Network


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

Hotspot


Dive into the research topics where Xiaoding Xu is active.

Publication


Featured researches published by Xiaoding Xu.


Angewandte Chemie | 2016

Ultra-pH-Responsive and Tumor-Penetrating Nanoplatform for Targeted siRNA Delivery with Robust Anti-Cancer Efficacy

Xiaoding Xu; Jun Wu; Yanlan Liu; Mikyung Yu; Lili Zhao; Xi Zhu; Sushant Bhasin; Qing Li; Emily Ha; Jinjun Shi; Omid C. Farokhzad

RNA interference (RNAi) gene silencing technologies have shown significant potential for treating various diseases, including cancer. However, clinical success in cancer therapy remains elusive, mainly owing to suboptimal in vivo delivery of RNAi therapeutics such as small interference RNA (siRNA) to tumors. Herein, we developed a library of polymers that respond to a narrow pH change (ultra-pH-responsive), and demonstrated the utility of these materials in targeted and deep tumor-penetrating nanoparticle (NP) for in vivo RNAi. The new NP platform is mainly composed of the following key components: i) internalizing RGD (iRGD) to enhance tumor targeting and tissue penetration; ii) polyethylene glycol (PEG) chains to prolong blood circulation; and iii) sharp pH-responsive hydrophobic polymer to improve endosome escape. Through systematic studies of structure-function relationship, the optimized RNAi NPs (<70 nm) showed efficient gene silencing and significant inhibition of tumor growth with negligible toxicities in vivo.


Angewandte Chemie | 2017

Antimonene Quantum Dots: Synthesis and Application as Near‐Infrared Photothermal Agents for Effective Cancer Therapy

Wei Tao; Xiaoyuan Ji; Xiaoding Xu; Mohammad Ariful Islam; Zhongjun Li; Si Chen; Phei Er Saw; Han Zhang; Zameer Bharwani; Zilei Guo; Jinjun Shi; Omid C. Farokhzad

Photothermal therapy (PTT) has shown significant potential for cancer therapy. However, developing nanomaterials (NMs)-based photothermal agents (PTAs) with satisfactory photothermal conversion efficacy (PTCE) and biocompatibility remains a key challenge. Herein, a new generation of PTAs based on two-dimensional (2D) antimonene quantum dots (AMQDs) was developed by a novel liquid exfoliation method. Surface modification of AMQDs with polyethylene glycol (PEG) significantly enhanced both biocompatibility and stability in physiological medium. The PEG-coated AMQDs showed a PTCE of 45.5 %, which is higher than many other NMs-based PTAs such as graphene, Au, MoS2 , and black phosphorus (BP). The AMQDs-based PTAs also exhibited a unique feature of NIR-induced rapid degradability. Through both in vitro and in vivo studies, the PEG-coated AMQDs demonstrated notable NIR-induced tumor ablation ability. This work is expected to expand the utility of 2D antimonene (AM) to biomedical applications through the development of an entirely novel PTA platform.


Advanced Materials | 2017

ROS-Responsive Polyprodrug Nanoparticles for Triggered Drug Delivery and Effective Cancer Therapy

Xiaoding Xu; Phei Er Saw; Wei Tao; Yujing Li; Xiaoyuan Ji; Sushant Bhasin; Yanlan Liu; Dana Ayyash; Jonathan Rasmussen; Marc Huo; Jinjun Shi; Omid C. Farokhzad

The application of nanoparticles (NPs) to drug delivery has led to the development of novel nanotherapeutics for the treatment of various diseases including cancer. However, clinical use of NP-mediated drug delivery has not always translated into improved survival of cancer patients, in part due to the suboptimal properties of NP platforms, such as premature drug leakage during preparation, storage, or blood circulation, lack of active targeting to tumor tissue and cells, and poor tissue penetration. Herein, an innovative reactive oxygen species (ROS)-responsive polyprodrug is reported that can self-assemble into stable NPs with high drug loading. This new NP platform is composed of the following key components: (i) polyprodrug inner core that can respond to ROS for triggered release of intact therapeutic molecules, (ii) polyethylene glycol (PEG) outer shell to prolong blood circulation; and (iii) surface-encoded internalizing RGD (iRGD) to enhance tumor targeting and tissue penetration. These targeted ROS-responsive polyprodrug NPs show significant inhibition of tumor cell growth both in vitro and in vivo.


