Network


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

Hotspot


Dive into the research topics where Yi-Ju Ho is active.

Publication


Featured researches published by Yi-Ju Ho.


Theranostics | 2016

Ultrasound/Magnetic Targeting with SPIO-DOX-Microbubble Complex for Image-Guided Drug Delivery in Brain Tumors

Ching-Hsiang Fan; Yu-Hang Cheng; Chien-Yu Ting; Yi-Ju Ho; Po-Hung Hsu; Hao-Li Liu; Chih-Kuang Yeh

One of the greatest challenges in the deployment of chemotherapeutic drugs against brain tumors is ensuring that sufficient drug concentrations reach the tumor, while minimizing drug accumulation at undesired sites. Recently, injection of therapeutic agents following blood-brain barrier (BBB) opening by focused ultrasound (FUS) with microbubbles (MBs) has been shown to enhance drug delivery in targeted brain regions. Nevertheless, the distribution and quantitative deposition of agents delivered to the brain are still hard to estimate. Based on our previous work on superparamagnetic iron oxide (SPIO)-loaded MBs, we present a novel theranostic complex of SPIO-Doxorubicin (DOX)-conjugated MB (SD-MB) for drug delivery to the brain. Magnetic labeling of the drug enables direct visualization via magnetic resonance imaging, and also facilitates magnetic targeting (MT) to actively enhance targeted deposition of the drug. In a rat glioma model, we demonstrated that FUS sonication can be used with SD-MBs to simultaneously facilitate BBB opening and allow dual ultrasound/magnetic targeting of chemotherapeutic agent (DOX) delivery. The accumulation of SD complex within brain tumors can be significantly enhanced by MT (25.7 fold of DOX, 7.6 fold of SPIO). The change in relaxation rate R2 (1/T2) within tumors was highly correlated with SD deposition as quantified by high performance liquid chromatography (R2 = 0.93) and inductively coupled plasma-atomic emission spectroscopy (R2 = 0.94), demonstrating real-time monitoring of DOX distribution. Our results suggest that SD-MBs can serve as multifunction agents to achieve advanced molecular theranostics.


Acta Biomaterialia | 2017

Concurrent anti-vascular therapy and chemotherapy in solid tumors using drug-loaded acoustic nanodroplet vaporization.

Yi-Ju Ho; Chih-Kuang Yeh

Drug-loaded nanodroplets (NDs) can be converted into gas bubbles through ultrasound (US) stimulation, termed acoustic droplet vaporization (ADV), which provides a potential strategy to simultaneously induce vascular disruption and release drugs for combined physical anti-vascular therapy and chemotherapy. Doxorubicin-loaded NDs (DOX-NDs) with a mean size of 214nm containing 2.48mg DOX/mL were used in this study. High-speed images displayed bubble formation and cell debris, demonstrating the reduction in cell viability after ADV. Intravital imaging provided direct visualization of disrupted tumor vessels (vessel size <30μm), the extravasation distance was 12μm in the DOX-NDs group and increased over 100μm in the DOX-NDs+US group. Solid tumor perfusion on US imaging was significantly reduced to 23% after DOX-NDs vaporization, but gradually recovered to 41%, especially at the tumor periphery after 24h. Histological images of the DOX-NDs+US group revealed tissue necrosis, a large amount of drug extravasation, vascular disruption, and immune cell infiltration at the tumor center. Tumor sizes decreased 22%, 36%, and 68% for NDs+US, DOX-NDs, and DOX-NDs+US, respectively, to prolong the survival of tumor-bearing mice. Therefore, this study demonstrates that the combination of physical anti-vascular therapy and chemotherapy with DOX-NDs vaporization promotes uniform treatment to improve therapeutic efficacy. STATEMENT OF SIGNIFICANCE Tumor vasculature plays an important role for tumor cell proliferation by transporting oxygen and nutrients. Previous studies combined anti-vascular therapy and drug release to inhibit tumor growth by ultrasound-stimulated microbubble destruction or acoustic droplet vaporization. Although the efficacy of combined therapy has been demonstrated; the relative spatial distribution of vascular disruption, drug delivery, and accompanied immune responses within solid tumors was not discussed clearly. Herein, our study used drug-loaded nanodroplets to combined physical anti-vascular and chemical therapy. The in vitro cytotoxicity, intravital imaging, and histological assessment were used to evaluate the temporal and spatial cooperation between physical and chemical effect. These results revealed some evidences for complementary action to explain the high efficacy of tumor inhibition by combined therapy.


Theranostics | 2015

Biomimetic Acoustically-Responsive Vesicles for Theranostic Applications

Chen-Chan Hsieh; Shih-Tsung Kang; Yee-Hsien Lin; Yi-Ju Ho; Chung-Hsin Wang; Chih-Kuang Yeh; Chien-Wen Chang

In recent years, biomimetic cell membrane-derived particles have emerged as a new class of drug delivery system with advantages of biocompatibility, ease of isolation and long circulation profile. Here we report the development and potential theranostic applications of a new biomimetic acoustically-responsive droplet system derived from mammalian red blood cell membrane (RBCM). We hypothesized that drug-loaded RBCM droplets (RBCMDs) would undergo a transition from liquid (droplets) to gas (bubbles) upon high intensity focused ultrasound (HIFU) insonation, resulting in on-demand drug release. The generated microbubbles could also serve as a contrast agent to enhance ultrasound imaging. As-synthesized RBCMDs exhibited uniform size, good dispersity and preservation of RBCM-associated proteins that prevented uptake by macrophages. Camptothecin (CPT), an anti-cancer drug, was successfully loaded in the RBCMDs with a loading efficiency of 2-3% and an encapsulation efficiency of 62-97%. A short (3 min) exposure to HIFU irradiation triggered release of CPT from the RBCMDs and the physical explosion of droplets damaged nearby cancer cells resulting in significant cell death. In addition, the acoustically vaporized RBCMDs significantly increased the ultrasound echo signal to 30 dB. Lastly, we demonstrated that RBCMDs could be acoustically vaporized in vivo in target tissues, and enhancing ultrasound imaging. Taken together, we have developed a new class of naturally derived RBCMDs which show great potential for future application in remotely triggered drug delivery and ultrasound imaging enhancement.


Theranostics | 2017

Theranostic Performance of Acoustic Nanodroplet Vaporization-Generated Bubbles in Tumor Intertissue

Yi-Ju Ho; Chih-Kuang Yeh

Solid tumors with poorly perfused regions reveal some of the treatment limitations that restrict drug delivery and therapeutic efficacy. Acoustic droplet vaporization (ADV) has been applied to directly disrupt vessels and release nanodroplets, ADV-generated bubbles (ADV-Bs), and drugs into tumor tissue. In this study, we investigated the in vivo behavior of ADV-Bs stimulated by US, and evaluated the possibility of moving intertissue ADV-Bs into the poorly perfused regions of solid tumors. Intravital imaging revealed intertissue ADV-B formation, movement, and cavitation triggered by US, where the distance of intertissue ADV-B movement was 33-99 µm per pulse. When ADV-Bs were applied to tumor cells, the cell membrane was damaged, increasing cellular permeability or inducing cell death. The poorly perfused regions within solid tumors show enhancement due to ADV-B accumulation after application of US-triggered ADV-B. The intratumoral nanodroplet or ADV-B distribution around the poorly perfused regions with tumor necrosis or hypoxia were demonstrated by histological assessment. ADV-B formation, movement and cavitation could induce cell membrane damage by mechanical force providing a mechanism to overcome treatment limitations in poorly perfused regions of tumors.


Drug Discovery Today | 2017

Current progress in antivascular tumor therapy

Yi-Ju Ho; Tzu-Chia Wang; Ching-Hsiang Fan; Chih-Kuang Yeh

The tumor vasculature transports oxygen, nutrients and drugs for crucial roles in tumor therapy. Antivascular therapy directly targets existing tumor vessels to reduce blood perfusion and then inhibit tumor growth. Vascular disrupting agents and ultrasound-stimulated microbubble destruction use chemical toxicity and physical effect, respectively, to damage vascular endothelial cells for antivascular therapy. Moreover, antivascular therapy can break vessel wall barriers and change the tumor microenvironment to compensate for the limitations of conventional chemotherapy or radiotherapy. This review presents current progress and an overview of antivascular therapy, which can inform the development and application in cancer research.


Ultrasound in Medicine and Biology | 2018

Macrophages as Drug Delivery Carriers for Acoustic Phase-Change Droplets

Ching-Hsiang Fan; Ya-Hsuan Lee; Yi-Ju Ho; Chung-Hsin Wang; Shih-Tsung Kang; Chih-Kuang Yeh

The major challenges in treating malignant tumors are transport of therapeutic agents to hypoxic regions and real-time assessment of successful drug release via medical imaging modalities. In this study, we propose the use of macrophages (RAW 264.7 cells) as carriers of drug-loaded phase-change droplets to penetrate ischemic or hypoxic regions within tumors. The droplets consist of perfluoropentane, lipid and the chemotherapeutic drug doxorubicin (DOX, DOX-droplets). The efficiency of DOX-droplet uptake, migration mobility and viability of DOX-droplet-loaded macrophages (DLMs) were measured using a transmembrane cell migration assay, the alamarBlue assay and flow cytometric analysis, respectively. Our results indicate the feasibility of utilizing macrophages as DOX-droplet carriers (DOX payload of DOX-droplets: 459.3 ± 35.8 µg/mL, efficiency of cell uptake DOX-droplets: 88.8 ± 3.5%). The migration mobility (total number of migrated microphages) of DLMs decreased to 32.3% compared with that of healthy macrophages, but the DLMs provided contrast-enhanced ultrasound imaging (1.7-fold enhancement) and anti-tumor effect (70.9% cell viability) after acoustic droplet vaporization, suggesting the potential theranostic applications of DLMs. Future work will assess the tumor penetration ability of DLMs, mechanical effect of droplet vaporization on in vivo anti-tumor therapy and the release of the carried drug by ultrasound-triggered vaporization.


Journal of Controlled Release | 2018

Thermal-sensitive acoustic droplets for dual-mode ultrasound imaging and drug delivery

Yi-Ju Ho; Chin-Chou Wu; Zong-Han Hsieh; Ching-Hsiang Fan; Chih-Kuang Yeh

ABSTRACT Mild hyperthermia (40–43°C) could assist with acoustic droplet vaporization (ADV) via focused ultrasound heating and enable ultrasound thermal imaging to monitor the heating position. We explored the possibility of predicting the treatment position of ADV using thermal‐sensitive droplets (TSDs) and ultrasound thermal imaging. The TSDs were created with an encapsulated mixture of C5F12 and C6F14 that could be vaporized by ultrasound under mild hyperthermia. The ultrasound imaging‐guided high‐intensity focused ultrasound system was used to collect ultrasound images, heat tumors, and vaporize the TSDs. The overlap between the location of heating and ADV‐induced bubble formation was used to evaluate the accuracy of predicting the treatment position. The optimal fabrication of TSDs (1.21±0.19&mgr;m, volume ratio of C5F12:C6F14=7:3) increased ADV efficiency by 33±11% at 41°C under an acoustic pressure of 8.6MPa. The accuracy of ADV region prediction by ultrasound thermal imaging was 87.2±3.5% for the in vitro study and 83.2±8.6% for the in vivo study. The similarity among the location of bubble enhancement and distribution of 41°C areas demonstrated the credibility of our estimates. Therefore, in this study, we validated the feasibility of applying TSDs and ultrasound thermal imaging to predict the in vivo treatment position of ADV


Journal of Controlled Release | 2018

Camptothecin-loaded fusogenic nanodroplets as ultrasound theranostic agent in stem cell-mediated drug-delivery system

Yi-Ju Ho; Yu-Jung Chiang; Shih-Tsung Kang; Ching-Hsiang Fan; Chih-Kuang Yeh

ABSTRACT Adipose‐derived stem cells (ADSCs) have been utilized in cellular delivery systems to carry therapeutic agents into tumors by migration. Drug‐loaded nanodroplets release drugs and form bubbles after acoustic droplet vaporization (ADV) triggered by ultrasound stimulation, providing a system for ultrasound‐induced cellular delivery of theranostic agents. In order to improve the efficiency of drug release, fusogenic nanodroplets were designed to go from nano to micron size upon uptake by ADSCs for reducing ADV threshold. The purpose of our study was to demonstrate the utility of camptothecin‐loaded fusogenic nanodroplets (CPT‐FNDs) as ultrasound theranostic agents in an ADSCs delivery system. CPT‐FNDs showed an increase in size from 81.6±3.5 to 1043.5±28.3nm and improved CPT release from 22.0±1.8% to 37.6±2.1%, demonstrating the fusion ability of CPT‐FNDs. CPT‐FNDs‐loaded ADSCs demonstrated a cell viability of 77±4%, and the in vitro migration ability was 3.2±1.2‐fold for the tumor condition compared to the cell growth condition. Ultrasound enhancement imaging showed intratumoral ADV‐generated bubble formation (increasing 3.24±0.47dB) triggered by ultrasound after CPT‐FNDs‐loaded ADSCs migration into B16F0 tumors. Histological images revealed intratumoral distribution of CPT‐FNDs‐loaded ADSCs and tissue damage due to the ADV. The CPT‐FNDs can be used as theranostic agents in an ADSCs delivery system to provide the ultrasound contrast imaging and deliver combination therapy of drug release and physical damage after ADV.


ACS Applied Materials & Interfaces | 2018

Spatially Uniform Tumor Treatment and Drug Penetration by Regulating Ultrasound with Microbubbles

Yi-Ju Ho; Tzu-Chia Wang; Ching-Hsiang Fan; Chih-Kuang Yeh

Tumor microenvironment has different morphologies of vessels in the core and rim regions, which influences the efficacy of tumor therapy. Our study proposed to improve the spatial uniformity of the antivascular effect and drug penetration within the tumor core and rim in combination therapies by regulating ultrasound-stimulated microbubble destruction (USMD). Focused ultrasound at 2 MHz and lipid-shell microbubbles (1.12 ± 0.08 μm, mean ± standard deviation) were used to perform USMD. The efficiency of the antivascular effect was evaluated by intravital imaging to determine the optimal USMD parameters. Tumor perfusion and histological alterations in the tumor core and rim were used to analyze the spatial uniformity of the antivascular effect and liposomal-doxorubicin (5 mg/kg) penetration in the combination therapy. Tumor vessels of specific sizes were disrupted by regulating USMD: vessels with sizes of 11 ± 3, 14 ± 5, 19 ± 7, and 23 ± 10 μm were disrupted by stimulation at acoustic pressures of 3, 5, 7, and 9 MPa, respectively (each p < 0.05). The effective treatment time of USMD (at 2 × 107 microbubbles/mouse, 7 MPa, and three cycles) was 60-120 min, which resulted in the disruption of 21-44% of vessels smaller than 50 μm. The reductions in perfusion and vascular density after combination therapy did not differ significantly between the tumor core and rim. This study found that regulating USMD can result in homogeneous antivascular effects and drug penetration within tumors and thereby improve the efficacy of combination therapies.


internaltional ultrasonics symposium | 2017

Bioeffects of acoustic droplet vaporization-generated bubbles in tissue

Yi-Ju Ho; Yi-Tim Lin; Chih-Kuang Yeh

Acoustic droplet vaporization (ADV) is a process when a droplet is converted into a gaseous bubble under ultrasound (US) stimulation. Our previous study has shown that the mechanical force generated by ADV can release drugs and disrupt vessels. It would result in the streaming intact droplets leaking into tissue. In addition, the intact droplets may occur ADV process by subsequent US stimulation. Therefore, this study explored the bioeffects between ADV-Bs and cells within tissue.

Collaboration


Dive into the Yi-Ju Ho's collaboration.

Top Co-Authors

Avatar

Chih-Kuang Yeh

National Tsing Hua University

View shared research outputs
Top Co-Authors

Avatar

Ching-Hsiang Fan

National Tsing Hua University

View shared research outputs
Top Co-Authors

Avatar

Shih-Tsung Kang

National Tsing Hua University

View shared research outputs
Top Co-Authors

Avatar

Chien-Yu Ting

National Tsing Hua University

View shared research outputs
Top Co-Authors

Avatar

Chung-Hsin Wang

National Tsing Hua University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tzu-Chia Wang

National Tsing Hua University

View shared research outputs
Top Co-Authors

Avatar

Yu-Hang Cheng

National Tsing Hua University

View shared research outputs
Top Co-Authors

Avatar

Chen-Chan Hsieh

National Tsing Hua University

View shared research outputs
Researchain Logo
Decentralizing Knowledge