Network


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

Hotspot


Dive into the research topics where Jinghan Su is active.

Publication


Featured researches published by Jinghan Su.


Advanced Materials | 2016

Cancer-Cell-Biomimetic Nanoparticles for Targeted Therapy of Homotypic Tumors

Huiping Sun; Jinghan Su; Qingshuo Meng; Qi Yin; Lingli Chen; Wangwen Gu; Pengcheng Zhang; Zhiwen Zhang; Haijun Yu; Siling Wang; Yaping Li

A unique biomimetic drug-delivery system composed of 4T1-breast-cancer-cell membranes and paclitaxel-loaded polymeric nanoparticles (PPNs) (cell-membrane-coated PPNs), demonstrates superior interactions to its source tumor cells and elongated blood circulation, and displays highly cell-specific targeting of the homotypic primary tumor and metastases, with successful inhibition of the growth and lung metastasis of the breast cancer cells.


Biomaterials | 2015

Inhibition of metastasis and growth of breast cancer by pH-sensitive poly (β-amino ester) nanoparticles co-delivering two siRNA and paclitaxel.

Shan Tang; Qi Yin; Jinghan Su; Huiping Sun; Qingshuo Meng; Yi Chen; Lingli Chen; Yongzhuo Huang; Wangwen Gu; Minghua Xu; Haijun Yu; Zhiwen Zhang; Yaping Li

Breast cancer is the most vicious killer for womens health, while metastasis is the main culprit, which leads to failure of treatment by increasing relapse rate. In this work, a new complexes nanoparticles loading two siRNA (Snail siRNA (siSna) and Twist siRNA (siTwi)) and paclitaxel (PTX) were designed and constructed using two new amphiphilic polymer, polyethyleneimine-block-poly[(1,4-butanediol)-diacrylate-β-5-hydroxyamylamine] (PEI-PDHA) and polyethylene glycol-block-poly[(1,4-butanediol)-diacrylate-β-5-hydroxyamylamine] (PEG-PDHA) by self-assembly. The experimental results showed that in the 4T1 tumor-bearing mice models, PEI-PDHA/PEG-PDHA/PTX/siSna/siTwi) complex nanoparticles (PPSTs) raised the accumulation and retention of both PTX and siRNA in tumor after administrated intravenously, resulted in the strong inhibition of the tumor growth and metastasis simultaneously. It was found that co-delivery of siSna and siTwi had more significant anti-metastasis effect than delivering a single siRNA, as a result of simultaneously inhibiting the motility of cancer cells and degradation of ECM. Therefore, PPSTs could be a promising co-delivery vector for effective therapy of metastatic breast cancer.


Theranostics | 2017

Enhanced Blood Suspensibility and Laser-Activated Tumor-specific Drug Release of Theranostic Mesoporous Silica Nanoparticles by Functionalizing with Erythrocyte Membranes

Jinghan Su; Huiping Sun; Qingshuo Meng; Pengcheng Zhang; Qi Yin; Yaping Li

Mesoporous silica nanoparticles (MSNs), with their large surface area and tunable pore sizes, have been widely applied for anticancer therapeutic cargos delivery with a high loading capacity. However, easy aggregation in saline buffers and limited blood circulation lifetime hinder their delivery efficiency and the anticancer efficacy. Here, new multifunctional MSNs-supported red-blood-cell (RBC)-mimetic theranostic nanoparticles with long blood circulation, deep-red light-activated tumor imaging and drug release were reported. High loading capacities were achieved by camouflaging MSNs with RBC membrane to co-load an anticancer drug doxorubicin (Dox) (39.1 wt%) and a near-infrared photosensitizer chlorin e6 (Ce6) (21.1 wt%). The RBC membrane-coating protected drugs from leakage, and greatly improved the colloidal stability of MSNs, with negligible particle size change over two weeks. Upon an external laser stimuli, the RBC membrane could be destroyed, resulting in 10 times enhancement of Dox release. In a 4T1 breast cancer mouse model, the RBC-mimetic MSNs could realize in vivo tumor imaging with elongated tumor accumulation lifetime for over 24 h, and laser-activated tumor-specific Dox accumulation. The RBC-mimetic MSNs could integrate the Ce6-based photodynamic therapy and Dox-based chemotherapy, completely suppress the primary tumor growth and inhibit metastasis of breast cancer, which could provide a new strategy for optimization of MSNs and efficient anticancer drug delivery.


Biomaterials | 2017

Dual pH-sensitive micelles with charge-switch for controlling cellular uptake and drug release to treat metastatic breast cancer

Shan Tang; Qingshuo Meng; Huiping Sun; Jinghan Su; Qi Yin; Zhiwen Zhang; Haijun Yu; Lingli Chen; Wangwen Gu; Yaping Li

For successful chemotherapy against metastatic breast cancer, the great efforts are still required for designing drug delivery systems that can be selectively internalized by tumor cells and release the cargo in a controlled manner. In this work, the chemotherapeutic agent paclitaxel (PTX) was loaded with the dual-pH sensitive micelle (DPM), which consisted of a pH-sensitive core, an acid-cleavable anionic shell, and a polyethylene glycol (PEG) corona. In the slightly acidic environment of tumor tissues, the anionic shell was taken off, inducing the conversion of the surface charge of DPM from negative to positive, which resulted in more efficient cellular uptake, stronger cytotoxicity and higher intra-tumor accumulation of PTX in the murine breast cancer 4T1 tumor-bearing mice models compared to the micelles with irremovable anionic or non-ionic shell. Meanwhile, the pH-sensitive core endowed DPM with rapid drug release in endo/lysosomes. The inhibitory rates of DPM against tumor growth and lung metastasis achieved 77.7% and 88.3%, respectively, without significant toxicity. Therefore, DPM is a promising nanocarrier for effective therapy of metastatic breast cancer due to satisfying the requirements of both selective uptake by tumor cells and sufficient and fast intracellular drug release.


Theranostics | 2017

Preparation and Application of Cell Membrane-Camouflaged Nanoparticles for Cancer Therapy

Yihui Zhai; Jinghan Su; Wei Ran; Pengcheng Zhang; Qi Yin; Zhiwen Zhang; Haijun Yu; Yaping Li

Cancer is one of the leading causes of death worldwide. Many treatments have been developed so far, although effective, suffer from severe side effects due to low selectivity. Nanoparticles can improve the therapeutic index of their delivered drugs by specifically transporting them to tumors. However, their exogenous nature usually leads to fast clearance by mononuclear phagocytic system. Recently, cell membrane-camouflaged nanoparticles have been investigated for cancer therapy, taking advantages of excellent biocompatibility and versatile functionality of cell membranes. In this review, we summarized source materials and procedures that have been used for constructing and characterizing biomimetic nanoparticles with a focus on their application in cancer therapy.


Molecular Pharmaceutics | 2015

Inhibition of Breast Cancer Metastasis by Pluronic Copolymers with Moderate Hydrophilic–Lipophilic Balance

Huiping Sun; Qingshuo Meng; Shan Tang; Jinghan Su; Qi Yin; Lingli Chen; Wangwen Gu; Haijun Yu; Zhiwen Zhang; Siling Wang; Yaping Li

Metastasis is the primary cause resulting in the high mortality of breast cancer. The inherent antimetastasis bioactivity of Pluronic copolymers with a wide range of hydrophilic-lipophilic balance (HLB) including Pluronic L61, P85, P123, F127, F68, and F108 was first explored on metastatic 4T1 breast cancer cells. The results indicated that P85 and P123 could strongly inhibit the migration and invasion of 4T1 cells. The effects of the polymers on cell healing, migration, and invasion exhibited bell-shaped dependencies on HLB of Pluronic copolymers, and the better antimetastasis effects of Pluronic copolymers could be achieved with the HLB between 8 and 16. P85 and P123 themselves could significantly inhibit pulmonary metastasis in 4T1 mammary tumor metastasis model in situ. In addition, a synergetic antimetastasis effect could be achieved during drug combination of doxorubicin hydrochloride (DOX) and P85 or P123 intravenously. The metastasis effects of P85 and P123 both in vitro and in vivo were partially attributed to the downregulation of matrix metalloproteinase-9 (MMP-9). Therefore, Pluronic copolymers with moderate HLB 8-16 such as P85 and P123 could be promising excipients with therapeutics in drug delivery systems to inhibit breast cancer metastasis.


Acta Pharmacologica Sinica | 2016

Silibinin and indocyanine green-loaded nanoparticles inhibit the growth and metastasis of mammalian breast cancer cells in vitro.