ACS Nano | 2017

Multifunctional Envelope-Type siRNA Delivery Nanoparticle Platform for Prostate Cancer Therapy

Xiaoding Xu; Jun Wu; Yanlan Liu; Phei Er Saw; Wei Tao; Mikyung Yu; Harshal Zope; Michelle Si; Amanda Victorious; Jonathan Rasmussen; Dana Ayyash; Omid C. Farokhzad; Jinjun Shi

With the capability of specific silencing of target gene expression, RNA interference (RNAi) technology is emerging as a promising therapeutic modality for the treatment of cancer and other diseases. One key challenge for the clinical applications of RNAi is the safe and effective delivery of RNAi agents such as small interfering RNA (siRNA) to a particular nonliver diseased tissue (e.g., tumor) and cell type with sufficient cytosolic transport. In this work, we proposed a multifunctional envelope-type nanoparticle (NP) platform for prostate cancer (PCa)-specific in vivo siRNA delivery. A library of oligoarginine-functionalized and sharp pH-responsive polymers was synthesized and used for self-assembly with siRNA into NPs with the features of long blood circulation and pH-triggered oligoarginine-mediated endosomal membrane penetration. By further modification with ACUPA, a small molecular ligand specifically recognizing prostate-specific membrane antigen (PSMA) receptor, this envelope-type nanoplatform with multifunctional properties can efficiently target PSMA-expressing PCa cells and silence target gene expression. Systemic delivery of the siRNA NPs can efficiently silence the expression of prohibitin 1 (PHB1), which is upregulated in PCa and other cancers, and significantly inhibit PCa tumor growth. These results suggest that this multifunctional envelope-type nanoplatform could become an effective tool for PCa-specific therapy.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Theranostic near-infrared fluorescent nanoplatform for imaging and systemic siRNA delivery to metastatic anaplastic thyroid cancer

Yanlan Liu; Viswanath Gunda; Xi Zhu; Xiaoding Xu; Jun Wu; Diana Askhatova; Omid C. Farokhzad; Sareh Parangi; Jinjun Shi

Significance Current therapies have shown limited success in improving outcomes for lethal anaplastic thyroid cancer (ATC) characterized by rapid tumor growth and metastatic dissemination caused by the activation of genetic mutations. RNAi nanotechnology is emerging as a promising strategy for effectively treating such cancers and suppressing metastasis. However, suboptimal systemic delivery of RNAi agents to tumors and variable therapeutic responses because of tumor heterogeneity represent challenging hurdles to widespread clinical use. We describe an innovative near-infrared nanoplatform for systemic delivery of siRNA to ATC and real-time tracking of tumor accumulation. Antitumor growth and antimetastasis effects in an orthotopic ATC mouse model suggest this nanoplatform as a valuable tool for personalized treatment of ATC and other advanced malignancies. Anaplastic thyroid cancer (ATC), one of the most aggressive solid tumors, is characterized by rapid tumor growth and severe metastasis to other organs. Owing to the lack of effective treatment options, ATC has a mortality rate of ∼100% and median survival of less than 5 months. RNAi nanotechnology represents a promising strategy for cancer therapy through nanoparticle (NP) -mediated delivery of RNAi agents (e.g., siRNA) to solid tumors for specific silencing of target genes driving growth and/or metastasis. Nevertheless, the clinical success of RNAi cancer nanotherapies remains elusive in large part because of the suboptimal systemic siRNA NP delivery to tumors and the fact that tumor heterogeneity produces variable NP accumulation and thus, therapeutic response. To address these challenges, we here present an innovative theranostic NP platform composed of a near-infrared (NIR) fluorescent polymer for effective in vivo siRNA delivery to ATC tumors and simultaneous tracking of the tumor accumulation by noninvasive NIR imaging. The NIR polymeric NPs are small (∼50 nm), show long blood circulation and high tumor accumulation, and facilitate tumor imaging. Systemic siRNA delivery using these NPs efficiently silences the expression of V-Raf murine sarcoma viral oncogene homolog B (BRAF) in tumor tissues and significantly suppresses tumor growth and metastasis in an orthotopic mouse model of ATC. These results suggest that this theranostic NP system could become an effective tool for NIR imaging-guided siRNA delivery for personalized treatment of advanced malignancies.


Nano Letters | 2017

Tumor Microenvironment-Responsive Multistaged Nanoplatform for Systemic RNAi and Cancer Therapy

Xiaoding Xu; Phei Er Saw; Wei Tao; Yujing Li; Xiaoyuan Ji; Mikyung Yu; Morteza Mahmoudi; Jonathan Rasmussen; Dana Ayyash; Yuxiao Zhou; Omid C. Farokhzad; Jinjun Shi

While RNA interference (RNAi) therapy has demonstrated significant potential for cancer treatment, the effective and safe systemic delivery of RNAi agents such as small interfering RNA (siRNA) into tumor cells in vivo remains challenging. We herein reported a unique multistaged siRNA delivery nanoparticle (NP) platform, which is comprised of (i) a polyethylene glycol (PEG) surface shell, (ii) a sharp tumor microenvironment (TME) pH-responsive polymer that forms the NP core, and (iii) charge-mediated complexes of siRNA and tumor cell-targeting- and penetrating-peptide-amphiphile (TCPA) that are encapsulated in the NP core. When the rationally designed, long circulating polymeric NPs accumulate in tumor tissues after intravenous administration, the targeted siRNA-TCPA complexes can be rapidly released via TME pH-mediated NP disassembly for subsequent specific targeting of tumor cells and cytosolic transport, thus achieving efficient gene silencing. In vivo results further demonstrate that the multistaged NP delivery of siRNA against bromodomain 4 (BRD4), a recently discovered target protein that regulates the development and progression of prostate cancer (PCa), can significantly inhibit PCa tumor growth.


ACS Nano | 2018

Intracellular Mechanistic Understanding of 2D MoS2 Nanosheets for Anti-Exocytosis-Enhanced Synergistic Cancer Therapy

Xianbing Zhu; Xiaoyuan Ji; Na Kong; Yunhan Chen; Morteza Mahmoudi; Xiaoding Xu; Li Ding; Wei Tao; Ting Cai; Yujing Li; Tian Gan; Austin Barrett; Zameer Bharwani; Hongbo Chen; Omid C. Farokhzad

Emerging two-dimensional (2D) nanomaterials, such as transition-metal dichalcogenide (TMD) nanosheets (NSs), have shown tremendous potential for use in a wide variety of fields including cancer nanomedicine. The interaction of nanomaterials with biosystems is of critical importance for their safe and efficient application. However, a cellular-level understanding of the nano-bio interactions of these emerging 2D nanomaterials ( i. e., intracellular mechanisms) remains elusive. Here we chose molybdenum disulfide (MoS2) NSs as representative 2D nanomaterials to gain a better understanding of their intracellular mechanisms of action in cancer cells, which play a significant role in both their fate and efficacy. MoS2 NSs were found to be internalized through three pathways: clathrin → early endosomes → lysosomes, caveolae → early endosomes → lysosomes, and macropinocytosis → late endosomes → lysosomes. We also observed autophagy-mediated accumulation in the lysosomes and exocytosis-induced efflux of MoS2 NSs. Based on these findings, we developed a strategy to achieve effective and synergistic in vivo cancer therapy with MoS2 NSs loaded with low doses of drug through inhibiting exocytosis pathway-induced loss. To the best of our knowledge, this is the first systematic experimental report on the nano-bio interaction of 2D nanomaterials in cells and their application for anti-exocytosis-enhanced synergistic cancer therapy.


