Network


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

Hotspot


Dive into the research topics where Haifa Shen is active.

Publication


Featured researches published by Haifa Shen.


Nature Biotechnology | 2015

Principles of nanoparticle design for overcoming biological barriers to drug delivery

Elvin Blanco; Haifa Shen; Mauro Ferrari

Biological barriers to drug transport prevent successful accumulation of nanotherapeutics specifically at diseased sites, limiting efficacious responses in disease processes ranging from cancer to inflammation. Although substantial research efforts have aimed to incorporate multiple functionalities and moieties within the overall nanoparticle design, many of these strategies fail to adequately address these barriers. Obstacles, such as nonspecific distribution and inadequate accumulation of therapeutics, remain formidable challenges to drug developers. A reimagining of conventional nanoparticles is needed to successfully negotiate these impediments to drug delivery. Site-specific delivery of therapeutics will remain a distant reality unless nanocarrier design takes into account the majority, if not all, of the biological barriers that a particle encounters upon intravenous administration. By successively addressing each of these barriers, innovative design features can be rationally incorporated that will create a new generation of nanotherapeutics, realizing a paradigmatic shift in nanoparticle-based drug delivery.


Nature | 2014

XBP1 promotes triple-negative breast cancer by controlling the HIF1α pathway

Xi Chen; Dimitrios Iliopoulos; Qing Zhang; Qianzi Tang; Matthew B. Greenblatt; Maria Hatziapostolou; Elgene Lim; Wai Leong Tam; Min Ni; Yiwen Chen; Junhua Mai; Haifa Shen; Dorothy Hu; Stanley Adoro; Bella Hu; Minkyung Song; Chen Tan; Melissa D. Landis; Mauro Ferrari; Sandra J. Shin; Myles Brown; Jenny Chang; X. Shirley Liu; Laurie H. Glimcher

Cancer cells induce a set of adaptive response pathways to survive in the face of stressors due to inadequate vascularization. One such adaptive pathway is the unfolded protein (UPR) or endoplasmic reticulum (ER) stress response mediated in part by the ER-localized transmembrane sensor IRE1 (ref. 2) and its substrate XBP1 (ref. 3). Previous studies report UPR activation in various human tumours, but the role of XBP1 in cancer progression in mammary epithelial cells is largely unknown. Triple-negative breast cancer (TNBC)—a form of breast cancer in which tumour cells do not express the genes for oestrogen receptor, progesterone receptor and HER2 (also called ERBB2 or NEU)—is a highly aggressive malignancy with limited treatment options. Here we report that XBP1 is activated in TNBC and has a pivotal role in the tumorigenicity and progression of this human breast cancer subtype. In breast cancer cell line models, depletion of XBP1 inhibited tumour growth and tumour relapse and reduced the CD44highCD24low population. Hypoxia-inducing factor 1α (HIF1α) is known to be hyperactivated in TNBCs. Genome-wide mapping of the XBP1 transcriptional regulatory network revealed that XBP1 drives TNBC tumorigenicity by assembling a transcriptional complex with HIF1α that regulates the expression of HIF1α targets via the recruitment of RNA polymerase II. Analysis of independent cohorts of patients with TNBC revealed a specific XBP1 gene expression signature that was highly correlated with HIF1α and hypoxia-driven signatures and that strongly associated with poor prognosis. Our findings reveal a key function for the XBP1 branch of the UPR in TNBC and indicate that targeting this pathway may offer alternative treatment strategies for this aggressive subtype of breast cancer.


Cancer Gene Therapy | 2012

Nanovector delivery of siRNA for cancer therapy

Haifa Shen; Tong Sun; Mauro Ferrari

RNA interference holds the promise to knock down expression of every cancer gene. Both academic laboratories and pharmaceutical companies have committed heavily on manpower and financial resources to develop small interfering RNA (siRNA) cancer therapeutics over the last decade. Although significant advances have been made in the design of siRNA therapeutics and mechanism of action on cancer cell killing, there are still many hurdles to overcome including effective delivery of therapeutics in vivo. Nanotechnology has had an important role in the development of delivery vectors so far. This article summarizes current nanovectors for siRNA delivery, discusses technical challenges in overcoming biological barriers, and introduces the multistage vector system for tumor-specific delivery.


Nature Biotechnology | 2016

An injectable nanoparticle generator enhances delivery of cancer therapeutics

Rong Xu; Guodong Zhang; Junhua Mai; Xiaoyong Deng; Victor Segura-Ibarra; Suhong Wu; Jianliang Shen; Haoran Liu; Zhenhua Hu; Lingxiao Chen; Yi Huang; Eugene Koay; Yu Huang; Jun Liu; Joe E. Ensor; Elvin Blanco; Xuewu Liu; Mauro Ferrari; Haifa Shen

The efficacy of cancer drugs is often limited because only a small fraction of the administered dose accumulates in tumors. Here we report an injectable nanoparticle generator (iNPG) that overcomes multiple biological barriers to cancer drug delivery. The iNPG is a discoidal micrometer-sized particle that can be loaded with chemotherapeutics. We conjugate doxorubicin to poly(L-glutamic acid) by means of a pH-sensitive cleavable linker, and load the polymeric drug (pDox) into iNPG to assemble iNPG-pDox. Once released from iNPG, pDox spontaneously forms nanometer-sized particles in aqueous solution. Intravenously injected iNPG-pDox accumulates at tumors due to natural tropism and enhanced vascular dynamics and releases pDox nanoparticles that are internalized by tumor cells. Intracellularly, pDox nanoparticles are transported to the perinuclear region and cleaved into Dox, thereby avoiding excretion by drug efflux pumps. Compared to its individual components or current therapeutic formulations, iNPG-pDox shows enhanced efficacy in MDA-MB-231 and 4T1 mouse models of metastatic breast cancer, including functional cures in 40–50% of treated mice.


Journal of Clinical Investigation | 2014

Transport properties of pancreatic cancer describe gemcitabine delivery and response

Eugene J. Koay; Mark J. Truty; Vittorio Cristini; Ryan M. Thomas; Rong Chen; Deyali Chatterjee; Ya’an Kang; Priya Bhosale; Eric P. Tamm; Christopher H. Crane; Milind Javle; Matthew H. Katz; Vijaya Gottumukkala; Marc A. Rozner; Haifa Shen; J. E. Lee; Huamin Wang; Yuling Chen; William Plunkett; James L. Abbruzzese; Robert A. Wolff; Gauri R. Varadhachary; Mauro Ferrari; Jason B. Fleming

BACKGROUND The therapeutic resistance of pancreatic ductal adenocarcinoma (PDAC) is partly ascribed to ineffective delivery of chemotherapy to cancer cells. We hypothesized that physical properties at vascular, extracellular, and cellular scales influence delivery of and response to gemcitabine-based therapy. METHODS We developed a method to measure mass transport properties during routine contrast-enhanced CT scans of individual human PDAC tumors. Additionally, we evaluated gemcitabine infusion during PDAC resection in 12 patients, measuring gemcitabine incorporation into tumor DNA and correlating its uptake with human equilibrative nucleoside transporter (hENT1) levels, stromal reaction, and CT-derived mass transport properties. We also studied associations between CT-derived transport properties and clinical outcomes in patients who received preoperative gemcitabine-based chemoradiotherapy for resectable PDAC. RESULTS Transport modeling of 176 CT scans illustrated striking differences in transport properties between normal pancreas and tumor, with a wide array of enhancement profiles. Reflecting the interpatient differences in contrast enhancement, resected tumors exhibited dramatic differences in gemcitabine DNA incorporation, despite similar intravascular pharmacokinetics. Gemcitabine incorporation into tumor DNA was inversely related to CT-derived transport parameters and PDAC stromal score, after accounting for hENT1 levels. Moreover, stromal score directly correlated with CT-derived parameters. Among 110 patients who received preoperative gemcitabine-based chemoradiotherapy, CT-derived parameters correlated with pathological response and survival. CONCLUSION Gemcitabine incorporation into tumor DNA is highly variable and correlates with multiscale transport properties that can be derived from routine CT scans. Furthermore, pretherapy CT-derived properties correlate with clinically relevant endpoints. TRIAL REGISTRATION Clinicaltrials.gov NCT01276613. FUNDING Lustgarten Foundation (989161), Department of Defense (W81XWH-09-1-0212), NIH (U54CA151668, KCA088084).


Advanced Healthcare Materials | 2012

Cooperative, nanoparticle-enabled thermal therapy of breast cancer

Haifa Shen; Jian You; Guodong Zhang; Arturas Ziemys; Qingpo Li; Litao Bai; Xiaoyong Deng; Donald R. Erm; Xuewu Liu; Chun Li; Mauro Ferrari

Hollow gold nanoshells are more efficient in heat generation triggered by near infrared laser when they are loaded into porous silicon particles, which results in effective cancer-cell killing in vitro and in vivo. Collective electromagnetic coupling of nanoconfined hollow gold nanoshells leads to dramatic enhancement of thermal ablation.


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.


Colloids and Surfaces B: Biointerfaces | 2014

The nano-plasma interface: implications of the protein corona

Joy Wolfram; Yong Yang; Jianliang Shen; Asad Moten; Chunying Chen; Haifa Shen; Mauro Ferrari; Yuliang Zhao

The interactions between nanoparticles and macromolecules in the blood plasma dictate the biocompatibility and efficacy of nanotherapeutics. Accordingly, the properties of nanoparticles and endogenous biomolecules change at the nano-plasma interface. Here, we review the implications of such changes including toxicity, immunological recognition, molecular targeting, biodistribution, intracellular uptake, and drug release. Although this interface poses several challenges for nanomedicine, it also presents opportunities for exploiting nanoparticle-protein interactions.


Clinical Cancer Research | 2013

Enhancing chemotherapy response with sustained EphA2 silencing using multistage vector delivery

Haifa Shen; Cristian Rodriguez-Aguayo; Rong Xu; Vianey Gonzalez-Villasana; Junhua Mai; Yi Huang; Guodong Zhang; Xiaojing Guo; Litao Bai; Guoting Qin; Xiaoyong Deng; Qingpo Li; Donald R. Erm; Xuewu Liu; Jason Sakamoto; Arturo Chavez-Reyes; Hee Dong Han; Anil K. Sood; Mauro Ferrari; Gabriel Lopez-Berestein

Purpose: RNA interference has the potential to specifically knockdown the expression of target genes and thereby transform cancer therapy. However, lack of effective delivery of siRNA has dramatically limited its in vivo applications. We have developed a multistage vector (MSV) system, composed of discoidal porous silicon particles loaded with nanotherapeutics, that directs effective delivery and sustained release of siRNA in tumor tissues. In this study, we evaluated therapeutic efficacy of MSV-loaded EphA2 siRNA (MSV/EphA2) with murine orthotopic models of metastatic ovarian cancers as a first step toward development of a new class of nanotherapeutics for the treatment of ovarian cancer. Experimental Design: Tumor accumulation of MSV/EphA2 and sustained release of siRNA from MSV were analyzed after intravenous administration of MSV/siRNA. Nude mice with metastatic SKOV3ip2 tumors were treated with MSV/EphA2 and paclitaxel, and therapeutic efficacy was assessed. Mice with chemotherapy-resistant HeyA8 ovarian tumors were treated with a combination of MSV/EphA2 and docetaxel, and enhanced therapeutic efficacy was evaluated. Results: Treatment of SKOV3ip2 tumor mice with MSV/EphA2 biweekly for 6 weeks resulted in dose-dependent (5, 10, and 15 μg/mice) reduction of tumor weight (36%, 64%, and 83%) and number of tumor nodules compared with the control groups. In addition, tumor growth was completely inhibited when mice were treated with MSV/EphA2 in combination with paclitaxel. Furthermore, combination treatment with MSV/EphA2 and docetaxel inhibited growth of HeyA8-MDR tumors, which were otherwise resistant to docetaxel treatment. Conclusion: These findings indicate that MSV/EphA2 merits further development as a novel therapeutic agent for ovarian cancer. Clin Cancer Res; 19(7); 1806–15. ©2013 AACR.


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.

Collaboration


Dive into the Haifa Shen's collaboration.

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

Xuewu Liu

Houston Methodist Hospital

View shared research outputs
Top Co-Authors

Avatar

Jianliang Shen

Houston Methodist Hospital

View shared research outputs
Top Co-Authors

Avatar

Chaofeng Mu

Houston Methodist Hospital

View shared research outputs
Top Co-Authors

Avatar

Guodong Zhang

Houston Methodist Hospital

View shared research outputs
Top Co-Authors

Avatar

Rong Xu

Houston Methodist Hospital

View shared research outputs
Top Co-Authors

Avatar

Xiaojun Xia

Houston Methodist Hospital

View shared research outputs
Top Co-Authors

Avatar

Elvin Blanco

Houston Methodist Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge