outputs
Top Co-Authors

Avatar

Jodi Nicolai

Northwestern University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

A. Sheu

Northwestern University

View shared research outputs
Researchain Logo
Decentralizing Knowledge