Advanced Healthcare Materials | 2017

Targeted Nanotherapeutics Encapsulating Liver X Receptor Agonist GW3965 Enhance Antiatherogenic Effects without Adverse Effects on Hepatic Lipid Metabolism in Ldlr−/− Mice

Mikyung Yu; Jaume Amengual; Arjun Menon; Nazila Kamaly; Felix Zhou; Xiaoding Xu; Phei Er Saw; Seung Joo Lee; Kevin Si; Carleena Angelica Ortega; Won Il Choi; In Hyun Lee; Yazan Bdour; Jinjun Shi; Morteza Mahmoudi; Sangyong Jon; Edward A. Fisher; Omid C. Farokhzad

The pharmacological manipulation of liver X receptors (LXRs) has been an attractive therapeutic strategy for atherosclerosis treatment as they control reverse cholesterol transport and inflammatory response. This study presents the development and efficacy of nanoparticles (NPs) incorporating the synthetic LXR agonist GW3965 (GW) in targeting atherosclerotic lesions. Collagen IV (Col IV) targeting ligands are employed to functionalize the NPs to improve targeting to the atherosclerotic plaque, and formulation parameters such as the length of the polyethylene glycol (PEG) coating molecules are systematically optimized. In vitro studies indicate that the GW-encapsulated NPs upregulate the LXR target genes and downregulate proinflammatory mediator in macrophages. The Col IV-targeted NPs encapsulating GW (Col IV-GW-NPs) successfully reaches atherosclerotic lesions when administered for 5 weeks to mice with preexisting lesions, substantially reducing macrophage content (≈30%) compared to the PBS group, which is with greater efficacy versus nontargeting NPs encapsulating GW (GW-NPs) (≈18%). In addition, mice administered the Col IV-GW-NPs do not demonstrate increased hepatic lipid biosynthesis or hyperlipidemia during the treatment period, unlike mice injected with the free GW. These findings suggest a new form of LXR-based therapeutics capable of enhanced delivery of the LXR agonist to atherosclerotic lesions without altering hepatic lipid metabolism.


Small | 2018

Redox-Responsive Nanoparticle-Mediated Systemic RNAi for Effective Cancer Therapy

Xiaoding Xu; Jun Wu; Shuaishuai Liu; Phei Er Saw; Wei Tao; Yujing Li; Lisa Krygsman; Srinivasan Yegnasubramanian; Angelo M. De Marzo; Jinjun Shi; Charles J. Bieberich; Omid C. Farokhzad

Biodegradable polymeric nanoparticles (NPs) have demonstrated significant potential to improve the systemic delivery of RNA interference (RNAi) therapeutics, such as small interfering RNA (siRNA), for cancer therapy. However, the slow and inefficient siRNA release inside tumor cells generally observed for most biodegradable polymeric NPs may result in compromised gene silencing efficacy. Herein, a biodegradable and redox-responsive NP platform, composed of a solid poly(disulfide amide) (PDSA)/cationic lipid core and a lipid-poly(ethylene glycol) (lipid-PEG) shell for systemic siRNA delivery to tumor cells, is developed. This newly generated NP platform can efficiently encapsulate siRNA under extracellular environments and can respond to the highly concentrated glutathione (GSH) in the cytoplasm to induce fast intracellular siRNA release. By screening a library of PDSA polymers with different structures and chain lengths, the optimized NP platform shows the unique features of i) long blood circulation, ii) high tumor accumulation, iii) fast GSH-triggered intracellular siRNA release, and iv) exceptionally effective gene silencing. Together with the facile polymer synthesis technique and robust NP formulation enabling scale-up, this new redox-responsive NP platform may become an effective tool for RNAi-based cancer therapy.


Angewandte Chemie | 2015

Hydrophobic Cysteine Poly(disulfide)-based Redox-Hypersensitive Nanoparticle Platform for Cancer Theranostics†

Jun Wu; Lili Zhao; Xiaoding Xu; Nicolas Bertrand; Won Ii Choi; Basit Yameen; Jinjun Shi; Vishva Shah; Matthew Mulvale; James L. MacLean; Omid C. Farokhzad

Collaboration


Dive into the Xiaoding Xu's collaboration.

Top Co-Authors

Avatar

Omid C. Farokhzad

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Jinjun Shi

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Wei Tao

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Xiaoyuan Ji

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Jun Wu

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Mikyung Yu

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Yanlan Liu

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Morteza Mahmoudi

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Yujing Li

Brigham and Women's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge