Yu-cai Wang

Find an error

Name:
Organization: University of Science and Technology of China
Department: Department of Polymer Science and Engineering, Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, and Department of Chemical Physics
Title:

TOPICS

Co-reporter:Shi-Yong Li, Yang Liu, Cong-Fei Xu, Song Shen, Rong Sun, Xiao-Jiao Du, Jin-Xing Xia, Yan-Hua Zhu, Jun Wang
Journal of Controlled Release 2016 Volume 231() pp:17-28
Publication Date(Web):10 June 2016
DOI:10.1016/j.jconrel.2016.01.044
The core purpose of cancer immunotherapy is the sustained activation and expansion of the tumor specific T cells, especially tumor-infiltrating cytotoxic T lymphocytes (CTLs). Currently, one of the main foci of immunotherapy involving nano-sized carriers is on cancer vaccines and the role of professional antigen presenting cells, such as dendritic cells (DCs) and other phagocytic immune cells. Besides the idea that cancer vaccines promote T cell immune responses, targeting immune inhibitory pathways with nanoparticle delivered regulatory agents such as small interfering RNA (siRNA) to the difficultly-transfected tumor-infiltrating T cells may provide more information on the utility of nanoparticle-mediated cancer immunotherapy. In this study, we constructed nanoparticles to deliver cytotoxic T lymphocyte-associated molecule-4 (CTLA-4)-siRNA (NPsiCTLA-4) and showed the ability of this siRNA delivery system to enter T cells both in vitro and in vivo. Furthermore, T cell activation and proliferation were enhanced after NPsiCTLA-4 treatment in vitro. The ability of direct regulation of T cells of this CTLA-4 delivery system was assessed in a mouse model bearing B16 melanoma. Our results demonstrated that this nanoparticle delivery system was able to deliver CTLA-4-siRNA into both CD4+ and CD8+ T cell subsets at tumor sites and significantly increased the percentage of anti-tumor CD8+ T cells, while it decreased the ratio of inhibitory T regulatory cells (Tregs) among tumor infiltrating lymphocytes (TILs), resulting in augmented activation and anti-tumor immune responses of the tumor-infiltrating T cells. These data support the use of potent nanoparticle-based cancer immunotherapy for melanoma.T cell mediated immunotherapy is an effective treatment option for malignant melanoma. It is critical for such immunotherapy to obtain a sufficient number of functional/activated T cells. However, CTLA-4 plays a potent inhibitory role in T cell activation and proliferation, which significantly curbs T cell-mediated tumor rejection. Hence, we investigated a method to exploit a nanoparticle delivery system to efficiently deliver siRNA (NPsiCTLA-4) targeting an immune checkpoint molecule, i.e. cytotoxic T lymphocyte-associated molecule-4, to manipulate or modulate tumor-infiltrating T cells and to assess the effects of NPsiCTLA4 on the blockade of CTLA-4 and the resulting enhancement of T cell mediated anti-tumor immunotherapy.
Co-reporter:Hong-Xia Wang, Zu-Qi Zuo, Jin-Zhi Du, Yu-Cai Wang, Rong Sun, Zhi-Ting Cao, Xiao-Dong Ye, Ji-Long Wang, Kam W. Leong, Jun Wang
Nano Today 2016 Volume 11(Issue 2) pp:133-144
Publication Date(Web):April 2016
DOI:10.1016/j.nantod.2016.04.008
•Positively charged PEGylated nanomedicines exhibit shorter blood circulation and lower tumor accumulation than their neutral and anionic counterparts.•Positively charged PEGylated nanomedicines exhibit superior extravasation and penetration in tumors.•Positively charged PEGylated nanomedicines show higher cellular uptake in disaggregated tumors.•Positively charged PEGylated nanomedicines exhibit enhanced antitumour efficacy in a variety of tumor models.Physiochemical properties of nanomedicines determine their in vivo fate and ultimate therapeutic efficacy. Establishing correlations between nanoparticle properties and their physiological response is vitally important for nanomedicine design and optimization. To date, the correlation between surface charge, a fundamental property of a nanomedicine, and its therapeutic efficacy remains poorly understood. Here, we systematically investigated the influence of surface charge on the pharmacokinetics, tumor accumulation, penetration, and antitumor efficacy of nanoparticles constructed from PEG-b-PLA, loaded with docetaxel, and tuned by various lipids to yield three groups of ∼100 nm nanoparticles with positive, neutral or negative charge. Our results indicate that cationic PEGylated nanoparticles, although slightly inferior in blood circulation time and tumor accumulation, outperform their anionic or neutral counterparts in inhibiting tumor growth in five different tumor models. Docetaxel-loaded cationic nanoparticles significantly suppressed tumor growth with an inhibition ratio of ∼90%, compared with the ∼60% achieved by their anionic or neutral counterparts. Further studies reveal that better tumor penetration and 2.5-fold higher cellular uptake of cationic PEGylated nanoparticles is responsible for their superior treatment efficacy. This fundamental study provides a foundation for engineering the next generation of nano-delivery systems for in vivo applications.
Co-reporter:Zu-Qi Zuo, Kai-Ge Chen, Xiao-Yuan Yu, Gui Zhao, Song Shen, Zhi-Ting Cao, Ying-Li Luo, Yu-Cai Wang, Jun Wang
Biomaterials 2016 82() pp: 48-59
Publication Date(Web):March 2016
DOI:10.1016/j.biomaterials.2015.12.014
Cancer stem cells (CSCs), which hold a high capacity for self-renewal, play a central role in the development, metastasis, and recurrence of various malignancies. CSCs must be eradicated to cure instances of cancer; however, because they can reside far from tumor vessels, they are not easily targeted by drug agents carried by nanoparticle-based drug delivery systems. We herein demonstrate that promoting tumor penetration of nanoparticles by transforming growth factor β (TGF-β) signaling pathway inhibition facilitates CSC therapy. In our study, we observed that although nanoparticles carrying siRNA targeting the oncogene polo-like kinase 1 (Plk1) efficiently killed breast CSCs derived from MDA-MB-231 cells in vitro, this intervention enriched CSCs in the residual tumor tissue following systemic treatment. However, inhibition of the TGF-β signaling pathway with LY364947, an inhibitor of TGF-β type I receptor, promoted the penetration of nanoparticles in tumor tissue, significantly ameliorating the intratumoral distribution of nanoparticles in MDA-MB-231 xenografts and further leading to enhanced internalization of nanoparticles by CSCs. As a result, synergistic treatment with a nanoparticle drug delivery system and LY364947 inhibited tumor growth and reduced the proportion of CSCs in vivo. This study suggests that enhanced tumor penetration of drug-carrying nanoparticles can enhance CSCs clearance in vivo and consequently provide superior anti-tumor effects.
Co-reporter:Jun Wang
Nanomedicine: Nanotechnology, Biology and Medicine 2016 Volume 12(Issue 2) pp:464
Publication Date(Web):February 2016
DOI:10.1016/j.nano.2015.12.052
Co-reporter:Chun-Yang Sun;Yang Liu;Dr. Jin-Zhi Du;Zhi-Ting Cao;Cong-Fei Xu; Jun Wang
Angewandte Chemie International Edition 2016 Volume 55( Issue 3) pp:1010-1014
Publication Date(Web):
DOI:10.1002/anie.201509507

Abstract

Successful bench-to-bedside translation of nanomedicine relies heavily on the development of nanocarriers with superior therapeutic efficacy and high biocompatibility. However, the optimal strategy for improving one aspect often conflicts with the other. Herein, we report a tactic of designing tumor-pH-labile linkage-bridged copolymers of clinically validated poly(d,l-lactide) and poly(ethylene glycol) (PEG-Dlinkm-PDLLA) for safe and effective drug delivery. Upon arriving at the tumor site, PEG-Dlinkm-PDLLA nanoparticles will lose the PEG layer and increase zeta potential by responding to tumor acidity, which significantly enhances cellular uptake and improves the in vivo tumor inhibition rate to 78.1 % in comparison to 47.8 % of the non-responsive control. Furthermore, PEG-Dlinkm-PDLLA nanoparticles show comparable biocompatibility with the clinically used PEG-b-PDLLA micelle. The improved therapeutic efficacy and safety demonstrate great promise for our strategy in future translational studies.

Co-reporter:Chun-Yang Sun;Yang Liu;Dr. Jin-Zhi Du;Zhi-Ting Cao;Cong-Fei Xu; Jun Wang
Angewandte Chemie 2016 Volume 128( Issue 3) pp:1022-1026
Publication Date(Web):
DOI:10.1002/ange.201509507

Abstract

Successful bench-to-bedside translation of nanomedicine relies heavily on the development of nanocarriers with superior therapeutic efficacy and high biocompatibility. However, the optimal strategy for improving one aspect often conflicts with the other. Herein, we report a tactic of designing tumor-pH-labile linkage-bridged copolymers of clinically validated poly(d,l-lactide) and poly(ethylene glycol) (PEG-Dlinkm-PDLLA) for safe and effective drug delivery. Upon arriving at the tumor site, PEG-Dlinkm-PDLLA nanoparticles will lose the PEG layer and increase zeta potential by responding to tumor acidity, which significantly enhances cellular uptake and improves the in vivo tumor inhibition rate to 78.1 % in comparison to 47.8 % of the non-responsive control. Furthermore, PEG-Dlinkm-PDLLA nanoparticles show comparable biocompatibility with the clinically used PEG-b-PDLLA micelle. The improved therapeutic efficacy and safety demonstrate great promise for our strategy in future translational studies.

Co-reporter:Hong-Jun Li, Jin-Zhi Du, Jing Liu, Xiao-Jiao Du, Song Shen, Yan-Hua Zhu, Xiaoyan Wang, Xiaodong Ye, Shuming Nie, and Jun Wang
ACS Nano 2016 Volume 10(Issue 7) pp:6753
Publication Date(Web):May 31, 2016
DOI:10.1021/acsnano.6b02326
The currently low delivery efficiency and limited tumor penetration of nanoparticles remain two major challenges of cancer nanomedicine. Here, we report a class of pH-responsive nanoparticle superstructures with ultrasensitive size switching in the acidic tumor microenvironment for improved tumor penetration and effective in vivo drug delivery. The superstructures were constructed from amphiphilic polymer directed assembly of platinum-prodrug conjugated polyamidoamine (PAMAM) dendrimers, in which the amphiphilic polymer contains ionizable tertiary amine groups for rapid pH-responsiveness. These superstructures had an initial size of ∼80 nm at neutral pH (e.g., in blood circulation), but once deposited in the slightly acidic tumor microenvironment (pH ∼6.5–7.0), they underwent a dramatic and sharp size transition within a very narrow range of acidity (less than 0.1–0.2 pH units) and dissociated instantaneously into the dendrimer building blocks (less than 10 nm in diameter). This rapid size-switching feature not only can facilitate nanoparticle extravasation and accumulation via the enhanced permeability and retention effect but also allows faster nanoparticle diffusion and more efficient tumor penetration. We have further carried out comparative studies of pH-sensitive and insensitive nanostructures with similar size, surface charge, and chemical composition in both multicellular spheroids and poorly permeable BxPC-3 pancreatic tumor models, whose results demonstrate that the pH-triggered size switching is a viable strategy for improving drug penetration and therapeutic efficacy.Keywords: drug delivery; nanoparticle; particle size; pH-responsive; tumor microenvironment; tumor penetration
Co-reporter:Cong-Fei Xu;Chun-Yang Sun;Xian-Zhu Yang;Hong-Xia Wang;Hong-Jun Li;Zhi-Ting Cao;Xiao-Jiao Du;Jin-Zhi Du;Yan-Hua Zhu;Shuming Nie;Jun Wang
PNAS 2016 Volume 113 (Issue 15 ) pp:4164-4169
Publication Date(Web):2016-04-12
DOI:10.1073/pnas.1522080113
A principal goal of cancer nanomedicine is to deliver therapeutics effectively to cancer cells within solid tumors. However, there are a series of biological barriers that impede nanomedicine from reaching target cells. Here, we report a stimuli-responsive clustered nanoparticle to systematically overcome these multiple barriers by sequentially responding to the endogenous attributes of the tumor microenvironment. The smart polymeric clustered nanoparticle (iCluster) has an initial size of ∼100 nm, which is favorable for long blood circulation and high propensity of extravasation through tumor vascular fenestrations. Once iCluster accumulates at tumor sites, the intrinsic tumor extracellular acidity would trigger the discharge of platinum prodrug-conjugated poly(amidoamine) dendrimers (diameter ∼5 nm). Such a structural alteration greatly facilitates tumor penetration and cell internalization of the therapeutics. The internalized dendrimer prodrugs are further reduced intracellularly to release cisplatin to kill cancer cells. The superior in vivo antitumor activities of iCluster are validated in varying intractable tumor models including poorly permeable pancreatic cancer, drug-resistant cancer, and metastatic cancer, demonstrating its versatility and broad applicability.
Co-reporter:Chun-Yang Sun; Song Shen; Cong-Fei Xu; Hong-Jun Li; Yang Liu; Zhi-Ting Cao; Xian-Zhu Yang; Jin-Xing Xia;Jun Wang
Journal of the American Chemical Society 2015 Volume 137(Issue 48) pp:15217-15224
Publication Date(Web):November 16, 2015
DOI:10.1021/jacs.5b09602
Although surface PEGylation of siRNA vectors is effective for preventing protein adsorption and thereby helps these vectors to evade the reticuloendothelial system (RES) in vivo, it also suppresses the cellular uptake of these vectors by target cells. This dilemma could be overcome by employing stimuli-responsive shell-detachable nanovectors to achieve enhanced cellular internalization while maintaining prolonged blood circulation. Among the possible stimuli, dysregulated pH in tumor (pHe) is the most universal and practical. However, the design of pHe-sensitive system is problematic because of the subtle differences between the pHe and pH in other tissues. Here, a simple acid-sensitive bridged copolymer is developed and used for tumor-targeted systemic delivery of siRNA. After forming the micelleplex delivery system, the corresponding nanoparticles (Dm-NP) might undergo several modifications as follows: (i) a poly(ethylene glycol) (PEG) corona, which is stable in the circulatory system and protects nanovectors from RES clearance; (ii) a pHe responsive linkage breakage, which induces PEG detachment at tumor sites and thereby facilitates cell targeting; and (iii) a cell-penetration peptide, which is exposed upon the removal of PEG and further enhances cellular uptake. Thus, Dm-NP achieved both prolonged circulation and effective accumulation in tumor cells and resulted in the safe and enhanced inhibition of non-small cell lung cancer growth.
Co-reporter:Shi Liang;Xian-Zhu Yang;Xiao-Jiao Du;Hong-Xia Wang;Hong-Jun Li;Wei-Wei Liu;Yan-Dan Yao;Yan-Hua Zhu;Yin-Chu Ma;Jun Wang;Er-Wei Song
Advanced Functional Materials 2015 Volume 25( Issue 30) pp:4778-4787
Publication Date(Web):
DOI:10.1002/adfm.201501548

Delivery of small interfering RNA (siRNA) by nanocarriers has shown promising therapeutic potential in cancer therapy. However, poor understanding of the correlation between the physicochemical properties of nanocarriers and their interactions with biological systems has significantly hindered its anticancer efficacy. Herein, in order to identify the optimal size of nanocarriers for siRNA delivery, different sized cationic micellar nanoparticles (MNPs) (40, 90, 130, and 180 nm) are developed that exhibit similar siRNA binding efficacies, shapes, surface charges, and surface chemistries (PEGylation) to ensure size is the only variable. Size-dependent biological effects are carefully and comprehensively evaluated through both in vitro and in vivo experiments. Among these nanocarriers, the 90 nm MNPs show the optimal balance of prolonged circulation and cellular uptake by tumor cells, which result in the highest retention in tumor cells. In contrast, larger MNPs are rapidly cleared from the circulation and smaller MNPs are inefficiently taken up by tumor cells. Accordingly, 90 nm MNPs carrying polo-like kinase 1 (Plk1)-specific siRNA (siPlk1) show superior antitumor efficacy, indicating that 90 nm could either be the optimal size for systemic delivery of siRNA or close to it. Our findings provide valuable information for rationally designing nanocarriers for siRNA-based cancer therapy in the future.

Co-reporter:Yang Liu, Xin Wang, Chun-Yang Sun, and Jun Wang
ACS Applied Materials & Interfaces 2015 Volume 7(Issue 1) pp:1012
Publication Date(Web):December 18, 2014
DOI:10.1021/am508262j
Hepatocellular carcinoma (HCC) is one of the most common malignant human tumors worldwide, but no effective therapeutic options are currently available. The cancer stem cell (CSC) has proven to play a central role in the development, metastasis, and recurrence of HCC. In this study, we report a dual functional mitogen-activated protein kinase inhibitor (U0126)-based therapy for treating both bulk HCC and HCC CSCs, using poly(ethylene glycol)-b-poly(d,l-lactide) (PEG-PLA) nanoparticles as the drug carrier. It is demonstrated that nanoparticle encapsulation enhanced the cell uptake of U0126 in HCC CSCs and that enhanced endocytosis lead to augmented cytotoxicity of U0126 in HCC CSCs. Moreover, the nanoparticle encapsulation increased the inhibition of self-renewal capability, prolonged the circulation time, and increased the tumor accumulation of U0126 when compared with the use of the free inhibitor. The systemic delivery of U0126 remarkably enhanced the suppression of tumor development with decreased CSCs in the HepG2 xenograft simultaneously with reduced systemic toxicity.Keywords: CSC; hepatocellular carcinoma; mitogen-activated protein kinase; nanoparticle; U0126
Co-reporter:Qinqin Cheng, Hongdong Shi, Hai Huang, Zhiting Cao, Jun Wang and Yangzhong Liu  
Chemical Communications 2015 vol. 51(Issue 99) pp:17536-17539
Publication Date(Web):12 Oct 2015
DOI:10.1039/C5CC07853A
Self-assembled cholesterol–asplatin-incorporated nanoparticles (SCANs) were prepared for oral delivery of a Pt(IV) prodrug. SCANs exhibit high gastrointestinal stability, sustained drug release and enhanced cell uptake. The oral bioavailability of SCANs was 4.32-fold higher than that of free Pt(IV) prodrugs. The oral administration of SCANs efficaciously inhibits tumor growth with negligible toxicity.
Co-reporter:Shi-Yong Li, Rong Sun, Hong-Xia Wang, Song Shen, Yang Liu, Xiao-Jiao Du, Yan-Hua Zhu, Wang Jun
Journal of Controlled Release 2015 Volume 205() pp:7-14
Publication Date(Web):10 May 2015
DOI:10.1016/j.jconrel.2014.11.011
Aberrant DNA hypermethylation is critical in the regulation of renewal and maintenance of cancer stem cells (CSCs), which represent targets for carcinogenic initiation by chemical and environmental agents. The administration of decitabine (DAC), which is a DNA hypermethylation inhibitor, is an attractive approach to enhancing the chemotherapeutic response and overcoming drug resistance by CSCs. In this study, we investigated whether low-dose DAC encapsulated in nanoparticles could be used to sensitize bulk breast cancer cells and CSCs to chemotherapy. In vitro studies revealed that treatment with nanoparticles loaded with low-dose DAC (NPDAC) combined with nanoparticles loaded with doxorubicin (NPDOX) better reduced the proportion of CSCs with high aldehyde dehydrogenase activity (ALDHhi) in the mammospheres of MDA-MB-231 cells, and better overcame the drug resistance by ALDHhi cells. Subsequently, systemic delivery of NPDAC significantly down-regulated the expression of DNMT1 and DNMT3b in a MB-MDA-231 xenograft murine model and induced increased caspase-9 expression, which contributed to the increased sensitivity of the bulk cancer cells and CSCs to NPDOX treatment. Importantly, the combined treatment of NPDAC and NPDOX resulted in the lowest proportion of ALDHhi CSCs and the highest proportion of apoptotic tumor cells, and the best tumor suppressive effects in inhibiting breast cancer growth.Combination therapy with nanoparticles loaded with epigenetic-targeted decitabine (NPDAC) increases the sensitivity of cancer stem cells to treatment of nanoparticles loaded with doxorubicin (NPDOX).
Co-reporter:Cong-fei Xu, Yang Liu, Song Shen, Yan-hua Zhu, Jun Wang
Journal of Controlled Release 2015 Volume 213() pp:e23-e24
Publication Date(Web):10 September 2015
DOI:10.1016/j.jconrel.2015.05.035
Co-reporter:Song Shen, Xiao-Jiao Du, Jing Liu, Rong Sun, Yan-Hua Zhu, Jun Wang
Journal of Controlled Release 2015 Volume 208() pp:14-24
Publication Date(Web):28 June 2015
DOI:10.1016/j.jconrel.2014.12.043
Basal-like triple negative breast cancer (TNBC) has received particular clinical interest due to its high frequency, poor baseline prognosis and lack of effective clinical therapy. Bortezomib, which was the first proteasome inhibitor approved for the treatment of multiple myeloma, has been proven to be worth investigating for this subtype of breast cancer. In our study, the amphiphilic copolymer poly(ethylene glycol)-block-poly(d,l-lactide) (PEG-b-PLA) was utilized as an excellent delivery carrier of bortezomib (BTZ) to overcome its clinical limitations including low water solubility and unstable properties. Bortezomib encapsulated nanoparticles (NPBTZ) can efficiently deliver the drug into both CSCs (cancer stem cells) and non-CSCs, resulting in proliferation inhibition and apoptosis induction. Remarkably, NPBTZ can more effectively affect the stemness of CSCs compared with free BTZ. Administration of this drug delivery system can markedly prolong the bortezomib circulation half-life and augment the enrichment of drugs in tumor tissue, then enhance the suppression of tumor growth, suggesting the therapeutic promise of NPBTZ delivery in basal-like TNBC therapy.
Co-reporter:Rong Sun, Xiao-Jiao Du, Chun-Yang Sun, Song Shen, Yang Liu, Xian-Zhu Yang, Yan Bao, Yan-Hua Zhu and Jun Wang  
Biomaterials Science 2015 vol. 3(Issue 7) pp:1105-1113
Publication Date(Web):04 Mar 2015
DOI:10.1039/C4BM00430B
Polymeric nanoparticles have been widely used as nano-drug delivery systems in preclinical and clinical trials for cancer therapy, and these systems usually need to be sterically stabilized by poly(ethylene glycol) (PEG) to maintain stability and avoid rapid clearance by the immune system. Recently, zwitterionic materials have been demonstrated to be potential alternatives to the classic PEG. Herein, we developed two drug delivery systems stabilized by zwitterionic polyphosphoesters. These nanoparticles showed favourable stability and anti-protein absorption ability in vitro. Meanwhile, as drug carriers, these zwitterionic polyphosphoester-stabilized nanoparticles significantly prolonged drug circulation half-lives and increased drug accumulation in tumors, which was comparable to PEG-stabilized nanoparticles. Systemic delivery of doxorubicin (DOX) by zwitterionic polyphosphoester-stabilized nanoparticles significantly inhibited tumor growth in a MDA-MB-231 tumor model, suggesting the potential of zwitterionic polyphosphoester-based nanoparticles in anticancer drug delivery.
Co-reporter:Rong Sun, Yang Liu, Shi-Yong Li, Song Shen, Xiao-Jiao Du, Cong-Fei Xu, Zhi-Ting Cao, Yan Bao, Yan-Hua Zhu, Ya-Ping Li, Xian-Zhu Yang, Jun Wang
Biomaterials 2015 37() pp: 405-414
Publication Date(Web):
DOI:10.1016/j.biomaterials.2014.10.018
Co-reporter:Cong-Fei Xu, Yang Liu, Song Shen, Yan-Hua Zhu, Jun Wang
Biomaterials 2015 51() pp: 1-11
Publication Date(Web):
DOI:10.1016/j.biomaterials.2015.01.068
Co-reporter:Xiao-Jiao Du, Ji-Long Wang, Wei-Wei Liu, Jin-Xian Yang, Chun-Yang Sun, Rong Sun, Hong-Jun Li, Song Shen, Ying-Li Luo, Xiao-Dong Ye, Yan-Hua Zhu, Xian-Zhu Yang, Jun Wang
Biomaterials 2015 69() pp: 1-11
Publication Date(Web):
DOI:10.1016/j.biomaterials.2015.07.048
Co-reporter:Xian-Zhu Yang;Xiao-Jiao Du;Yang Liu;Yan-Hua Zhu;Yang-Zhong Liu;Ya-Ping Li;Jun Wang
Advanced Materials 2014 Volume 26( Issue 6) pp:931-936
Publication Date(Web):
DOI:10.1002/adma.201303360
Co-reporter:Yang Liu, Yan-Hua Zhu, Cheng-Qiong Mao, Shuang Dou, Song Shen, Zi-Bin Tan, Jun Wang
Journal of Controlled Release 2014 Volume 192() pp:114-121
Publication Date(Web):28 October 2014
DOI:10.1016/j.jconrel.2014.07.001
There is no effective clinical therapy yet for triple-negative breast cancer (TNBC) without particular human epidermal growth factor receptor-2, estrogen and progesterone receptor expression. In this study, we report a molecularly targeted and synthetic lethality-based siRNA therapy for TNBC treatment, using cationic lipid assisted poly(ethylene glycol)-b-poly(d,l-lactide) (PEG-PLA) nanoparticles as the siRNA carrier. It is demonstrated that only in c-Myc overexpressed TNBC cells, while not in normal mammary epithelial cells, delivery of siRNA targeting cyclin-dependent kinase 1 (CDK1) with the nanoparticle carrier (NPsiCDK1) induces cell viability decreasing and cell apoptosis through RNAi-mediated CDK1 expression inhibition, indicating the synthetic lethality between c-Myc with CDK1 in TNBC cells. Moreover, systemic delivery of NPsiCDK1 is able to suppress tumor growth in mice bearing SUM149 and BT549 xenograft and cause no systemic toxicity or activate the innate immune response, suggesting the therapeutic promise with such nanoparticles carrying siCDK1 for c-Myc overexpressed triple negative breast cancer.Schematic illustration of cancer therapy of triple negative breast cancer (TNBC) over-expressed c-Myc with CDK1 siRNA delivered by cationic lipid (BHEM-Chol) assisted poly(ethylene glycol)-b-poly(d,l-lactide) (PEG5K-PLA11K) nanoparticles.
Co-reporter:Qinqin Cheng, Hongdong Shi, Hongxia Wang, Yuanzeng Min, Jun Wang and Yangzhong Liu  
Chemical Communications 2014 vol. 50(Issue 56) pp:7427-7430
Publication Date(Web):28 Feb 2014
DOI:10.1039/C4CC00419A
Asplatin, a fusion of aspirin and cisplatin, exhibits significant cytotoxicity in tumor cells and almost fully overcomes the drug resistance of cisplatin resistant cells. Asplatin is highly accumulated in cancer cells and is activated upon the reduction by ascorbic acid.
Co-reporter:Chun-Yang Sun;Shuang Dou;Jin-Zhi Du;Xian-Zhu Yang;Ya-Ping Li;Jun Wang
Advanced Healthcare Materials 2014 Volume 3( Issue 2) pp:261-272
Publication Date(Web):
DOI:10.1002/adhm.201300091

Abstract

Polyphosphoesters with repeating phosphoester linkages in the backbone can be easily functionalized, are biodegradable and potentially biocompatible, and may be potential candidates as polymer carriers of drug conjugates. Here, the efficacy of a polyphosphoester drug conjugate as an anticancer agent in vivo is assessed for the first time. With controlled synthesis, doxorubicin conjugated to poly(ethylene glycol)-block-polyphosphoester (PPEH–DOX) via labile hydrazone bonds form spherical nanoparticles in aqueous solution with an average diameter of ≈60 nm. These nanoparticles are effectively internalized by MDA-MB-231 breast cancer cells and release the conjugated doxorubicin in response to the intracellular pH of endosomes and lysosomes, resulting in significant antiproliferative activity in cancer cells. Compared with free doxorubicin injection, PPEH–DOX injection exhibits much longer circulation behavior in the plasma of mice and leads to enhanced drug accumulation in tumor cells. In an MDA-MB-231 xenograft murine model, inhibition of tumor growth with systemic delivery of PPEH–DOX nanoparticles is more pronounced compared with free doxorubicin injection, suggesting the potential of polyphosphoesters as carriers of drug conjugates in cancer therapy.

Co-reporter:Shuang Dou;Xian-Zhu Yang;Meng-Hua Xiong;Chun-Yang Sun;Yan-Dan Yao;Yan-Hua Zhu;Jun Wang
Advanced Healthcare Materials 2014 Volume 3( Issue 11) pp:1792-1803
Publication Date(Web):
DOI:10.1002/adhm.201400037

Patients with Her2-overexpressing (Her2+) breast cancers generally have a poorer prognosis due to the high aggressiveness and chemoresistance of the disease. Small interfering RNA (siRNA) targeting the gene encoding polo-like kinase 1 (Plk1; siPlk1) has emerged as an efficient therapeutic agent for Her2+ breast cancers. Poly(ethylene glycol)-block-poly(d,l-lactide) (PEG-PLA)-based nanoparticles for siRNA delivery were previously developed and optimized. In this study, for targeted delivery of siPlk1 to Her2+ breast cancer, anti-Her2 single-chain variable fragment antibody (ScFvHer2)-decorated PEG-PLA-based nanoparticles with si Plk1 encapsulation (ScFvHer2-NPsi Plk1) are developed. With the rationally designed conjugation site, ScFvHer2-NPsiRNA can specifically bind to the Her2 antigen overexpressed on the surface of Her2+ breast cancer cells. Therefore, ScFvHer2-NPsi Plk1 exhibits improved cellular uptake, promoted Plk1 silencing efficiency, and induced enhanced tumor cell apoptosis in Her2+ breast cancer cells, when compared with nontargeted NPsi Plk1. More importantly, ScFvHer2-NPsiRNA markedly enhances the accumulation of siRNA in Her2+ breast tumor tissue, and remarkably improves the efficacy of tumor suppression. Dose-dependent anti-tumor efficacy further demonstrates that ScFvHer2-decorated PEG-PLA-based nanoparticles with siPlk1 encapsulation can significantly enhance the inhibition of Her2+ breast tumor growth and reduce the dose of injected siRNA. These results suggest that ScFvHer2-decorated PEG-PLA-based nanoparticles show great potential for targeted RNA interference therapy of Her2+ breast tumor.

Co-reporter:Song Shen, Chong-Qiong Mao, Xian-Zhu Yang, Xiao-Jiao Du, Yang Liu, Yan-Hua Zhu, and Jun Wang
Molecular Pharmaceutics 2014 Volume 11(Issue 8) pp:2612-2622
Publication Date(Web):February 12, 2014
DOI:10.1021/mp400714z
Synthetic lethal interaction provides a conceptual framework for the development of wiser cancer therapeutics. In this study, we exploited a therapeutic strategy based on the interaction between GATA binding protein 2 (GATA2) downregulation and the KRAS mutation status by delivering small interfering RNA targeting GATA2 (siGATA2) with cationic lipid-assisted polymeric nanoparticles for treatment of non-small-cell lung carcinoma (NSCLC) harboring oncogenic KRAS mutations. Nanoparticles carrying siGATA2 (NPsiGATA2) were effectively taken up by NSCLC cells and resulted in targeted gene suppression. NPsiGATA2 selectively inhibited cell proliferation and induced cell apoptosis in KRAS mutant NSCLC cells. However, this intervention was harmless to normal KRAS wild-type NSCLC cells and HL7702 hepatocytes, confirming the advantage of synthetic lethality-based therapy. Moreover, systemic delivery of NPsiGATA2 significantly inhibited tumor growth in the KRAS mutant A549 NSCLC xenograft murine model, suggesting the therapeutic promise of NPsiGATA2 delivery in KRAS mutant NSCLC therapy.Keywords: cationic lipid-assisted polymeric nanoparticle; GATA2; non-small-cell lung carcinoma; siRNA delivery; synthetic lethal therapy;
Co-reporter:Hong-Jun Li, Hong-Xia Wang, Chun-Yang Sun, Jin-Zhi Du and Jun Wang  
RSC Advances 2014 vol. 4(Issue 4) pp:1961-1964
Publication Date(Web):22 Nov 2013
DOI:10.1039/C3RA44866E
Novel light-responsive nanoparticles based on an amphiphile with a single photolabile linker between its hydrophilic head and hydrophobic tail was developed for small interfering RNA (siRNA) delivery. Upon UV exposure, cleavage of the linkage resulted in rapid shell detachment of the nanoparticles, which facilitated siRNA release and enhanced gene silencing efficiency.
Co-reporter:Weiwei Liu;Yucai Wang;Yang Li;Feng Wang;Xianzhu Yang;Tianmeng Sun;Jinzhi Du;Jun Wang
Chinese Journal of Chemistry 2014 Volume 32( Issue 1) pp:51-56
Publication Date(Web):
DOI:10.1002/cjoc.201300736

Abstract

Inspired by the influence of chemical structure of end groups on the phase transition temperature of thermoresponsive polymers, we demonstrated a strategy to control the multi-responsiveness of polymer assemblies via subtle modification of end groups of thermoresponsive polymer segments and revealed its potential application for drug delivery. By developing polymer assemblies composed of poly(aliphatic ester) as the inner core and thermoresponsive polyphosphoester as the outer shell, we showed that end groups of thermoresponsive polyphosphoester segments controlled the surface property of assemblies and further determined the stimuli-responsive behavior. The phase-transition temperatures of the unmodified polymer assemblies are tightly controlled by their surface properties due to the hydrophilic to hydrophobic transitions of end groups in response to an environmental stimulus (e.g. pH or light irradiation). External control over these surface properties can by asserted by adjusting the chemical structure and composition of the terminal groups of the thermoresponsive polyphosphoesters.

Co-reporter:Yang Li;Feng Wang;TianMeng Sun;JinZhi Du;XianZhu Yang
Science China Chemistry 2014 Volume 57( Issue 4) pp:579-585
Publication Date(Web):2014 April
DOI:10.1007/s11426-013-5056-9
The chemical structure of end groups influenced the phase transition temperature of thermoresponsive polymers. We demonstrated a strategy for the preparation of the pH/thermo-responsive polymeric nanoparticles via subtle modification of end groups of thermoresponsive polymer segments with a carboxyl group and revealed its potential application for enhanced intracellular drug delivery. By developing a polymeric nanoparticle composed of poly(aliphatic ester) as the inner core and thermoresponsive polyphosphoester as the outer shell, we showed that end groups of thermoresponsive polyphosphoester segments modified by carboxyl groups exhibited a pH/thermo-responsive behavior due to the hydrophilic to hydrophobic transitions of the end groups in response to the pH. Moreover, by encapsulating doxorubicin into the hydrophobic core of such pH/thermo-responsive polymer nanoparticles, their intracellular delivery and cytotoxicity to wild-type and drug-resistant tumor cells were significantly enhanced through the phase-transition-dependent drug release that was triggered by endosomal/lysosomal pH. This novel strategy and the multi-responsive polymer nanoparticles achieved by the subtle chain-terminal modification of thermoresponsive polymers provide a smart platform for biomedical applications.
Co-reporter:Tian-Meng Sun, Yu-Cai Wang, Feng Wang, Jin-Zhi Du, Cheng-Qiong Mao, Chun-Yang Sun, Rui-Zhi Tang, Yang Liu, Jing Zhu, Yan-Hua Zhu, Xian-Zhu Yang, Jun Wang
Biomaterials 2014 35(2) pp: 836-845
Publication Date(Web):
DOI:10.1016/j.biomaterials.2013.10.011
Co-reporter:Hong-Xia Wang, Xian-Zhu Yang, Chun-Yang Sun, Cheng-Qiong Mao, Yan-Hua Zhu, Jun Wang
Biomaterials 2014 35(26) pp: 7622-7634
Publication Date(Web):
DOI:10.1016/j.biomaterials.2014.05.050
Co-reporter:Wen-qing Li, Chun-yang Sun, Feng Wang, Yu-cai Wang, Yi-wen Zhai, Meng Liang, Wen-jing Liu, Zhi-min Liu, Jun Wang, and Fei Sun
Nano Letters 2013 Volume 13(Issue 6) pp:2477-2484
Publication Date(Web):May 20, 2013
DOI:10.1021/nl400536d
During the process of human civilization, owning household pets has become increasingly popular. However, dogs and cats may be reservoirs or vectors of transmissible diseases to humans. Confronted with the overpopulation of pets, traditional contraception methods, surgical methods of sterilization, for animals are used, namely, ovariohysterectomy and orchidectomy. Therefore, a simple, nonsurgical, controllable, more effective and less expensive contraception method is highly desirable. In this study, we show that in situ testicular injection of methoxy poly(ethylene glycol)-modified gold nanorods with near-infrared irradiation in male mice can achieve short-lived or permanent male infertility. In a lower hyperthermia treatment, the morphology of testes and seminiferous tubules is only partly injured, and fertility indices are decreased to ∼10% at day 7, then recovered to 50% at day 60. In a higher hyperthermia treatment, the morphology of testes and seminiferous tubules are totally destroyed, and fertility indices are decreased to 0 at day 7. Overall, our results indicate a potential application of plasmonic nanomaterials for male contraception.
Co-reporter:Hong-Xia Wang, Meng-Hua Xiong, Yu-Cai Wang, Jing Zhu, Jun Wang
Journal of Controlled Release 2013 Volume 166(Issue 2) pp:106-114
Publication Date(Web):10 March 2013
DOI:10.1016/j.jconrel.2012.12.017
Due to its efficient and specific gene silencing ability, RNA interference has shown great potential in the treatment of liver diseases. However, achieving in vivo delivery of siRNA to critical liver cells remains the biggest obstacle for this technique to be a real clinic therapeutic modality. Here, we describe a promising liver targeting siRNA delivery system based on N-acetylgalactosamine functionalized mixed micellar nanoparticles (Gal-MNP), which can efficiently deliver siRNA to hepatocytes and silence the target gene expression after systemic administration. The Gal-MNP were assembled in aqueous solution from mixed N-acetylgalactosamine functionalized poly(ethylene glycol)-b-poly(ε-caprolactone) and cationic poly(ε-caprolactone)-b-poly(2-aminoethyl ethylene phosphate) (PCL-b-PPEEA); the properties of nanoparticles, including particle size, zeta potential and the density of poly(ethylene glycol) could be easily regulated. The hepatocyte-targeting effect of Gal-MNP was demonstrated by significant enriching of fluorescent siRNA in primary hepatocytes in vitro and in vivo. Successful down-regulation of liver-specific apolipoprotein B (apoB) expression was achieved in mouse liver, at both the transcriptional and protein level, following intravenous injection of Gal-MNP/siapoB to BALB/c mice. Systemic delivery of Gal-MNP/siRNA did not induce the innate immune response or positive hepatotoxicity. The results of this study suggested therapeutic potential for the Gal-MNP/siRNA system in liver disease.
Co-reporter:Juan Wu, Tian-Meng Sun, Xian-Zhu Yang, Jing Zhu, Xiao-Jiao Du, Yan-Dan Yao, Meng-Hua Xiong, Hong-Xia Wang, Yu-Cai Wang and Jun Wang  
Biomaterials Science 2013 vol. 1(Issue 11) pp:1143-1150
Publication Date(Web):17 Jul 2013
DOI:10.1039/C3BM60099H
Effective systemic therapy is often necessary to treat hepatocellular carcinoma (HCC). We synthesized a Gal-PPE nanogel consisting of a cross-linked polyphosphate core and galactosylated poly(ethylene glycol) arms for enhanced doxorubicin delivery to diethylnitrosamine-induced HCC in rats. The Gal-PPE nanogel exhibited high affinity to HepG2 cells in vitro, mediated by the asialoglycoprotein receptor. In vivo studies revealed that the Gal-PPE nanogel was taken up more efficiently by hepatocytes, in contrast to m-PPE nanogel. Consequently, doxorubicin delivery with Gal-PPE significantly inhibited the progress of HCC, reducing neoplastic liver nodules and prolonging the survival time of HCC rats more significantly. These results demonstrate the potential of Gal-PPE as a nanocarrier for improved HCC chemotherapy.
Co-reporter:Meng-Hua Xiong, Yan Bao, Xiao-Jiao Du, Zi-Bin Tan, Qiu Jiang, Hong-Xia Wang, Yan-Hua Zhu, and Jun Wang
ACS Nano 2013 Volume 7(Issue 12) pp:10636
Publication Date(Web):November 7, 2013
DOI:10.1021/nn403146t
Differential anticancer drug delivery that selectively releases a drug within a tumor represents an ideal cancer therapy strategy. Herein, we report differential drug delivery to the tumor through the fabrication of a special bacteria-accumulated tumor environment that responds to bacteria-sensitive triple-layered nanogel (TLN). We demonstrate that the attenuated bacteria SBY1 selectively accumulated in tumors and were rapidly cleared from normal tissues after intravenous administration, leading to a unique bacteria-accumulated tumor environment. Subsequent administrated doxorubicin-loaded TLN (TLND) was thus selectively degraded in the bacteria-accumulated tumor environment after its accumulation in tumors, triggering differential doxorubicin release and selectively killing tumor cells. This concept can be extended and improved by using other factors secreted by bacteria or materials to fabricate a unique tumor environment for differential drug delivery, showing potential applications in drug delivery.Keywords: bacteria; cancer therapy; differential drug delivery; doxorubicin; nanogel
Co-reporter:You-Yong Yuan;Cheng-Qiong Mao;Xiao-Jiao Du;Jin-Zhi Du;Feng Wang;Jun Wang
Advanced Materials 2012 Volume 24( Issue 40) pp:5476-5480
Publication Date(Web):
DOI:10.1002/adma.201202296
Co-reporter:Meng-Hua Xiong ; Yan Bao ; Xian-Zhu Yang ; Yu-Cai Wang ; Baolin Sun ;Jun Wang
Journal of the American Chemical Society 2012 Volume 134(Issue 9) pp:4355-4362
Publication Date(Web):February 3, 2012
DOI:10.1021/ja211279u
We report a new strategy for differential delivery of antimicrobials to bacterial infection sites with a lipase-sensitive polymeric triple-layered nanogel (TLN) as the drug carrier. The TLN was synthesized by a convenient arm-first procedure using an amphiphilic diblock copolymer, namely, monomethoxy poly(ethylene glycol)-b-poly(ε-caprolactone), to initiate the ring-opening polymerization of the difunctional monomer 3-oxapentane-1,5-diyl bis(ethylene phosphate). The hydrophobic poly(ε-caprolactone) (PCL) segments collapsed and surrounded the polyphosphoester core, forming a hydrophobic and compact molecular fence in aqueous solution which prevented antibiotic release from the polyphosphoester core prior to reaching bacterial infection sites. However, once the TLN sensed the lipase-secreting bacteria, the PCL fence of the TLN degraded to release the antibiotic. Using Staphylococcus aureus (S. aureus) as the model bacterium and vancomycin as the model antimicrobial, we demonstrated that the TLN released almost all the encapsulated vancomycin within 24 h only in the presence of S. aureus, significantly inhibiting S. aureus growth. The TLN further delivered the drug into bacteria-infected cells and efficiently released the drug to kill intracellular bacteria. This technique can be generalized to selectively deliver a variety of antibiotics for the treatment of various infections caused by lipase-secreting bacteria and thus provides a new, safe, effective, and universal approach for the treatment of extracellular and intracellular bacterial infections.
Co-reporter:You-Yong Yuan, Jin-Zhi Du, Wen-Jing Song, Feng Wang, Xian-Zhu Yang, Meng-Hua Xiong and Jun Wang  
Journal of Materials Chemistry A 2012 vol. 22(Issue 18) pp:9322-9329
Publication Date(Web):2012/03/09
DOI:10.1039/C2JM30663H
Biocompatible and biodegradable nanogels with a branched structure designed as potential carriers for drug delivery are reported here. The nanogels were prepared by the reaction of 3,6-dioxaoctan-1,8-diyl bis(ethylene phosphate) (TEGDP) with tris(2-aminoethyl)amine (TREN) in an ionic liquid containing miniemulsion; the particle size was tunable in the nanoscopic range. The biocompatibility of the nanogels was evaluated using both in vitro and in vivo experiments. The nanogels could efficiently load the anticancer drug doxorubicin and showed enzyme-responsive drug release behavior. The nanogels were further labelled with fluorescein isothiocyanate through a reaction with the surface amino groups to demonstrate functionalization. Flow cytometric analyses and confocal laser scanning microscope observations demonstrated that the nanogels could be efficiently taken up by the human breast cancer cell line MDA-MB-231. These biocompatible, biodegradable and surface functionalizable nanogels are expected to have utility in the field of drug delivery.
Co-reporter:Shuang Dou, Yan-Dan Yao, Xian-Zhu Yang, Tian-Meng Sun, Cheng-Qiong Mao, Er-Wei Song, Jun Wang
Journal of Controlled Release 2012 Volume 161(Issue 3) pp:875-883
Publication Date(Web):10 August 2012
DOI:10.1016/j.jconrel.2012.05.015
The targeted delivery of small interfering RNA (siRNA) to specific tumor tissues and tumor cells remains as one of the key challenges in the development of RNA interference as a therapeutic application. To target breast cancer, we developed a therapeutic delivery system using a fusion protein of an anti-Her2 single-chain antibody fragment with a positively charged protamine, namely F5-P, as the carrier to specifically deliver siRNA-targeting DNA methyltransferases 1 and/or 3b genes (siDNMTs) into Her2-expressing breast tumor cells. The carrier F5-P, expressed by the Escherichia coli system, was able to bind siRNA molecules and specifically deliver the siRNA to Her2-expressing BT474 breast cancer cells but not Her2-nonexpressing MDA-MB-231 breast cancer cells, while delivery of siDNMTs to BT474 cells successfully silenced the expression of targeted DNA methyltransferases (DNMTs) and facilitated the de-methylation of the RASSF1A tumor suppressor gene promoter, leading to the suppression of tumor cell proliferation. Moreover, as demonstrated in the BT474 xenograft murine model, F5-P successfully delivered siRNA into a Her2-expressing breast tumor, and tumor growth inhibition was mediated by an intravenous injection of F5-P/siDNMTs complex by down-regulating the expression of DNMTs and restoring tumor suppressor gene expression. These data suggest that the delivery of siDNMTs by F5-P could be used to treat Her2-expressing breast cancer.
Co-reporter:Jin-Zhi Du, Hong-Yan Long, You-Yong Yuan, Meng-Meng Song, Liang Chen, Hong Bi and Jun Wang  
Chemical Communications 2012 vol. 48(Issue 9) pp:1257-1259
Publication Date(Web):16 Dec 2011
DOI:10.1039/C2CC16363B
Micelle-to-vesicle morphological transition has been achieved by light-induced rapid hydrophilic arm detachment from a star polymer. This provides a remote and clean method to control morphology transition of polymeric assemblies.
Co-reporter:You-Yong Yuan, Jin-Zhi Du and Jun Wang  
Chemical Communications 2012 vol. 48(Issue 4) pp:570-572
Publication Date(Web):07 Nov 2011
DOI:10.1039/C1CC16065F
A simple and universal route to functional cyclic polyesters has been demonstrated, combining two consecutive click reactions of azide–alkyne cycloaddition of linear hetero-bifunctional precursors and thiol–ene coupling for post cyclization functionalizations. Functional cationic and thermo-responsive cyclic polyphosphoesters have been synthesized to demonstrate the efficiency of the procedures.
Co-reporter:Xi-Qiu Liu, Meng-Hua Xiong, Xiao-Ting Shu, Rui-Zhi Tang, and Jun Wang
Molecular Pharmaceutics 2012 Volume 9(Issue 10) pp:2863-2874
Publication Date(Web):August 27, 2012
DOI:10.1021/mp300193f
The particular characteristics of the tumor microenvironment have the potential to strongly promote tumor growth, metastasis and angiogenesis and induce drug resistance. Therefore, the development of effective, systemic therapeutic approaches specifically based on the tumor microenvironment is highly desirable. Hypoxia-inducible factor-1α (HIF-1α) is an attractive therapeutic target because it is a key transcription factor in tumor development and only accumulates in hypoxic tumors. We report here that a cationic mixed micellar nanoparticle (MNP) consisting of amphiphilic block copolymers poly(ε-caprolactone)-block-poly(2-aminoethylethylene phosphate) (PCL29-b-PPEEA21) and poly(ε-caprolactone)-block-poly(ethylene glycol) (PCL40-b-PEG45) was a suitable carrier for HIF-1α siRNA to treat hypoxic tumors, which showed an average diameter of 58.0 ± 3.4 nm. The complex MNPsiRNA, formed by the interaction of MNP and siRNA, was transfected into PC3 prostate cancer cells efficiently, while the inhibition of HIF-1α expression by MNP loaded with HIF-1α siRNA (MNPsiHIF) blocked PC3 cell proliferation, suppressed cell migration and disturbed angiogenesis under in vitro hypoxic mimicking conditions. It was further demonstrated that systemic delivery of MNPsiHIF effectively inhibited tumor growth in a PC3 prostate cancer xenograft murine model without activating innate immune responses. Moreover, delivery of MNPsiHIF sensitized PC3 tumor cells to doxorubicin chemotherapy in vitro and in vivo by downregulating MDR1 gene expression which was induced by hypoxia. The underlying concept of use of MNPsiHIF to block HIF-1α holds promise as an example of a clinical approach using specific siRNA therapy for cancer treatment aimed at the hypoxic tumor microenvironment.Keywords: cancer therapy; HIF-1 Alpha; hypoxic tumor microenvironment; micellar nanoparticles; siRNA delivery;
Co-reporter:Xian-Zhu Yang, Shuang Dou, Yu-Cai Wang, Hong-Yan Long, Meng-Hua Xiong, Cheng-Qiong Mao, Yan-Dan Yao, and Jun Wang
ACS Nano 2012 Volume 6(Issue 6) pp:4955
Publication Date(Web):May 30, 2012
DOI:10.1021/nn300500u
The clinical success of therapeutics of small interfering RNA (siRNA) is still hindered by its delivery systems. Cationic polymer or lipid-based vehicles as the major delivery systems of siRNA cannot sufficiently satisfy siRNA therapeutic applications. It is hypothesized that cationic lipid–polymer hybrid nanoparticles may take advantage of both polymeric and lipid-based nanoparticles for siRNA delivery, while diminishing the shortcomings of both. In this study, cationic lipid–polymer hybrid nanoparticles were prepared by a single-step nanoprecipitation of a cationic lipid (N,N-bis(2-hydroxyethyl)-N-methyl-N-(2-cholesteryloxycarbonyl aminoethyl) ammonium bromide, BHEM-Chol) and amphiphilic polymers for systemic delivery of siRNA. The formed hybrid nanoparticles comprised a hydrophobic polylactide core, a hydrophilic poly(ethylene glycol) shell, and a cationic lipid monolayer at the interface of the core and the shell. Such hybrid nanoparticles exhibited excellent stability in serum and showed significantly improved biocompatibility compared to that of pure BHEM-Chol particles. The hybrid nanoparticles were capable of delivering siRNA into BT474 cells and facilitated the escape of loaded siRNA from the endosome into the cytoplasm. The hybrid nanoparticles carrying polo-like kinase 1 (Plk1)-specific siRNA (siPlk1) remarkably and specifically downregulated expression of the oncogene Plk1 and induced cancer cell apoptosis both in vitro and in vivo and significantly suppressed tumor growth following systemic administration. We demonstrate that this system is stable, nontoxic, highly efficient, and easy to scale up, bringing the clinical application of siRNA therapy one important step closer to reality.Keywords: cancer therapy; cationic lipid−polymer hybrid nanoparticles; polo-like kinase 1; single-step assembly; siRNA delivery
Co-reporter:Shuang Dou;Jian-bin Ruan;Peter Zhang;Jun Wang;Mu-sheng Zeng;Yan-dan Yao;Feng-xi Su;Feng-yan Yu;Cheng-qiong Mao;Qiang Liu;Tian-meng Sun;Jian-ye Zang;Erwei Song;Song-yin Huang;Jia-ning Chen;Judy Lieberman;Ling Lin
Science Translational Medicine 2012 Volume 4(Issue 130) pp:130ra48
Publication Date(Web):18 Apr 2012
DOI:10.1126/scitranslmed.3003601

Antibody-mediated delivery of anticancer siRNAs suppresses Her2+ breast cancer growth and metastasis.

Co-reporter:Yuanzeng Min;Cheng-Qiong Mao;Siming Chen;Guolin Ma; Jun Wang; Yangzhong Liu
Angewandte Chemie 2012 Volume 124( Issue 27) pp:6846-6851
Publication Date(Web):
DOI:10.1002/ange.201201562
Co-reporter:Yuanzeng Min;Cheng-Qiong Mao;Siming Chen;Guolin Ma; Jun Wang; Yangzhong Liu
Angewandte Chemie International Edition 2012 Volume 51( Issue 27) pp:6742-6747
Publication Date(Web):
DOI:10.1002/anie.201201562
Co-reporter:Xian-Zhu Yang, Jin-Zhi Du, Shuang Dou, Cheng-Qiong Mao, Hong-Yan Long, and Jun Wang
ACS Nano 2012 Volume 6(Issue 1) pp:771
Publication Date(Web):December 3, 2011
DOI:10.1021/nn204240b
Drug delivery systems for cancer therapy usually need to be sterically stabilized by a poly(ethylene glycol) (PEG) layer during blood circulation to minimize nonspecific interactions with serum components. However, PEGylation significantly reduces cellular uptake of the delivery systems after they accumulate at the tumor site, which markedly impairs the in vivo antitumor efficiency. Here, we develop a ternary small interfering RNA (siRNA) delivery system with tumor acidity-activated sheddable PEG layer to overcome the challenge. The sheddable nanoparticle is fabricated by introducing a tumor acidity-responsive PEGylated anionic polymer to the surface of positively charged polycation/siRNA complexes via electrostatic interaction. We show clear evidence that introducing the PEGylated anionic polymer to the surface of a nanoparticle markedly reduces its nonspecific interactions with protein. We further demonstrate that the nanoparticle is capable of deshielding the PEG layer at the slightly acidic tumor extracellular microenvironment to facilitate the delivery of siRNA to the tumor cells after accumulation at the tumor site. Accordingly, this promotes the RNA-interfering efficiencies and enhances the inhibition of tumor growth. Such delivery system with the ability to deshield the PEG layer at the target tissues has remarkable potential in cancer therapy.Keywords: nanotechnology; pH-responsive; sheddable nanoparticles; siRNA delivery; tumor acidity
Co-reporter:Jin-Zhi Du ; Xiao-Jiao Du ; Cheng-Qiong Mao ;Jun Wang
Journal of the American Chemical Society 2011 Volume 133(Issue 44) pp:17560-17563
Publication Date(Web):October 10, 2011
DOI:10.1021/ja207150n
Efficient delivery of therapeutics into tumor cells to increase the intracellular drug concentration is a major challenge for cancer therapy due to drug resistance and inefficient cellular uptake. Herein, we have designed a tailor-made dual pH-sensitive polymer–drug conjugate nanoparticulate system to overcome the challenges. The nanoparticle is capable of reversing its surface charge from negative to positive at tumor extracellular pH (∼6.8) to facilitate cell internalization. Subsequently, the significantly increased acidity in subcellular compartments such as the endosome (∼5.0) further promotes doxorubicin release from the endocytosed drug carriers. This dual pH-sensitive nanoparticle has showed enhanced cytotoxicity in drug-resistant cancer stem cells, indicating its great potential for cancer therapy.
Co-reporter:Yu-Cai Wang, Feng Wang, Tian-Meng Sun, and Jun Wang
Bioconjugate Chemistry 2011 Volume 22(Issue 10) pp:1939
Publication Date(Web):August 25, 2011
DOI:10.1021/bc200139n
Multidrug resistance (MDR) is a major impediment to the success of cancer chemotherapy. The intracellular accumulation of drug and the intracellular release of drug molecules from the carrier could be the most important barriers for nanoscale carriers in overcoming MDR. We demonstrated that the redox-responsive micellar nanodrug carrier assembled from the single disulfide bond-bridged block polymer of poly(ε-caprolactone) and poly(ethyl ethylene phosphate) (PCL-SS-PEEP) achieved more drug accumulation and retention in MDR cancer cells. Such drug carrier rapidly released the incorporated doxorubicin (DOX) in response to the intracellular reductive environment. It therefore significantly enhanced the cytotoxicity of DOX to MDR cancer cells. It was demonstrated that nanoparticular drug carrier with either poly(ethylene glycol) or poly(ethyl ethylene phosphate) (PEEP) shell increased the influx but decreased the efflux of DOX by the multidrug resistant MCF-7/ADR breast cancer cells, in comparison with the direct incubation of MCF-7/ADR cells with DOX, which led to high cellular retention of DOX. Nevertheless, nanoparticles bearing PEEP shell exhibited higher affinity to the cancer cells. The shell detachment of the PCL-SS-PEEP nanoparticles caused by the reduction of intracellular glutathione significantly accelerated the drug release in MCF-7/ADR cells, demonstrated by the flow cytometric analyses, which was beneficial to the entry of DOX into the nuclei of MCF-7/ADR cells. It therefore enhanced the efficiency in overcoming MDR of cancer cells, which renders the redox-responsive nanoparticles promising in cancer therapy.
Co-reporter:Xi-Qiu Liu, Wen-Jing Song, Tian-Meng Sun, Pei-Zhuo Zhang, and Jun Wang
Molecular Pharmaceutics 2011 Volume 8(Issue 1) pp:250-259
Publication Date(Web):December 7, 2010
DOI:10.1021/mp100315q
MiRNAs are viable therapeutic targets for cancer therapy, but the targeted delivery of miRNA or its anti-miRNA antisense oligonucleotides (AMOs) remains a challenge. We report here a PEGylated LPH (liposome-polycation-hyaluronic acid) nanoparticle formulation modified with cyclic RGD peptide (cRGD) for specific and efficient delivery of AMO into endothelial cells, targeting αvβ3 integrin present on the tumor neovasculature. The nanoparticles effectively delivered anti-miR-296 AMO to the cytoplasm and downregulated the target miRNA in human umbilical vein endothelial cells (HUVECs), which further efficiently suppressed blood tube formulation and endothelial cell migration, owing to significant upregulation of hepatocyte growth factor-regulated tyrosine kinase substrate (HGS), whereas nanoparticles without cRGD modification showed only little AMO uptake and miRNA silencing activity. In vivo assessment of angiogenesis using Matrigel plug assay also demonstrated that cRGD modified LPH nanoparticles have potential for antiangiogenesis in miRNA therapeutics. With the delivery of anti-miR-296 AMO by targeted nanoparticles, significant decrease in microvessel formulation within Matrigel was achieved through suppressing the invasion of CD31-positive cells into Matrigel and prompting HGS expression in angiogenic endothelial cells.Keywords: Anti-miRNA therapy; antiangiogenesis; cRGD; targeted delivery;
Co-reporter:JinZhi Du;LingYan Tang;YouYong Yuan;Jun Wang
Science China Chemistry 2011 Volume 54( Issue 2) pp:351-358
Publication Date(Web):2011 February
DOI:10.1007/s11426-010-4210-x
Cyclic phosphoester monomer ethyl ethylene phosphate (EEP) modified poly(ethylenimine) (PEI), denoted as PEI-EEP, was developed for gene delivery. Three PEI-EEP polymers were synthesized and their structures were characterized by 1H and 31P NMR methods. All the PEI-EEP polymers could condense DNA efficiently at N/P ratios higher than 0.5/1. The physiochemical characteristics of PEI-EEP/DNA complexes were analyzed by particle size and zeta potential measurements. The particle sizes of complexes were around 160–250 nm, and their zeta potentials were around 30–45 mV at the N/P ratios ranging from 10/1 to 50/1. In vitro cell viability and transfection ability were evaluated in HEK293 and HeLa cells using PEI as the control. The cytotoxicity of PEI-EEP and PEI-EEP/DNA complexes was lower than that of PEI and its complexes with DNA. The transfection efficiency of PEI-EEP/DNA complexes was correlated to modification degrees with phosphoester. When the modification of phosphoester to PEI was moderate, the PEI-EEP1/DNA and PEI-EEP2/DNA complexes exhibited comparable or even higher transfection ability than PEI/DNA complex at its optimal N/P ratio in the absence of serum. However, transfection efficiency of PEI-EEP3 reduced dramatically. More importantly, the PEI-EEP exhibited higher transfection efficiency in the presence of 10% serum than that without serum. Therefore, PEI-EEP polymers may be attractive vectors for non-viral gene therapy.
Co-reporter:You-Yong Yuan, Jun Wang
Colloids and Surfaces B: Biointerfaces 2011 Volume 85(Issue 1) pp:81-85
Publication Date(Web):15 June 2011
DOI:10.1016/j.colsurfb.2010.10.044
This work reports temperature-induced morphological change of ABC 3-miktoarm star terpolymer assemblies in aqueous solution. The terpolymer (MPEG)(PCL)(PPE) is composed of hydrophilic monomethoxy poly(ethylene glycol) (MPEG), hydrophobic poly(ɛ-caprolactone) (PCL) and thermosensitive polyphosphoester (PPE) chains, emanating from a central junction point. It is thermosensitive in aqueous solution, forming spherical micelles at lower temperature, which transition to short nano-rod morphology at temperature higher than the cloud point. The temperature induced morphological transition of this biodegradable miktoarm star terpolymer shows that it has potential in stimulus-controlled drug delivery applications.Graphical abstractResearch highlights▶ Successful preparation of the thermo-responsive miktoarm star terpolymer. ▶ Self-assembly of the terpolymer into micelle in aqueous solution. ▶ Temperature-induced morphological change of the terpolymer from micelle into nano-rod.
Co-reporter:Cheng-Qiong Mao, Jin-Zhi Du, Tian-Meng Sun, Yan-Dan Yao, Pei-Zhuo Zhang, Er-Wei Song, Jun Wang
Biomaterials 2011 32(11) pp: 3124-3133
Publication Date(Web):
DOI:10.1016/j.biomaterials.2011.01.006
Co-reporter:Yu-Cai Wang;You-Yong Yuan;Feng Wang;Jun Wang
Journal of Polymer Science Part A: Polymer Chemistry 2011 Volume 49( Issue 2) pp:487-494
Publication Date(Web):
DOI:10.1002/pola.24462

Abstract

We report a series of biocompatible and biodegradable block copolymers of poly(ε-caprolactone) with “clickable” polyphosphoester (PPE). The block copolymers are synthesized through controlled ring-opening polymerization of five-membered cyclic phosphoester monomer, propargyl ethylene phosphate (PAEP), initiated with poly(ε-caprolactone) macroinitiator. The polymerization followed first-order kinetics with living polymerization characteristics, thus the molecular weight and composition of copolymers are tunable by adjusting the feed ratio of PAEP monomer to macroinitiator. Azide-functionalized poly(ethylene glycol) has been grafted to the copolymer to demonstrate the reactive feasibility by Cu(I)-catalyzed “click” chemistry of azides and alkynes, generating “brush-coil” polymers. The mild conditions associated with the click reaction are shown to be compatible with poly(ε-caprolactone) and PPE backbones, rendering the click reaction a generally useful method for grafting numerous types of functionality onto the block copolymers. The block copolymers also show good biocompatibility to cells, suggesting their suitability for a range of biomaterial applications. © 2010 Wiley Periodicals, Inc. J Polym Sci Part A: Polym Chem, 2011

Co-reporter:Tian-Meng Sun, Jin-Zhi Du, Yan-Dan Yao, Cheng-Qiong Mao, Shuang Dou, Song-Yin Huang, Pei-Zhuo Zhang, Kam W. Leong, Er-Wei Song, and Jun Wang
ACS Nano 2011 Volume 5(Issue 2) pp:1483
Publication Date(Web):January 4, 2011
DOI:10.1021/nn103349h
Combination of two or more therapeutic strategies with different mechanisms can cooperatively prohibit cancer development. Combination of chemotherapy and small interfering RNA (siRNA)-based therapy represents an example of this approach. Hypothesizing that the chemotherapeutic drug and the siRNA should be simultaneously delivered to the same tumoral cell to exert their synergistic effect, the development of delivery systems that can efficiently encapsulate two drugs and successfully deliver payloads to targeted sites via systemic administration has proven to be challenging. Here, we demonstrate an innovative “two-in-one” micelleplex approach based on micellar nanoparticles of a biodegradable triblock copolymer poly(ethylene glycol)-b-poly(ε-caprolactone)-b-poly(2-aminoethyl ethylene phosphate) to systemically deliver the siRNA and chemotherapeutic drug. We show clear evidence that the micelleplex is capable of delivering siRNA and paclitaxel simultaneously to the same tumoral cells both in vitro and in vivo. We further demonstrate that systemic administration of the micelleplex carrying polo-like kinase 1 (Plk1) specific siRNA and paclitaxel can induce a synergistic tumor suppression effect in the MDA-MB-435s xenograft murine model, requiring a thousand-fold less paclitaxel than needed for paclitaxel monotherapy delivered by the micelleplex and without activation of the innate immune response or generation of carrier-associated toxicity.Keywords (keywords): cancer therapy; codelivery; micelleplex; nanoparticle; siRNA delivery; synergistic effect
Co-reporter:Feng Wang, Yu-Cai Wang, Shuang Dou, Meng-Hua Xiong, Tian-Meng Sun, and Jun Wang
ACS Nano 2011 Volume 5(Issue 5) pp:3679
Publication Date(Web):April 4, 2011
DOI:10.1021/nn200007z
Multidrug resistance (MDR) is a major impediment to the success of cancer chemotherapy. Through the development of a drug delivery system that tethers doxorubicin onto the surface of gold nanoparticles with a poly(ethylene glycol) spacer via an acid-labile linkage (DOX-Hyd@AuNPs), we have demonstrated that multidrug resistance in cancer cells can be significantly overcome by a combination of highly efficient cellular entry and a responsive intracellular release of doxorubicin from the gold nanoparticles in acidic organelles. DOX-Hyd@AuNPs achieved enhanced drug accumulation and retention in multidrug resistant MCF-7/ADR cancer cells when it was compared with free doxorubicin. It released doxorubicin in response to the pH of acidic organelles following endocytosis, opposite to the noneffective drug release from doxorubicin-tethered gold nanoparticles via the carbamate linkage (DOX-Cbm@AuNPs), which was shown by the recovered fluorescence of doxorubicin from quenching due to the nanosurface energy transfer between the doxorubicinyl groups and the gold nanoparticles. DOX-Hyd@AuNPs therefore significantly enhanced the cytotoxicity of doxorubicin and induced elevated apoptosis of MCF-7/ADR cancer cells. With a combined therapeutic potential and ability to probe drug release, DOX-Hyd@AuNPs represent a model with dual roles in overcoming MDR in cancer cells and probing the intracellular release of drug from its delivery system.Keywords: doxorubicin; drug delivery; gold nanoparticles; intracellular drug release; multidrug resistance; nanosurface energy transfer
Co-reporter:Yu-Cai Wang, Juan Wu, Yang Li, Jin-Zhi Du, You-Yong Yuan and Jun Wang  
Chemical Communications 2010 vol. 46(Issue 20) pp:3520-3522
Publication Date(Web):08 Apr 2010
DOI:10.1039/C002620D
We report the preparation of biodegradable nanoscopic hydrogels and their application for targeted drug delivery. The nanogel is synthesized in a template-free method by photo-crosslinking salt-induced polymer assemblies. With convenient incorporation of targeted lactosyl moieties, the nanogels efficiently deliver doxorubicin to HepG2 cells through receptor-mediated internalization.
Co-reporter:Yu-Cai Wang;Yang Li;Tian-Meng Sun;Meng-Hua Xiong;Juan Wu;Yi-Yan Yang;Jun Wang
Macromolecular Rapid Communications 2010 Volume 31( Issue 13) pp:1201-1206
Publication Date(Web):
DOI:10.1002/marc.200900863
Co-reporter:Jin-Zhi Du;Tian-Meng Sun;Wen-Jing Song;Juan Wu;Jun Wang
Angewandte Chemie International Edition 2010 Volume 49( Issue 21) pp:3621-3626
Publication Date(Web):
DOI:10.1002/anie.200907210
Co-reporter:Jin-Zhi Du;Tian-Meng Sun;Wen-Jing Song;Juan Wu;Jun Wang
Angewandte Chemie 2010 Volume 122( Issue 21) pp:3703-3708
Publication Date(Web):
DOI:10.1002/ange.200907210
Co-reporter:You-Yong Yuan, Qing Du, Yu-Cai Wang and Jun Wang
Macromolecules 2010 Volume 43(Issue 4) pp:1739-1746
Publication Date(Web):January 27, 2010
DOI:10.1021/ma9023763
Amphiphilic centipede-like brush copolymers with biodegradable poly(ε-caprolactone) and poly(ethyl ethylene phosphate) side segments were prepared by a one-pot syntheses strategy. The syntheses combined ring-opening polymerization of 2-ethoxy-2-oxo-1,3,2-dioxaphospholane through a “grafting from” strategy and “click” reaction with α-propargyl-ω-acetyl-poly(ε-caprolactone) through a “grafting to” strategy, using multifunctional poly(tert-butyl methacrylate)-co-poly(2-hydroxy-3-azidopropyl methacrylate) that bears hydroxyl and azide groups from junction points. The reactions are controllable, and the structure of obtained centipede-like brush copolymer is well characterized. These brush copolymers are amphiphilic and self-assemble into spherical micellar structure in aqueous solution with critical aggregation concentration around 10−3 mg mL−1 and average diameters of 50−90 nm. Such micelles formed from centipede-like brush copolymers can be used as drug carriers for biomedical applications.
Co-reporter:Juan Wu, Xi-Qiu Liu, Yu-Cai Wang and Jun Wang  
Journal of Materials Chemistry A 2009 vol. 19(Issue 42) pp:7856-7863
Publication Date(Web):16 Sep 2009
DOI:10.1039/B908768K
Biodegradable nanogels with tunable sizes, synthesized by a template-free method, as potential carriers for drug delivery are reported. The nanogels are obtained by crosslinking thermo-induced nanoparticles, with subsequent swelling at low temperatures, which were based on a thermosensitive and biocompatible triblock copolymer composed of poly(ethylene glycol) and poly(ethyl ethylene phosphate). The nanogels loaded with doxorubicin are efficiently taken up by A549 tumor cells and the drug could be released intracellularly, demonstrated by flow cytometric analyses and confocal laser scanning microscope observations. It results in enhanced growth inhibition activity to tumor cells in comparison with free doxorubicin treatment. These polyphosphoester and poly(ethylene glycol) based nanogels are biocompatible and biodegradable, rendering potential for drug delivery applications.
Co-reporter:Ling-Yan Tang, Yu-Cai Wang, Yang Li, Jin-Zhi Du and Jun Wang
Bioconjugate Chemistry 2009 Volume 20(Issue 6) pp:1095
Publication Date(Web):May 13, 2009
DOI:10.1021/bc900144m
Aiming at development of a micellar nanoparticle system for intracellular drug release triggered by glutathione in tumor cells, a disulfide-linked biodegradable diblock copolymer of poly(ε-caprolactone) and poly(ethyl ethylene phosphate) was synthesized. It formed biocompatible micelles loaded with doxorubicin in aqueous solution but detached the shell material under glutathione stimulus, resulting in rapid drug release with destruction of micellar structure. These glutathione-sensitive micelles also rapidly released the drug molecules intracellularly and led to enhanced growth inhibition to A549 tumor cells, suggesting that this nanoparticle system may have potential for improving drug delivery efficacy.
Co-reporter:Yu-Cai Wang;You-Yong Yuan;Jin-Zhi Du;Xian-Zhu Yang;Jun Wang
Macromolecular Bioscience 2009 Volume 9( Issue 12) pp:1154-1164
Publication Date(Web):
DOI:10.1002/mabi.200900253
Co-reporter:Jin-Zhi Du, Ling-Yan Tang, Wen-Jing Song, Yue Shi and Jun Wang
Biomacromolecules 2009 Volume 10(Issue 8) pp:
Publication Date(Web):July 8, 2009
DOI:10.1021/bm900345m
Brush polymers PHEMA-g-(PCL-b-PEG) with poly(2-hydroxyethyl methacrylate) (PHEMA) as the backbone and poly(ε-caprolactone)-b-poly(ethylene glycol) (PCL-b-PEG) block copolymers as side chains were synthesized and evaluated as drug delivery vehicles. Two brush polymers were synthesized, and their structures were confirmed by gel permeation chromatography analyses and 1H NMR measurements. The brush polymers self-assembled into micelles in aqueous solution, and the critical micellization concentrations of brush polymers were 2-fold lower than that of the linear diblock copolymer PCL-b-PEG with structure similar to that of the grafted side chains of brush polymers, indicating the higher aqueous stability of brush polymer micelles. The micelles were spherical with average diameters below 100 nm. Brush polymer micelles exhibited higher loading doxorubicin capacity compared with micelles from linear PCL-b-PEG block copolymer by the dialysis method, and the burst doxorubicin release from the brush polymer micelles was significantly suppressed. Doxorubicin-loaded brush polymer micelles can be effectively internalized by A549 human lung carcinoma cells and slowly released the encapsulated drug molecules as demonstrated by the drug accumulation in cytoplasm, which was opposite to free doxorubicin, which accumulated rapidly in the cell nuclei.
Co-reporter:Feng Wang, Yu-Cai Wang, Li-Feng Yan, Jun Wang
Polymer 2009 50(21) pp: 5048-5054
Publication Date(Web):
DOI:10.1016/j.polymer.2009.09.007
Co-reporter:Jing Cheng;Jun Wang
Science China Chemistry 2009 Volume 52( Issue 7) pp:961-968
Publication Date(Web):2009 July
DOI:10.1007/s11426-009-0121-0
Biodegradable and amphiphilic triblock copolymers poly(ethyl ethylene phosphate)-poly(3-hydroxy-butyrate)-poly(ethyl ethylene phosphate) (PEEP-b-PHB-b-PEEP) have been successfully synthesized through ring-opening polymerization. The structures are confirmed by gel permeation chromatography and NMR analyses. Crystallization investigated by X-ray diffraction reveals that the block copolymer with higher content of poly(ethyl ethylene phosphate) (PEEP) is more amorphous, showing decreased crystallizability. The obtained copolymers self-assemble into biodegradable nanoparticles with a core-shell micellar structure in aqueous solution, verified by the probe-based fluorescence measurements and transmission electronic microscopy (TEM) observation. The hydrophobic poly(3-hydroxybutyrate) (PHB) block serves as the core of the micelles and the micelles are stabilized by the hydrophilic PEEP block. The size and size distribution are related to the compositions of the copolymers. Paclitaxel (PTX) has been encapsulated into the micelles as a model drug and a sustained drug release from the micelles is observed. MTT assay also demonstrates that the block copolymers are biocompatible, rendering these copolymers attractive for drug delivery.
Co-reporter:You-Yong Yuan, Xi-Qiu Liu, Yu-Cai Wang and Jun Wang
Langmuir 2009 Volume 25(Issue 17) pp:10298-10304
Publication Date(Web):June 1, 2009
DOI:10.1021/la901120x
Aqueous dispersions of thermosensitive gold nanoparticles protected by diblock copolymers of poly(ethylene glycol) and polyphosphoester were prepared and studied. Diblock copolymers MPEG-b-P(EEP-co-PEP) with different compositions that are composed of monomethoxy poly(ethylene glycol), random copolymer of ethyl ethylene phosphate (EEP), and isopropyl ethylene phosphate (PEP) were synthesized by ring-opening polymerization in bulk. Thioctic acid was then conjugated to the terminal hydroxyl group of the polyphosphoester block by esterification. Gold nanoparticles were then prepared by a one-step method and showed core−shell structure with an average gold core diameter of about 10 nm surrounded by a MPEG-b-P(EEP-co-PEP) shell with a thickness of about 30 nm. These polymer stabilized gold nanoparticles are reversibly thermosensitive in aqueous medium, exhibiting tunable collapse temperatures which are dependent on the composition of the diblock copolymers. Methyl tetrazolium (MTT) assay against HEK 293 cells demonstrated that these gold nanoparticles are with good biocompatibility. These gold nanoparticles protected by thermosensitive diblock copolymers with tunable collapse temperature are expected to be useful for biomedical applications.
Co-reporter:Meng-Hua Xiong, Juan Wu, Yu-Cai Wang, Lai-Sheng Li, Xiao-Bing Liu, Guang-Zhao Zhang, Li-Feng Yan and Jun Wang
Macromolecules 2009 Volume 42(Issue 4) pp:893-896
Publication Date(Web):January 21, 2009
DOI:10.1021/ma802688y
Co-reporter:Yu-Cai Wang;Hai Xia;Xian-Zhu Yang;Jun Wang
Journal of Polymer Science Part A: Polymer Chemistry 2009 Volume 47( Issue 22) pp:6168-6179
Publication Date(Web):
DOI:10.1002/pola.23660

Abstract

Pluronic analogs based on block copolymers of poly(propylene oxide) and poly(ethyl ethylene phosphate) (PEEP-PPO-PEEP) were synthesized, and the thermoresponsive behavior, including aggregation at low concentration and gelation at high concentration were studied. At lower concentrations up to 10 wt %, thermo-induced aggregation of PEEP-PPO-PEEP was demonstrated by UV-vis absorbance measurements using 1,6-diphenyl-1,3,5-hexatriene as a probe. Microthermal analyzes showed symmetrically endothermic and exothermic thermograms during the thermo-induced aggregation and de-association processes, which was also associated with the dehydration and rehydration of PPO blocks, as revealed by the variable temperature NMR measurements. Thermo-induced aggregation with the increased temperatures was also observed by dynamic light scattering. At higher concentration from 20 to 40 wt %, the aqueous solution of PEEP-PPO-PEEP underwent thermo-induced phase transitions from a clear solution to a turbid solution, then to opaque gel and syneresis phases, depending on the molecular weights of PEEP blocks. Such a thermoresponsive hydrogel was used for doxorubicin incorporation. Sustained release of drug was achieved from the gel, demonstrating the polyphosphoester-based Pluronic analogs' potential for biomedical applications. © 2009 Wiley Periodicals, Inc. J Polym Sci Part A: Polym Chem 47: 6168–6179, 2009

Co-reporter:Yu-Cai Wang, Yang Li, Xian-Zhu Yang, You-Yong Yuan, Li-Feng Yan and Jun Wang
Macromolecules 2009 Volume 42(Issue 8) pp:3026-3032
Publication Date(Web):March 31, 2009
DOI:10.1021/ma900288t
The development of thermoresponsive and biodegradable polymer nanoparticles with tunable thermosensitivity and biocompatibility is of great interest, especially for in vivo biomedical applications. In this study, biodegradable polymer micellar nanoparticles with tunable thermosensitivity are reported. They are based on various biodegradable block copolymers of hydrophobic poly(ε-caprolactone) and thermosensitive polyphosphoesters, obtained through poly(ε-caprolactone)/stannous octoate coinitiated random ring-opening polymerization of cyclic phosphoester monomers. The thermosensitive micellar shells of nanoparticles turn out to be more hydrophobic and result in aggregates in aqueous solution when the temperature is higher than their lower critical solution temperature (LCST). The phase transition temperatures can be adjusted by controlling the molecular weights and the compositions of biodegradable polyphosphoester blocks. Decreased molecular weights of poly(ethyl ethylene phosphate) (PEEP) lead to higher LCST, whereas copolymerization of PEEP with more hydrophobic component results in lower LCST and sharper response. It has also been observed that increased sodium chloride concentration in micelle solution leads to lower responsive temperature. Therefore, the thermosensitivity of micelles can be conveniently adjusted over a wide temperature range. With good biocompatibility and tunable thermosensitivity, these biodegradable polymer-based nanoparticles are potential stimuli-responsive materials for biomedical applications.
Co-reporter:Xian-Zhu Yang, Tian-Meng Sun, Shuang Dou, Juan Wu, Yu-Cai Wang and Jun Wang
Biomacromolecules 2009 Volume 10(Issue 8) pp:
Publication Date(Web):July 8, 2009
DOI:10.1021/bm900390k
Surface modification is often needed in tissue engineering to enhance the interaction between cells and synthetic materials and improve the cytocompatibility and cellular functions. In this study, block copolymers of poly(l-lactic acid) and poly(ethyl ethylene phosphate) (PLLA-b-PEEP) were synthesized and used to modify the PLLA surface via a spin-coating process, to understand whether surface modification with polyphosphoester-based polymer will be osteoinductive for potential bone tissue engineering applications. X-ray photoelectron spectra measurements revealed that phosphorus atomic compositions after surface modification increased from 2.09% to 4.39% with increasing PEEP length of PLLA-b-PEEP from 58 to 224 units, which also led to a more hydrophilic surface property compared with unmodified PLLA. The initial osteoblast attachment and proliferation on the modified surfaces were significantly enhanced. Moreover, cellular alkaline phosphatase activity and mineral calcium depositions were also promoted by PEEP modification. The gene expression determined by reverse transcription polymerase chain reaction further revealed that type I collagen and osteocalcin expression were upregulated in osteoblasts cultured on the modified surfaces, indicating that PEEP modification might be potentially osteoinductive and favorable for further application in bone tissue engineering.
Co-reporter:Yu-Cai Wang, Ling-Yan Tang, Yang Li and Jun Wang
Biomacromolecules 2009 Volume 10(Issue 1) pp:
Publication Date(Web):December 2, 2008
DOI:10.1021/bm800808q
Novel thermoresponsive block copolymers of poly(ethylene glycol) and polyphosphoester were synthesized, and the thermo-induced self-assembly, biocompatibility, and hydrolytic degradation behavior were studied. The block copolymers with various molecular weights and compositions were synthesized through ring-opening polymerization of 2-ethoxy-2-oxo-1,3,2-dioxaphospholane (EEP) and 2-isopropoxy-2-oxo-1,3,2-dioxaphospholane (PEP) using poly(ethylene glycol) monomethyl ether (mPEG) as the initiator and stannous octoate as the catalyst. The obtained block polymers exhibited thermo-induced self-assembly behavior, demonstrated by dynamic light scattering and UV−vis measurements using 1,6-diphenyl-1,3,5-hexatriene as the probe. It was found that the critical aggregation temperature (CAT) of the block copolymers shifted to higher temperature with increased molecular weight of mPEG, while copolymerization with more hydrophobic monomer PEP led to lower transition temperature; thus, the CAT can be conveniently adjusted. The block copolymers did not induce significant hemolysis and plasma protein precipitation. In vitro MTT and live/dead staining assays indicated they are biocompatible, and the biocompatibility was further demonstrated in vivo by the absence of local acute inflammatory response in mouse muscle following intramuscular injection. Unlike most frequently studied thermoresponsive poly(N-isopropylacrylamide), polyphosphoesters were hydrolytically degradable in aqueous solution that was proven by gel permeation chromatography and NMR analyses, and the degradation products were proven to be nontoxic to HEK293 cells. Therefore, with good biocompatibility and thermoresponsiveness, these biodegradable block copolymers of mPEG and polyphosphoesters are promising as stimuli-responsive materials for biomedical applications.
Co-reporter:Yu-Cai Wang, Xi-Qiu Liu, Tian-Meng Sun, Meng-Hua Xiong, Jun Wang
Journal of Controlled Release 2008 Volume 128(Issue 1) pp:32-40
Publication Date(Web):22 May 2008
DOI:10.1016/j.jconrel.2008.01.021
Cellular specific micellar systems from functional amphiphilic block copolymers are attractive for targeted intracellular drug delivery. In this study, we developed reactive micelles based on diblock copolymer of poly(ethyl ethylene phosphate) and poly(ɛ-caprolactone). The micelles were further surface conjugated with galactosamine to target asialoglycoprotein receptor (ASGP-R) of HepG2 cells. The size of micellar nanoparticles was about 70nm in diameter, and nanoparticles were negatively charged in aqueous solution. Through recognition between galactose ligands with ASGP-R of HepG2 cells, cell surface binding and internalization of galactosamine-conjugated micelles were significantly promoted, which were demonstrated by flow cytometric analyses using rhodamine 123 fluorescent dye. Paclitaxel-loaded micelles with galactose ligands exhibited comparable activity to free paclitaxel in inhibiting HepG2 cell proliferation, in contrast to the poor inhibition activity of micelles without galactose ligands particularly at lower paclitaxel doses. In addition, population of HepG2 cells arrested in G2/M phase was in positive response to paclitaxel dose when cells were incubated with paclitaxel-loaded micelles with galactosamine conjugation, which was against the performance of micelles without galactose ligand, owing to the ligand–receptor interaction. The surface functionalized micellar system is promising for specific anticancer drug transportation and intracellular drug release.
Co-reporter:Yu-Cai Wang, Ling-Yan Tang, Tian-Meng Sun, Chang-Hua Li, Meng-Hua Xiong and Jun Wang
Biomacromolecules 2008 Volume 9(Issue 1) pp:
Publication Date(Web):December 15, 2007
DOI:10.1021/bm700732g
A series of novel amphiphilic triblock copolymers of poly(ethyl ethylene phosphate) and poly(ϵ-caprolactone) (PEEP-PCL-PEEP) with various PEEP and PCL block lengths were synthesized and characterized. These triblock copolymers formed micelles composed of a hydrophobic core of poly(ϵ-caprolactone) (PCL) and a hydrophilic shell of poly(ethyl ethylene phosphate) (PEEP) in aqueous solution. The micelle morphology was spherical, determined by transmission electron microscopy. It was found that the size and critical micelle concentration values of the micelles depended on both hydrophobic PCL block length and PEEP hydrophilic block length. The in vitro degradation characteristics of the triblock copolymers were investigated in micellar form, showing that these copolymers were completely biodegradable under enzymatic catalysis of Pseudomonas lipase and phosphodiesterase I. These triblock copolymers were used for paclitaxel (PTX) encapsulation to demonstrate the potential in drug delivery. PTX was successfully loaded into the micelles, and the in vitro release profile was found to be correlative to the polymer composition. These biodegradable triblock copolymer micelles are potential as novel carriers for hydrophobic drug delivery.
Co-reporter:Xian-Zhu Yang;Ling-Yan Tang;Hai Xia;Jun Wang
Journal of Polymer Science Part A: Polymer Chemistry 2008 Volume 46( Issue 19) pp:
Publication Date(Web):
DOI:10.1002/pola.22951

Abstract

Aliphatic polyesters and polyphosphoesters (PPEs) have received much interest in medical applications due to their favorable biocompatibility and biodegradability. In this work, novel amphiphilic triblock copolymers of PPE and poly(L-lactic acid) (PLLA) with various compositions were synthesized and characterized. The blocky structure was confirmed by GPC analyses. These triblock copolymers formed micelles composed of hydrophobic PLLA core and hydrophilic PPE shell in aqueous solution. Critical micellization concentrations of these triblock copolymers were related to the polymer compositions. Incubation of micelles at neutral pH followed by GPC analyses revealed that these polymer micelles were hydrolysized and resulted in decreased molecular weights and small oligomers, whereas its degradation in basic and acid mediums was accelerated. MTT assay also demonstrated the biocompatibility against HEK293 cells. These biodegradable polymers are potential as drug carriers for biomedical application. © 2008 Wiley Periodicals, Inc. J Polym Sci Part A: Polym Chem 46: 6425–6434, 2008

Co-reporter:Wen-Jing Song, Jin-Zhi Du, Nan-Jun Liu, Shuang Dou, Jing Cheng and Jun Wang
Macromolecules 2008 Volume 41(Issue 19) pp:6935-6941
Publication Date(Web):September 3, 2008
DOI:10.1021/ma801043m
Novel diblock copolymers of poly(ε-caprolactone) and polyphosphoester bearing functional hydroxyl pendant groups, denoted as PCL-b-PHEP, were synthesized through ring-opening polymerization of functionalized cyclic phosphoester monomer using hydroxyl end-capped poly(ε-caprolactone) and Sn(Oct)2 as macroinitiator and catalyst, respectively. The chemical structure was proved by 1H, 13C, and 31P NMR, gel permeation chromatography (GPC), and Fourier transform infrared spectroscopy (FT-IR) analyses. These amphiphilic functionalized block copolymers could self-assemble into micellar or vesicular aggregates in aqueous solution, depending on the composition, which was demonstrated by transmission electron microscopy and confocal laser scanning microscope observations. The critical aggregation concentrations (CAC) of PCL-b-PHEP were also dependent on the composition, measured by the fluorescence probe technique. MTT assay for cytotoxicity of PCL-b-PHEP suggested these polymeric nanoparticles were biocompatible. Combining the advantages of poly(ε-caprolactone) and polyphosphoester with functional hydroxyl pendant groups for further biological modification, such amphiphilic block copolymers could potentially provide novel opportunities for design of drug delivery system and therapeutic application.
Co-reporter:You-Yong Yuan, Yu-Cai Wang, Jin-Zhi Du and Jun Wang
Macromolecules 2008 Volume 41(Issue 22) pp:8620-8625
Publication Date(Web):October 24, 2008
DOI:10.1021/ma801452n
Novel biodegradable amphiphilic ABC 3-miktoarm star terpolymers composed of poly(ε-caprolactone) (PCL), monomethoxy poly(ethylene glycol) (MPEG), and polyphosphoester (PPE) were synthesized by a combination of ring-opening polymerization and “click” chemistry. MPEG was first end-capped by epoxide ring, which was opened by sodium azide in the presence of ammonium chloride to give modified MPEG bearing reactive azide and hydroxyl groups (MPEG(−OH,−N3)). “Click” chemistry was then applied to conjugate α-propargyl-ω-acetyl-poly(ε-caprolactone) and MPEG(−OH,−N3), resulting in a diblock copolymer of MPEG and PCL with reactive hydroxyl groups at the junction point (MPEG(−OH)-b-PCL), which further initiated ring-opening polymerization of 2-ethoxy-2-oxo-1,3,2-dioxaphospholane (EEP) under the catalytic action of stannous octoate to obtain the desired well-defined (MPEG)(PCL)(PEEP) 3-miktoarm star terpolymers. Such terpolymers and their intermediates were characterized by 1H NMR, FT-IR, and gel permeation chromatography. These polymers are expected to be promising vehicles for drug delivery applications.
Co-reporter:Rong Sun, Song Shen, Yun-Jiao Zhang, Cong-Fei Xu, Zhi-Ting Cao, Long-Ping Wen, Jun Wang
Biomaterials (October 2016) Volume 103() pp:44-55
Publication Date(Web):October 2016
DOI:10.1016/j.biomaterials.2016.06.038
Cancer stem cells (CSCs) have garnered increasing attention over the past decade, as they are believed to play a crucial role in tumor initiation, progression and metastasis, relapse and drug resistance. Therapeutic strategies which simultaneously exterminate both bulk tumor cells and the rare CSC subpopulation may produce striking response and result in long-term tumor remission. Accumulating evidence provides insight into the function of autophagy in maintenance, plasticity and survival of CSCs. The role of autophagy in the susceptibility of breast CSCs to chemotherapeutics was investigated in the present work, reduced ‘stemness’ and increased susceptibility to chemotherapy drugs (doxorubicin, DOX and docetaxel, DTXL) were observed after chloroquine (CQ)-mediated autophagy inhibition in sorted ALDHhi cells of breast cancer cell line MDA-MB-231. We further proved that nanoparticle-mediated autophagy inhibition promoted the efficacy of chemotherapeutics against ALDHhi MDA-MB-231 cells in vitro. Administration of drug delivery systems significantly prolonged the circulation half-life and augmented enrichment of two different drugs in tumor tissues and ALDHhi cells. More importantly, compared with single treatment, the combined delivery systems NPCQ/NPDOX and NPCQ/DOX (NPCQ/NPDTXL and NPCQ/DTXL) showed most effective and persistent tumor growth inhibitory effect by eliminating bulk tumor cells as well as CSCs (p < 0.01) in an MDA-MB-231 orthotopic tumor murine model. Therefore, our research provides new insights into the nanoparticle-facilitated combination of autophagy inhibition and chemotherapy for effective therapy of breast cancer.
Co-reporter:Rong Sun, Song Shen, Yun-Jiao Zhang, Cong-Fei Xu, Zhi-Ting Cao, Long-Ping Wen, Jun Wang
Biomaterials (October 2016) Volume 103() pp:44-55
Publication Date(Web):October 2016
DOI:10.1016/j.biomaterials.2016.06.038
Co-reporter:Song Shen, Chun-Yang Sun, Xiao-Jiao Du, Hong-Jun Li, Yang Liu, Jin-Xing Xia, Yan-Hua Zhu, Jun Wang
Biomaterials (November 2015) Volume 70() pp:71-83
Publication Date(Web):November 2015
DOI:10.1016/j.biomaterials.2015.08.026
Co-reporter:Cong-Fei Xu, Hou-Bing Zhang, Chun-Yang Sun, Yang Liu, Song Shen, Xian-Zhu Yang, Yan-Hua Zhu, Jun Wang
Biomaterials (May 2016) Volume 88() pp:48-59
Publication Date(Web):May 2016
DOI:10.1016/j.biomaterials.2016.02.031
The design of ideal nanoparticle delivery systems should be capable of meeting the requirements of several stages of drug delivery, including prolonged circulation, enhanced accumulation and penetration in the tumor, facilitated cellular internalization and rapid release of the active drug in the tumor cells. However, among the current design strategies, meeting the requirements of one stage often conflicts with the other. Herein, a tumor pH-labile linkage-bridged block copolymer of poly(ethylene glycol) with poly(lacide-co-glycolide) (PEG-Dlinkm-PLGA) was used for siRNA delivery to fulfill all aforementioned requirements of these delivery stages. The obtained siRNA-encapsulating PEG-Dlinkm-PLGA nanoparticle gained efficiently prolonged circulation in the blood and preferential accumulation in tumor sites via the PEGylation. Furthermore, the PEG surface layer was detached in response to the tumor acidic microenvironment to facilitate cellular uptake, and the siRNA was rapidly released within tumor cells due to the hydrophobic PLGA layer. Hence, PEG-Dlinkm-PLGA nanoparticles met the requirements of several stages of drug delivery, and resulted in the enhanced therapeutic effect of the nanoparticular delivery systems.
Co-reporter:Cong-Fei Xu, Hou-Bing Zhang, Chun-Yang Sun, Yang Liu, Song Shen, Xian-Zhu Yang, Yan-Hua Zhu, Jun Wang
Biomaterials (May 2016) Volume 88() pp:48-59
Publication Date(Web):May 2016
DOI:10.1016/j.biomaterials.2016.02.031
Co-reporter:Cheng-Qiong Mao, Meng-Hua Xiong, Yang Liu, Song Shen, ... Jun Wang
Molecular Therapy (May 2014) Volume 22(Issue 5) pp:964-973
Publication Date(Web):1 May 2014
DOI:10.1038/mt.2014.18
The KRAS mutation is present in ∼20% of lung cancers and has not yet been effectively targeted for therapy. This mutation is associated with a poor prognosis in non-small-cell lung carcinomas (NSCLCs) and confers resistance to standard anticancer treatment drugs, including epidermal growth factor receptor tyrosine kinase inhibitors. In this study, we exploited a new therapeutic strategy based on the synthetic lethal interaction between cyclin-dependent kinase 4 (CDK4) downregulation and the KRAS mutation to deliver micellar nanoparticles (MNPs) containing small interfering RNA targeting CDK4 (MNPsiCDK4) for treatment in NSCLCs harboring the oncogenic KRAS mutation. Following MNPsiCDK4 administration, CDK4 expression was decreased, accompanied by inhibited cell proliferation, specifically in KRAS mutant NSCLCs. However, this intervention was harmless to normal KRAS wild-type cells, confirming the proposed mechanism of synthetic lethality. Moreover, systemic delivery of MNPsiCDK4 significantly inhibited tumor growth in an A549 NSCLC xenograft murine model, with depressed expression of CDK4 and mutational KRAS status, suggesting the therapeutic promise of MNPsiCDK4 delivery in KRAS mutant NSCLCs via a synthetic lethal interaction between KRAS and CDK4.
Co-reporter:Jin-Zhi Du, Cheng-Qiong Mao, You-Yong Yuan, Xian-Zhu Yang, Jun Wang
Biotechnology Advances (July–August 2014) Volume 32(Issue 4) pp:
Publication Date(Web):1 July 2014
DOI:10.1016/j.biotechadv.2013.08.002
pH-responsive nanoparticles (NPs) are currently under intense development as drug delivery systems for cancer therapy. Among various pH-responsiveness, NPs that are designed to target slightly acidic extracellular pH environment (pHe) of solid tumors provide a new paradigm of tumor targeted drug delivery. Compared to conventional specific surface targeting approaches, the pHe-targeting strategy is considered to be more general due to the common occurrence of acidic microenvironment in solid tumors. This review mainly focuses on the design and applications of pHe-activated NPs, with special emphasis on pHe-activated surface charge reversal NPs, for drug and siRNA delivery to tumors. The novel development of NPs described here offers great potential for achieving better therapeutic effects in cancer treatment.
Co-reporter:Zhi-Ting Cao, Zhi-Yao Chen, Chun-Yang Sun, Hong-Jun Li, Hong-Xia Wang, Qin-Qin Cheng, Zu-Qi Zuo, Ji-Long Wang, Yang-Zhong Liu, Yu-Cai Wang, Jun Wang
Biomaterials (July 2016) Volume 94() pp:9-19
Publication Date(Web):July 2016
DOI:10.1016/j.biomaterials.2016.04.001
Chemotherapy resistance has become a major challenge in the clinical treatment of lung cancer which is the leading cancer type for the estimated deaths. Recent studies have shown that nanoparticles as drug carriers can raise intracellular drug concentration by achieving effectively cellular uptake and rapid drug release, and therefore reverse the acquired chemoresistance of tumors. In this context, nanoparticles-based chemotherapy represents a promising strategy for treating malignancies with chemoresistance. In the present study, we developed cationic lipid assisted nanoparticles (CLAN) to deliver polylactide-cisplatin prodrugs to drug resistant lung cancer cells. The nanoparticles were formulated through self-assembly of a biodegradable poly(ethylene glycol)-block-poly(lactide) (PEG-PLA), a hydrophobic polylactide-cisplatin prodrug, and a cationic lipid. The cationic nanoparticles were proven to significantly improve cell uptake of cisplatin, leading to an increased DNA-Pt adduct and significantly promoted DNA damage in vitro. Moreover, our study reveals that cationic nanoparticles, although are slightly inferior in blood circulation and tumor accumulation, are more effective in blood vessel extravasation. The CLANs ultimately enhances the cellular drug availability and leads to the reversal of cisplatin resistance.
Co-reporter:Zhi-Ting Cao, Zhi-Yao Chen, Chun-Yang Sun, Hong-Jun Li, Hong-Xia Wang, Qin-Qin Cheng, Zu-Qi Zuo, Ji-Long Wang, Yang-Zhong Liu, Yu-Cai Wang, Jun Wang
Biomaterials (July 2016) Volume 94() pp:9-19
Publication Date(Web):July 2016
DOI:10.1016/j.biomaterials.2016.04.001
Co-reporter:Song Shen, Jin-Xing Xia, Jun Wang
Biomaterials (January 2016) Volume 74() pp:1-18
Publication Date(Web):January 2016
DOI:10.1016/j.biomaterials.2015.09.037
Circumstantial evidence suggests that most tumours are heterogeneous and contain a small population of cancer stem cells (CSCs) that exhibit distinctive self-renewal, proliferation and differentiation capabilities, which are believed to play a crucial role in tumour progression, drug resistance, recurrence and metastasis in multiple malignancies. Given that the existence of CSCs is a primary obstacle to cancer therapy, a tremendous amount of effort has been put into the development of anti-CSC strategies, and several potential approaches to kill therapeutically-resistant CSCs have been explored, including inhibiting ATP-binding cassette transporters, blocking essential signalling pathways involved in self-renewal and survival of CSCs, targeting CSCs surface markers and destroying the tumour microenvironment. Meanwhile, an increasing number of therapeutic agents (e.g. small molecule drugs, nucleic acids and antibodies) to selectively target CSCs have been screened or proposed in recent years. Drug delivery technology-based approaches hold great potential for tackling the limitations impeding clinical applications of CSC-specific agents, such as poor water solubility, short circulation time and inconsistent stability. Properly designed nanocarrier-based therapeutic agents (or nanomedicines) offer new possibilities of penetrating CSC niches and significantly increasing therapeutic drug accumulation in CSCs, which are difficult for free drug counterparts. In addition, intelligent nanomedicine holds great promise to overcome pump-mediated multidrug resistance which is driven by ATP and to decrease detrimental effects on normal somatic stem cells. In this review, we summarise the distinctive biological processes related to CSCs to highlight strategies against inherently drug-resistant CSCs. We then focus on some representative examples that give a glimpse into state-of-the-art nanomedicine approaches developed for CSCs elimination. A perspective on innovative therapeutic strategies and the potential direction of nanomedicine-based CSC therapy in the near future is also presented.
Co-reporter:Song Shen, Jin-Xing Xia, Jun Wang
Biomaterials (January 2016) Volume 74() pp:1-18
Publication Date(Web):January 2016
DOI:10.1016/j.biomaterials.2015.09.037
Co-reporter:Cong-fei Xu, Jun Wang
Asian Journal of Pharmaceutical Sciences (February 2015) Volume 10(Issue 1) pp:1-12
Publication Date(Web):February 2015
DOI:10.1016/j.ajps.2014.08.011
Co-reporter:You-Yong Yuan, Jin-Zhi Du, Wen-Jing Song, Feng Wang, Xian-Zhu Yang, Meng-Hua Xiong and Jun Wang
Journal of Materials Chemistry A 2012 - vol. 22(Issue 18) pp:NaN9329-9329
Publication Date(Web):2012/03/09
DOI:10.1039/C2JM30663H
Biocompatible and biodegradable nanogels with a branched structure designed as potential carriers for drug delivery are reported here. The nanogels were prepared by the reaction of 3,6-dioxaoctan-1,8-diyl bis(ethylene phosphate) (TEGDP) with tris(2-aminoethyl)amine (TREN) in an ionic liquid containing miniemulsion; the particle size was tunable in the nanoscopic range. The biocompatibility of the nanogels was evaluated using both in vitro and in vivo experiments. The nanogels could efficiently load the anticancer drug doxorubicin and showed enzyme-responsive drug release behavior. The nanogels were further labelled with fluorescein isothiocyanate through a reaction with the surface amino groups to demonstrate functionalization. Flow cytometric analyses and confocal laser scanning microscope observations demonstrated that the nanogels could be efficiently taken up by the human breast cancer cell line MDA-MB-231. These biocompatible, biodegradable and surface functionalizable nanogels are expected to have utility in the field of drug delivery.
Co-reporter:Juan Wu, Xi-Qiu Liu, Yu-Cai Wang and Jun Wang
Journal of Materials Chemistry A 2009 - vol. 19(Issue 42) pp:NaN7863-7863
Publication Date(Web):2009/09/16
DOI:10.1039/B908768K
Biodegradable nanogels with tunable sizes, synthesized by a template-free method, as potential carriers for drug delivery are reported. The nanogels are obtained by crosslinking thermo-induced nanoparticles, with subsequent swelling at low temperatures, which were based on a thermosensitive and biocompatible triblock copolymer composed of poly(ethylene glycol) and poly(ethyl ethylene phosphate). The nanogels loaded with doxorubicin are efficiently taken up by A549 tumor cells and the drug could be released intracellularly, demonstrated by flow cytometric analyses and confocal laser scanning microscope observations. It results in enhanced growth inhibition activity to tumor cells in comparison with free doxorubicin treatment. These polyphosphoester and poly(ethylene glycol) based nanogels are biocompatible and biodegradable, rendering potential for drug delivery applications.
Co-reporter:Qinqin Cheng, Hongdong Shi, Hongxia Wang, Yuanzeng Min, Jun Wang and Yangzhong Liu
Chemical Communications 2014 - vol. 50(Issue 56) pp:NaN7430-7430
Publication Date(Web):2014/02/28
DOI:10.1039/C4CC00419A
Asplatin, a fusion of aspirin and cisplatin, exhibits significant cytotoxicity in tumor cells and almost fully overcomes the drug resistance of cisplatin resistant cells. Asplatin is highly accumulated in cancer cells and is activated upon the reduction by ascorbic acid.
Co-reporter:Qinqin Cheng, Hongdong Shi, Hai Huang, Zhiting Cao, Jun Wang and Yangzhong Liu
Chemical Communications 2015 - vol. 51(Issue 99) pp:NaN17539-17539
Publication Date(Web):2015/10/12
DOI:10.1039/C5CC07853A
Self-assembled cholesterol–asplatin-incorporated nanoparticles (SCANs) were prepared for oral delivery of a Pt(IV) prodrug. SCANs exhibit high gastrointestinal stability, sustained drug release and enhanced cell uptake. The oral bioavailability of SCANs was 4.32-fold higher than that of free Pt(IV) prodrugs. The oral administration of SCANs efficaciously inhibits tumor growth with negligible toxicity.
Co-reporter:Juan Wu, Tian-Meng Sun, Xian-Zhu Yang, Jing Zhu, Xiao-Jiao Du, Yan-Dan Yao, Meng-Hua Xiong, Hong-Xia Wang, Yu-Cai Wang and Jun Wang
Biomaterials Science (2013-Present) 2013 - vol. 1(Issue 11) pp:NaN1150-1150
Publication Date(Web):2013/07/17
DOI:10.1039/C3BM60099H
Effective systemic therapy is often necessary to treat hepatocellular carcinoma (HCC). We synthesized a Gal-PPE nanogel consisting of a cross-linked polyphosphate core and galactosylated poly(ethylene glycol) arms for enhanced doxorubicin delivery to diethylnitrosamine-induced HCC in rats. The Gal-PPE nanogel exhibited high affinity to HepG2 cells in vitro, mediated by the asialoglycoprotein receptor. In vivo studies revealed that the Gal-PPE nanogel was taken up more efficiently by hepatocytes, in contrast to m-PPE nanogel. Consequently, doxorubicin delivery with Gal-PPE significantly inhibited the progress of HCC, reducing neoplastic liver nodules and prolonging the survival time of HCC rats more significantly. These results demonstrate the potential of Gal-PPE as a nanocarrier for improved HCC chemotherapy.
Co-reporter:Jin-Zhi Du, Hong-Yan Long, You-Yong Yuan, Meng-Meng Song, Liang Chen, Hong Bi and Jun Wang
Chemical Communications 2012 - vol. 48(Issue 9) pp:NaN1259-1259
Publication Date(Web):2011/12/16
DOI:10.1039/C2CC16363B
Micelle-to-vesicle morphological transition has been achieved by light-induced rapid hydrophilic arm detachment from a star polymer. This provides a remote and clean method to control morphology transition of polymeric assemblies.
Co-reporter:Yu-Cai Wang, Juan Wu, Yang Li, Jin-Zhi Du, You-Yong Yuan and Jun Wang
Chemical Communications 2010 - vol. 46(Issue 20) pp:NaN3522-3522
Publication Date(Web):2010/04/08
DOI:10.1039/C002620D
We report the preparation of biodegradable nanoscopic hydrogels and their application for targeted drug delivery. The nanogel is synthesized in a template-free method by photo-crosslinking salt-induced polymer assemblies. With convenient incorporation of targeted lactosyl moieties, the nanogels efficiently deliver doxorubicin to HepG2 cells through receptor-mediated internalization.
Co-reporter:Rong Sun, Xiao-Jiao Du, Chun-Yang Sun, Song Shen, Yang Liu, Xian-Zhu Yang, Yan Bao, Yan-Hua Zhu and Jun Wang
Biomaterials Science (2013-Present) 2015 - vol. 3(Issue 7) pp:NaN1113-1113
Publication Date(Web):2015/03/04
DOI:10.1039/C4BM00430B
Polymeric nanoparticles have been widely used as nano-drug delivery systems in preclinical and clinical trials for cancer therapy, and these systems usually need to be sterically stabilized by poly(ethylene glycol) (PEG) to maintain stability and avoid rapid clearance by the immune system. Recently, zwitterionic materials have been demonstrated to be potential alternatives to the classic PEG. Herein, we developed two drug delivery systems stabilized by zwitterionic polyphosphoesters. These nanoparticles showed favourable stability and anti-protein absorption ability in vitro. Meanwhile, as drug carriers, these zwitterionic polyphosphoester-stabilized nanoparticles significantly prolonged drug circulation half-lives and increased drug accumulation in tumors, which was comparable to PEG-stabilized nanoparticles. Systemic delivery of doxorubicin (DOX) by zwitterionic polyphosphoester-stabilized nanoparticles significantly inhibited tumor growth in a MDA-MB-231 tumor model, suggesting the potential of zwitterionic polyphosphoester-based nanoparticles in anticancer drug delivery.
Co-reporter:You-Yong Yuan, Jin-Zhi Du and Jun Wang
Chemical Communications 2012 - vol. 48(Issue 4) pp:NaN572-572
Publication Date(Web):2011/11/07
DOI:10.1039/C1CC16065F
A simple and universal route to functional cyclic polyesters has been demonstrated, combining two consecutive click reactions of azide–alkyne cycloaddition of linear hetero-bifunctional precursors and thiol–ene coupling for post cyclization functionalizations. Functional cationic and thermo-responsive cyclic polyphosphoesters have been synthesized to demonstrate the efficiency of the procedures.
4-(3-(Pyridin-2-yl)-1H-pyrazol-4-yl)quinoline
N-[1-(2,3-DIOLEYLOXY)PROPYL]-N,N,N-TRIMETHYLAMMONIUM CHLORIDE
1-Propanaminium,N,N,N-trimethyl-2,3-bis[[(9Z)-1-oxo-9-octadecen-1-yl]oxy]-
Adriamycin