Huiping Sun; Jinghan Su; Qingshuo Meng; Qi Yin; Zhiwen Zhang; Haijun Yu; Pengcheng Zhang; Siling Wang; Yaping Li

Aim:To improve the therapeutic efficacy of cancer treatments, combinational therapies based on nanosized drug delivery system (NDDS) has been developed recently. In this study we designed a new NDDS loaded with an anti-metastatic drug silibinin and a photothermal agent indocyanine green (ICG), and investigated its effects on the growth and metastasis of breast cancer cells in vitro.Methods:Silibinin and ICG were self-assembled into PCL lipid nanoparticles (SIPNs). Their physical characteristics including the particle size, zeta potential, morphology and in vitro drug release were examined. 4T1 mammalian breast cancer cells were used to evaluate their cellular internalization, cytotoxicity, and their influences on wound healing, in vitro cell migration and invasion.Results:SIPNs showed a well-defined spherical shape with averaged size of 126.3±0.4 nm and zeta potential of −10.3±0.2 mV. NIR laser irradiation substantially increased the in vitro release of silibinin from the SIPNs (58.3% at the first 8 h, and 97.8% for the total release). Furthermore, NIR laser irradiation markedly increased the uptake of SIPNs into 4T1 cells. Under the NIR laser irradiation, both SIPNs and IPNs (PCL lipid nanoparticles loaded with ICG alone) caused dose-dependent ablation of 4T1 cells. The wound healing, migration and invasion experiments showed that SIPNs exposed to NIR laser irradiation exhibited dramatic in vitro anti-metastasis effects.Conclusion:SIPNs show temperature-sensitive drug release following NIR laser irradiation, which can inhibit the growth and metastasis of breast cancer cells in vitro.


Advanced Materials | 2018

Traceable Bioinspired Nanoparticle for the Treatment of Metastatic Breast Cancer via NIR-Trigged Intracellular Delivery of Methylene Blue and Cisplatin

Yihui Zhai; Wei Ran; Jinghan Su; Tianqun Lang; Jia Meng; Guanru Wang; Pengcheng Zhang; Yaping Li

Cytotoxic T lymphocyte (CTL) eliminates abnormal cells through target recognition-triggered intracellular toxin delivery. Chimeric antigen receptor T-cell improves cancer cell recognition of CTL, but its effectiveness and safety in solid tumor treatment are still hampered by poor tumor infiltration, suppressive tumor microenvironment, and severe on-target off-tumor toxicity. Given the functionality and challenges of CTL in cancer therapy, herein, a CTL-inspired nanovesicle (MPV) with a cell membrane-derived shell and a methylene blue (MB) and cisplatin (Pt) loaded gelatin nanogel core is created. The MPV generates contrast for tumor photoacoustic imaging, and produces hyperthermia upon laser irradiation, enabling photothermal imaging and deep tumor penetration. Meanwhile, it releases MB and Pt, and then delivers them into the cytosol of cancer cells, which process can be visualized by imaging the recovery of MB-derived fluorescence. The localized hyperthermia, photodynamic therapy, and chemotherapy together kill 4T1 breast cancer cells effectively, resulting in primary tumor regression and 97% inhibition of pulmonary metastasis, without significant toxicity to the animals. Taken together, the MPV shows tumor-specific and stimuli-triggered intracellular toxin delivery with advantages in traceable accumulation and activation, high tumor penetration, and triple combination therapy, and thus can be an effective nanomedicine for combating metastatic breast cancer.


Advanced Functional Materials | 2016

Long Circulation Red-Blood-Cell-Mimetic Nanoparticles with Peptide-Enhanced Tumor Penetration for Simultaneously Inhibiting Growth and Lung Metastasis of Breast Cancer

Jinghan Su; Huiping Sun; Qingshuo Meng; Qi Yin; Shan Tang; Pengcheng Zhang; Yi Chen; Zhiwen Zhang; Haijun Yu; Yaping Li


Advanced Functional Materials | 2017

Cancer Cell Membrane-Coated Gold Nanocages with Hyperthermia-Triggered Drug Release and Homotypic Target Inhibit Growth and Metastasis of Breast Cancer

Huiping Sun; Jinghan Su; Qingshuo Meng; Qi Yin; Lingli Chen; Wangwen Gu; Zhiwen Zhang; Haijun Yu; Pengcheng Zhang; Siling Wang; Yaping Li

Collaboration


Dive into the Jinghan Su's collaboration.

Top Co-Authors

Avatar

Yaping Li

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Qi Yin

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Qingshuo Meng

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Haijun Yu

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Huiping Sun

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Zhiwen Zhang

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Pengcheng Zhang

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Lingli Chen

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Wangwen Gu

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Shan Tang

Chinese Academy of Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge