Network


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

Hotspot


Dive into the research topics where Chaofeng Mu is active.

Publication


Featured researches published by Chaofeng Mu.


Theranostics | 2014

Cyclodextrin and polyethylenimine functionalized mesoporous silica nanoparticles for delivery of siRNA cancer therapeutics

Jianliang Shen; Han Cheon Kim; Hua Su; Feng Wang; Joy Wolfram; Dickson K. Kirui; Junhua Mai; Chaofeng Mu; Liang Nian Ji; Zong-Wan Mao; Haifa Shen

Effective delivery holds the key to successful in vivo application of therapeutic small interfering RNA (siRNA). In this work, we have developed a universal siRNA carrier consisting of a mesoporous silica nanoparticle (MSNP) functionalized with cyclodextrin-grafted polyethylenimine (CP). CP provides positive charge for loading of siRNA through electrostatic interaction and enables effective endosomal escape of siRNA. Using intravital microscopy we were able to monitor tumor enrichment of CP-MSNP/siRNA particles in live mice bearing orthotopic MDA-MB-231 xenograft tumors. CP-MSNP delivery of siRNA targeting the M2 isoform of the glycolytic enzyme pyruvate kinase (PKM2) resulted in effective knockdown of gene expression in vitro and in vivo. Suppression of PKM2 led to inhibition of tumor cell growth, invasion, and migration.


ACS Nano | 2013

High Capacity Nanoporous Silicon Carrier for Systemic Delivery of Gene Silencing Therapeutics

Jianliang Shen; Rong Xu; Junhua Mai; Han Cheon Kim; Xiaojing Guo; Guoting Qin; Yong Yang; Joy Wolfram; Chaofeng Mu; Xiaojun Xia; Jianhua Gu; Xuewu Liu; Zong-Wan Mao; Mauro Ferrari; Haifa Shen

Gene silencing agents such as small interfering RNA (siRNA) and microRNA offer the promise to modulate expression of almost every gene for the treatment of human diseases including cancer. However, lack of vehicles for effective systemic delivery to the disease organs has greatly limited their in vivo applications. In this study, we developed a high capacity polycation-functionalized nanoporous silicon (PCPS) platform comprised of nanoporous silicon microparticles functionalized with arginine-polyethyleneimine inside the nanopores for effective delivery of gene silencing agents. Incubation of MDA-MB-231 human breast cancer cells with PCPS loaded with STAT3 siRNA (PCPS/STAT3) or GRP78 siRNA (PCPS/GRP78) resulted in 91 and 83% reduction of STAT3 and GRP78 gene expression in vitro. Treatment of cells with a microRNA-18a mimic in PCPS (PCPS/miR-18) knocked down 90% expression of the microRNA-18a target gene ATM. Systemic delivery of PCPS/STAT3 siRNA in murine model of MDA-MB-231 breast cancer enriched particles in tumor tissues and reduced STAT3 expression in cancer cells, causing significant reduction of cancer stem cells in the residual tumor tissue. At the therapeutic dosage, PCPS/STAT3 siRNA did not trigger acute immune response in FVB mice, including changes in serum cytokines, chemokines, and colony-stimulating factors. In addition, weekly dosing of PCPS/STAT3 siRNA for four weeks did not cause signs of subacute toxicity based on changes in body weight, hematology, blood chemistry, and major organ histology. Collectively, the results suggest that we have developed a safe vehicle for effective delivery of gene silencing agents.


Advanced Healthcare Materials | 2014

Multifunctional gold nanorods for siRNA gene silencing and photothermal therapy

Jianliang Shen; Han Cheon Kim; Chaofeng Mu; Emanuela Gentile; Junhua Mai; Joy Wolfram; Liang Nian Ji; Mauro Ferrari; Zong-Wan Mao; Haifa Shen

Cancer is a complex disease that usually requires several treatment modalities. A multifunctional nanotherapeutic system is designed, incorporating small interfering RNA (siRNA) and gold nanorods (Au NRs) for photothermal therapy. Surface-engineered Au NRs with polyethylenimine are synthesized using a layer-by-layer assembly and siRNA is absorbed on the surface. The siRNA is efficiently delivered into breast cancer cells, resulting in subsequent gene silencing. Cells are then irradiated with near-infrared (NIR) light, causing heat-induced anticancer activity. The combination of gene silencing and photothermal therapy results in effective inhibition of cell proliferation.


Cell Reports | 2015

Porous Silicon Microparticle Potentiates Anti-Tumor Immunity by Enhancing Cross-Presentation and Inducing Type I Interferon Response

Xiaojun Xia; Junhua Mai; Rong Xu; Jorge Enrique Tovar Perez; Maria L. Guevara; Qi Shen; Chaofeng Mu; Hui Ying Tung; David B. Corry; Scott E. Evans; Xuewu Liu; Mauro Ferrari; Zhiqiang Zhang; Xian Chang Li; Rong Fu Wang; Haifa Shen

Micro- and nanometer-size particles have become popular candidates for cancer vaccine adjuvants. However, the mechanism by which such particles enhance immune responses remains unclear. Here, we report a porous silicon microparticle (PSM)-based cancer vaccine that greatly enhances cross-presentation and activates type I interferon (IFN-I) response in dendritic cells (DCs). PSM-loaded antigen exhibited prolonged early endosome localization and enhanced cross-presentation through both proteasome- and lysosome-dependent pathways. Phagocytosis of PSM by DCs induced IFN-I responses through a TRIF- and MAVS-dependent pathway. DCs primed with PSM-loaded HER2 antigen produced robust CD8 T cell-dependent anti-tumor immunity in mice bearing HER2+ mammary gland tumors. Importantly, this vaccination activated the tumor immune microenvironment with elevated levels of intra-tumor IFN-I and MHCII expression, abundant CD11c+ DC infiltration, and tumor-specific cytotoxic T cell responses. These findings highlight the potential of PSM as an immune adjuvant to potentiate DC-based cancer immunotherapy.


Journal of Controlled Release | 2014

Bone marrow endothelium-targeted therapeutics for metastatic breast cancer

Junhua Mai; Yi Huang; Chaofeng Mu; Guodong Zhang; Rong Xu; Xiaojing Guo; Xiaojun Xia; David E. Volk; Ganesh L.R. Lokesh; Varatharasa Thiviyanathan; David G. Gorenstein; Xuewu Liu; Mauro Ferrari; Haifa Shen

Effective treatment of cancer metastasis to the bone relies on bone marrow drug accumulation. The surface proteins in the bone marrow vascular endothelium provide docking sites for targeted drug delivery. We have developed a thioaptamer that specifically binds to E-selectin that is overexpressed in the vasculature of tumor and inflammatory tissues. In this study, we tested targeted delivery of therapeutic siRNA loaded in the E-selectin thioaptamer-conjugated multistage vector (ESTA-MSV) drug carrier to bone marrow for the treatment of breast cancer bone metastasis. We evaluated tumor type- and tumor growth stage-dependent targeting in mice bearing metastatic breast cancer in the bone, and carried out studies to identify factors that determine targeting efficiency. In a subsequent study, we delivered siRNA to knock down expression of the human STAT3 gene in murine xenograft models of human MDA-MB-231 breast tumor, and assessed therapeutic efficacy. Our studies revealed that the CD31(+)E-selectin(+) population accounted for 20.8%, 26.4% and 29.9% of total endothelial cells respectively inside the femur of mice bearing early, middle and late stage metastatic MDA-MB-231 tumors. In comparison, the double positive cells remained at a basal level in mice with early stage MCF-7 tumors, and jumped to 23.9% and 28.2% when tumor growth progressed to middle and late stages. Accumulation of ESTA-MSV inside the bone marrow correlated with the E-selectin expression pattern. There was up to 5-fold enrichment of the targeted MSV in the bone marrow of mice bearing early or late stage MDA-MB-231 tumors and of mice with late stage, but not early stage, MCF-7 tumors. Targeted delivery of STAT3 siRNA in ESTA-MSV resulted in knockdown of STAT3 expression in 48.7% of cancer cells inside the bone marrow. Weekly systemic administration of ESTA-MSV/STAT3 siRNA significantly extended survival of mice with MDA-MB-231 bone metastasis. In conclusion, targeting the overexpressed E-selectin provides an effective approach for tissue-specific drug delivery to the bone marrow. Tumor growth in the bone can be effectively inhibited by blockage of the STAT3 signaling.


Advanced Healthcare Materials | 2016

A Micro/Nano Composite for Combination Treatment of Melanoma Lung Metastasis

Yu Mi; Chaofeng Mu; Joy Wolfram; Zaian Deng; Tony Y. Hu; Xuewu Liu; Elvin Blanco; Haifa Shen; Mauro Ferrari

The successful treatment of malignant disease generally requires the use of multiple therapeutic agents that are coordinated in a spatiotemporal manner to enable synergy. Here, a porous silicon-based micro/nano composite (MNC) that is capable of simultaneously delivering chemotherapeutic agents and small interfering RNA (siRNA) to the lungs following intravenous injection is designed. The pores of the silicon microparticles are loaded with B-Raf proto-oncogene serine/threonine kinase (BRAF) siRNA-containing liposomes, while the surface is conjugated with docetaxel-encapsulated polymeric nanoparticles. The synergistic antitumor effect of the MNC is demonstrated in vitro in melanoma cells and in vivo using a mouse model for melanoma lung metastasis. The MNC displays superior therapeutic efficacy and increased accumulation in metastatic melanoma lesions in the lungs in comparison to combination therapy with liposomes and polymers. The results indicate that the MNC can be used as an effective delivery vehicle for simultaneous enrichment of multiple therapeutic agents in the lungs.


Leukemia | 2016

In vivo targeting of leukemia stem cells by directing parthenolide-loaded nanoparticles to the bone marrow niche

Hongliang Zong; Siddhartha Sen; Guodong Zhang; Chaofeng Mu; Zaineb Fadhel Albayati; David G. Gorenstein; Xuewu Liu; Mauro Ferrari; Peter A. Crooks; Gail J. Roboz; Haifa Shen; Monica L. Guzman

In vivo targeting of leukemia stem cells by directing parthenolide-loaded nanoparticles to the bone marrow niche


Pharmacological Research | 2016

Enzyme-responsive multistage vector for drug delivery to tumor tissue.

Yu Mi; Joy Wolfram; Chaofeng Mu; Xuewu Liu; Elvin Blanco; Haifa Shen; Mauro Ferrari

Various nanodelivery systems have been designed to release therapeutic agents upon contact with specific enzymes. However, enzyme-triggered release typically takes place in the tissue interstitium, thereby resulting in the extracellular delivery of drugs. Here, we have designed an enzyme-stimulated multistage vector (ESMSV), which enables stimulus-triggered release of drug-encapsulated nanoparticles from a microparticle. Specifically, polymeric nanoparticles with a surface matrix metalloproteinase-2 (MMP2) peptide substrate were conjugated to the surface of porous silicon microparticles. In the presence of MMP2, the polymeric nanoparticles were released into the tumor interstitium. This platform can be used to attain triggered drug release, while simultaneously facilitating the cellular internalization of drugs. The results indicate that nanoparticle release was MMP2-specific and resulted in improved intracellular uptake of hydrophobic agents in the presence of MMP2. Furthermore, in a mouse model of melanoma lung metastasis, systemic delivery of ESMSVs caused a substantial increase in intracellular accumulation of agents in cancer cells in comparison to delivery with non-stimulus-responsive particles.


Mini-reviews in Medicinal Chemistry | 2016

Recent advances in discovering the role of CCL5 in metastatic breast cancer

Ayesha Khalid; Joy Wolfram; Ilaria Ferrari; Chaofeng Mu; Junhua Mai; Zhizhou Yang; Yuliang Zhao; Mauro Ferrari; Xiaojing Ma; Haifa Shen

A variety of therapeutic strategies are currently under investigation to inhibit factors that promote tumor invasion, as metastasis is the most common cause of mortality for cancer patients. Notably, considerable emphasis has been placed on studying metastasis as a dynamic process that is highly dependent on the tumor microenvironment. In regards to breast cancer, chemokine C-C motif ligand 5 (CCL5), which is produced by tumor-associated stromal cells, has been established as an important contributor to metastatic disease. This review summarizes recent discoveries uncovering the role of this chemokine in breast cancer metastasis, including conditions that increase the generation of CCL5 and effects induced by this signaling pathway. In particular, CCL-5-mediated cancer cell migration and invasion are discussed in the context of intertwined feedback loops between breast cancer cells and stromal cells. Moreover, the potential use of CCL5 and its receptor chemokine C-C motif receptor 5 (CCR5) as targets for preventing breast cancer metastasis is also reviewed.


Nano Letters | 2017

A Liposome Encapsulated Ruthenium Polypyridine Complex as a Theranostic Platform for Triple-Negative Breast Cancer

Jianliang Shen; Han Cheon Kim; Joy Wolfram; Chaofeng Mu; Wei Zhang; Haoran Liu; Yan Xie; Junhua Mai; Hang Zhang; Zhi Li; Maria L. Guevara; Zong-Wan Mao; Haifa Shen

Ruthenium coordination complexes have the potential to serve as novel theranostic agents for cancer. However, a major limitation in their clinical implementation is effective tumor accumulation. In this study, we have developed a liposome-based theranostic nanodelivery system for [Ru(phen)2dppz](ClO4)2 (Lipo-Ru). This ruthenium polypyridine complex emits a strong fluorescent signal when incorporated in the hydrophobic lipid bilayer of the delivery vehicle or in the DNA helix, enabling visualization of the therapeutic agent in tumor tissues. Incubation of MDA-MB-231 breast cancer cells with Lipo-Ru induced double-strand DNA breaks and triggers apoptosis. In a mouse model of triple-negative breast cancer, treatment with Lipo-Ru dramatically reduced tumor growth. Biodistribution studies of Lipo-Ru revealed that more than 20% of the injected dose accumulated in the tumor. These results suggest that Lipo-Ru could serve as a promising theranostic platform for cancer.

Collaboration


Dive into the Chaofeng Mu's collaboration.

Top Co-Authors

Avatar

Haifa Shen

Houston Methodist Hospital

View shared research outputs
Top Co-Authors

Avatar

Mauro Ferrari

Houston Methodist Hospital

View shared research outputs
Top Co-Authors

Avatar

Joy Wolfram

Houston Methodist Hospital

View shared research outputs
Top Co-Authors

Avatar

Junhua Mai

Houston Methodist Hospital

View shared research outputs
Top Co-Authors

Avatar

Jianliang Shen

Houston Methodist Hospital

View shared research outputs
Top Co-Authors

Avatar

Xuewu Liu

Houston Methodist Hospital

View shared research outputs
Top Co-Authors

Avatar

Han Cheon Kim

Houston Methodist Hospital

View shared research outputs
Top Co-Authors

Avatar

Xiaojun Xia

Houston Methodist Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Guodong Zhang

Houston Methodist Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge