Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 24
1.
Acta Pharm Sin B ; 13(9): 3849-3861, 2023 Sep.
Article En | MEDLINE | ID: mdl-37719382

As a representative chemotherapeutic drug, docetaxel (DTX) has been used for breast cancer treatment for decades. However, the poor solubility of DTX limits its efficacy, and the DTX based therapy increases the metastasis risk due to the upregulation of C-X-C chemokine receptor type 4 (CXCR4) expression during the treatment. Herein, we conjugated CXCR4 antagonist peptide (CTCE) with DTX (termed CTCE-DTX) as an anti-metastasis agent to treat breast cancer. CTCE-DTX could self-assemble to nanoparticles, targeting CXCR4-upregulated metastatic tumor cells and enhancing the DTX efficacy. Thus, the CTCE-DTX NPs achieved promising efficacy on inhibiting both bone-specific metastasis and lung metastasis of triple-negative breast cancer. Our work provided a rational strategy on designing peptide-drug conjugates with synergistic anti-tumor efficacy.

2.
Nanoscale ; 15(3): 1014-1023, 2023 Jan 19.
Article En | MEDLINE | ID: mdl-36602182

Otitis media (OM) is the main cause of pediatric antibiotic prescriptions. Nontypeable Haemophilus influenzae (NTHi) is a major OM pathogen, which forms a biofilm that resists conventional antimicrobials and immune clearance. Thus, novel treatments that are effective against NTHi and its biofilm are urgently required. Nanozymes (often inorganic nanoparticles) mimic natural enzymes' catalytic activities to generate strong antimicrobials at the site of infection, and thus represent one of the emerging solutions to the crisis of antimicrobial resistance. They mimic natural enzymes' activities, such as generating strong antimicrobials catalytically at the site of infection, to minimize overexposure. However, that in situ generation often relies on Reactive Oxygen Species (ROS) as precursors, a prerequisite that limits the broad deployment of nanozymes. To address this challenge, we designed a cascade nanozyme that generates an antiseptic, HOBr, from a ubiquitous non-ROS, i.e., O2, which successfully eradicates NTHi. The cascade nanozyme simultaneously exhibits glucose oxidase (GOx)-like activity from gold nanoparticles (AuNPs) and haloperoxidase (HPO)-mimicking activity from vanadium pentoxide nanowires (V2O5 NWs) connected using dopamine (DPA). The cascade nanozyme demonstrated strong antimicrobial efficacy against NTHi and its biofilm, while showing improved biocompatibility compared to the nanozyme of V2O5 NWs alone. The cascade nanozyme thus points to a material-oriented infectious disease treatment strategy, where small-molecule antimicrobials are generated in real time at the site of infection for the benefit of autonomous dosing. This strategy potentially mitigates the development of antimicrobial resistance and reduces side effects.


Anti-Infective Agents , Metal Nanoparticles , Otitis Media , Humans , Child , Gold/pharmacology , Haemophilus influenzae , Metal Nanoparticles/therapeutic use , Biofilms , Anti-Infective Agents/pharmacology
3.
ACS Biomater Sci Eng ; 9(7): 3923-3934, 2023 07 10.
Article En | MEDLINE | ID: mdl-33821617

Reduced biofilm formation is highly desirable in applications ranging from transportation to separations and healthcare. Biofilms often form at the three-phase interface where air, liquid, and solid coexist due to the close proximity to nutrients and oxygen. Reducing biofilm formation at the triple interface presents challenges because of the conflicting requirements for hydrophobicity at the air-solid interface (for self-cleaning properties) and for hydrophilicity at the liquid-solid interface (for reduced foulant adhesion). Meeting those needs simultaneously likely entails a dynamic surface, capable of shifting the surface energy landscape in response to wetting conditions and thus enabling hydrophobicity in air and hydrophilicity in water. Here, we designed a facile approach to render existing surfaces resistant to biofilm formation at the triple interface. By adding trace amounts (∼0.1 mM) of surfactants, biofilm formation of Pseudomonas aeruginosa (known to form biofilm at the triple interface) was reduced on all surfaces tested, ranging from hydrophilic to hydrophobic, polar to nonpolar. That reduced fouling was not a result of the known antimicrobial effects. Instead, it was attributed to the surface-adsorbed surfactants that dynamically control surface energy at the triple interface. To further understand the effect of surfactant-surface interactions on biofilm reduction, we systematically varied the surfactant charge type and surface properties (surface energy and charge). Electrostatic interactions between surfactants and surfaces were identified as an influential factor when predicting the relative fouling reduction upon introduction of surfactants. Nevertheless, biofilm formation was reduced even on the charge-neutral, fluorinated surface made of poly(1H, 1H, 2H, 2H-perfluorodecyl acrylate) by more than 2-fold simply via adding 0.2 mM dodecyl trimethylammonium chloride or 0.3 mM sodium dodecyl sulfate. Given its robustness, this strategy is broadly applicable for reducing fouling on existing surfaces, which in turn improves the cost-effectiveness of membrane separations and mitigates contaminations and nosocomial infections in healthcare.


Biofilms , Surface-Active Agents , Surface-Active Agents/pharmacology , Surface Properties , Sodium Dodecyl Sulfate/pharmacology , Wettability
4.
Adv Sci (Weinh) ; 10(3): e2204334, 2023 01.
Article En | MEDLINE | ID: mdl-36453580

Restoring sodium iodide symporter (NIS) expression and function remains a major challenge for radioiodine therapy in anaplastic thyroid cancer (ATC). For more efficient delivery of messenger RNA (mRNA) to manipulate protein expression, a lipid-peptide-mRNA (LPm) nanoparticle (NP) is developed. The LPm NP is prepared by using amphiphilic peptides to assemble a peptide core and which is then coated with cationic lipids. An amphiphilic chimeric peptide, consisting of nine arginine and hydrophobic segments (6 histidine, C18 or cholesterol), is synthesized for adsorption of mRNA encoding NIS in RNase-free conditions. In vitro studies show that LP(R9H6) m NP is most efficient at delivering mRNA and can increase NIS expression in ATC cells by more than 10-fold. After intratumoral injection of NIS mRNA formulated in optimized LPm NP, NIS expression in subcutaneous ATC tumor tissue increases significantly in nude mice, resulting in more iodine 131 (131 I) accumulation in the tumor, thereby significantly inhibiting tumor growth. Overall, this work designs three arginine-rich peptide nanoparticles, contributing to the choice of liposome cores for gene delivery. LPm NP can serve as a promising adjunctive therapy for patients with ATC by restoring iodine affinity and enhancing the therapeutic efficacy of radioactive iodine.


Iodine , Thyroid Carcinoma, Anaplastic , Thyroid Neoplasms , Animals , Mice , Cell Line, Tumor , Iodine Radioisotopes/therapeutic use , Lipids , Liposomes , Mice, Nude , Peptides , RNA, Messenger , Thyroid Carcinoma, Anaplastic/therapy , Thyroid Carcinoma, Anaplastic/genetics , Thyroid Carcinoma, Anaplastic/metabolism , Thyroid Neoplasms/radiotherapy , Thyroid Neoplasms/genetics , Humans
5.
Nanoscale ; 14(10): 3731-3737, 2022 Mar 10.
Article En | MEDLINE | ID: mdl-35191916

Despite the excellent antibacterial and antifouling effects of haloperoxidase (HPO)-mimicking CeO2-x nanorods, their antiviral efficiency has not been explored. Herein, we designed and synthesized CeO2-x nanorods with varying aspect ratios via the hydrothermal method. CeO2-x nanorods catalysed the oxidative bromination of Br- and H2O2 to HOBr, the kinetics of which were studied systematically using a phenol red assay. The CeO2-x nanorods with the optimized aspect ratio (i.e., 4.5) demonstrated strong antiviral efficacies against the human coronavirus OC43, with no visible toxicity to the HCT-8 host cells.


Cerium , Coronavirus OC43, Human , Nanotubes , Catalysis , Cerium/pharmacology , Hydrogen Peroxide
6.
Sci Adv ; 8(2): eabl8812, 2022 Jan 14.
Article En | MEDLINE | ID: mdl-35030022

To reduce the severe health risk and the huge economic impact associated with the fomite transmission of SARS-CoV-2, an imidazolium-based zwitterionic polymer was designed, synthesized, and demonstrated to achieve contact deactivation of a human coronavirus under dry ambient conditions that resemble fomite transmission. The zwitterionic polymer further demonstrated excellent antifouling properties, reducing the adhesion of coronavirus and the formation of bacteria biofilms under wetted conditions. The polymer was synthesized using a substrate-independent and solvent-free process, leveraging an all-dry technique named initiated chemical vapor deposition (iCVD). The broad applicability of this approach was demonstrated by applying the polymer to a range of substrates that are curved and/or with high-aspect-ratio nano/microporous structures, which remained intact after the coating process. The zwitterionic polymer and the synthesis approach reported here present an effective solution to mitigate viral transmission without the need for manual disinfection, reducing the health and economic impact of the ongoing pandemic.

7.
Nano Today ; 472022 Dec.
Article En | MEDLINE | ID: mdl-36968792

Otitis media (OM) is the main reason for pediatric antibiotic prescriptions. The current treatment mandates a rigorous regimen of multidose antibiotics over 5-10 days. The systemic antibiotic exposure and often prematurely terminated treatment due to the challenge of drug administration to young patients are believed to breed antibiotic resistance. To address these challenges, we designed a local treatment that converted a metabolic product (H2O2) of an OM pathogen (Streptococcus pneumoniae) into a potent antiseptic (HOBr), a reaction catalyzed by locally administered vanadium pentoxide nanowires. The therapeutic, HOBr, was only synthesized in the presence of the pathogen, enabling on-demand generation of therapeutics for OM treatment. Hypohalous acids are broad-spectrum and have a long history in general disinfection applications without breeding substantial drug resistance. A single dose of the nanowire formulation eradicated OM in a standard chinchilla model in 7 days with no observable tissue toxicity or negative impact on hearing sensitivity.

8.
Biomacromolecules ; 22(12): 4933-4944, 2021 12 13.
Article En | MEDLINE | ID: mdl-34694768

Biofilms are ubiquitous in nature, yet strategies to direct biofilm behavior without genetic manipulation are limited. Due to the small selection of materials that have been used to successfully grow biofilms, the availability of functional materials that are able to support growth and program microbial functions remains a critical bottleneck in the design and deployment of functional yet safe microbes. Here, we report the design of insoluble pyridine-rich polymer surfaces synthesized using initiated chemical vapor deposition, which led to modulated biofilm growth and virulence in Pseudomonas aeruginosa (PAO1). A variety of extracellular virulence factors exhibited decreased production in response to the functional polymer, most significantly biomolecules also associated with iron acquisition, validating the material design strategy reported here. This report signifies a rich potential for materials-based strategies to direct the behavior of naturally occurring biofilms, which complement the existing genetic engineering toolkits in advancing microbiology, translational medicine, and biomanufacturing.


Biofilms , Pseudomonas aeruginosa , Polymers/metabolism , Pseudomonas aeruginosa/genetics , Virulence , Virulence Factors/metabolism
9.
AIChE J ; 67(12)2021 Dec.
Article En | MEDLINE | ID: mdl-35450419

Otitis Media (OM) is the most common reason for U.S. children to receive prescribed oral antibiotics, leading to potential to cause antibiotic resistance. To minimize oral antibiotic usage, we developed polyvinylpyrrolidone-coated silver nanoparticles (AgNPs-PVP), which completely eradicated common OM pathogens, i.e., Streptococcus pneumoniae and non-typeable Haemophilus influenzae (NTHi) at 1.04µg/mL and 2.13µg/mL. The greater antimicrobial efficacy against S. pneumoniae was a result of the H2O2-producing ability of S. pneumoniae and the known synergistic interactions between H2O2 and AgNPs. To enable the sustained local delivery of AgNPs-PVP (e.g., via injection through perforated tympanic membranes), a hydrogel formulation of 18%(w/v)P407 was developed. Reverse thermal gelation of the AgNPs-PVP-P407 hydrogel could gel rapidly upon entering the warm auditory bullae and thereby sustained release of antimicrobials. This hydrogel-based local delivery system completely eradicated OM pathogens in vitro without cytotoxicity, and thus represents a promising strategy for treating bacterial OM without relying on conventional antibiotics.

11.
ACS Nano ; 13(11): 12357-12371, 2019 11 26.
Article En | MEDLINE | ID: mdl-31545587

Prostate cancer is one of the most common malignant tumors in men, and inhibiting metastasis is a key event but still a major challenge in prostate cancer treatment. Cancer-associated fibroblasts (CAFs) play an important role in prostate tumor metastasis by shaping the malignant tumor microenvironment. Herein, we constructed a CAF-targeting siRNA delivery system by loading the fibroblast activation protein-α (FAP-α) antibody onto the cell-penetrating peptide (CPP)-based nanoparticles, which specifically downregulated C-X-C motif chemokine ligand 12 (CXCL12) expression in CAFs. This regulation generated a series of changes through inactivating CAFs so that the malignant prostate tumor microenvironment was reshaped. The tumor cell invasion, migration, and tumor angiogenesis were significantly inhibited, which all contributed to the suppression of the metastasis of an orthotopic prostate tumor. This tumor microenvironment reshaping strategy via CAF targeting and inactivation provides an alternative approach for malignant prostate tumor metastasis inhibition.


Cancer-Associated Fibroblasts , Nanomedicine/methods , Prostatic Neoplasms , Tumor Microenvironment , Animals , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/metabolism , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/pharmacology , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Endopeptidases , Gelatinases/antagonists & inhibitors , Gelatinases/metabolism , Gene Silencing , Human Umbilical Vein Endothelial Cells , Humans , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Neoplasm Metastasis/prevention & control , PC-3 Cells , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA, Small Interfering , Serine Endopeptidases/metabolism , Tumor Microenvironment/drug effects , Tumor Microenvironment/physiology
12.
Angew Chem Int Ed Engl ; 58(43): 15287-15294, 2019 10 21.
Article En | MEDLINE | ID: mdl-31402550

Tumor complexity makes the development of highly sensitive tumor imaging probes an arduous task. Here, we construct a peptide-based near-infrared probe that is responsive to fibroblast activation protein-α (FAP-α), and specifically forms nanofibers on the surface of cancer-associated fibroblasts (CAFs) in situ. The assembly/aggregation-induced retention (AIR) effect results in enhanced accumulation and retention of the probe around the tumor, resulting in a 5.5-fold signal enhancement in the tumor 48 h after administration compared to that of a control molecule that does not aggregate. The probe provides a prolonged detectable window of 48 h for tumor diagnosis. The selective assembly of the probe results in a signal intensity over four- and fivefold higher in tumor than in the liver and kidney, respectively. With enhanced tumor imaging capability, this probe can visualize small tumors around 2 mm in diameter.


Gelatinases/metabolism , Membrane Proteins/metabolism , Nanofibers/chemistry , Peptides/metabolism , Serine Endopeptidases/metabolism , Spectroscopy, Near-Infrared/methods , Amino Acid Sequence , Animals , Cancer-Associated Fibroblasts/cytology , Cancer-Associated Fibroblasts/metabolism , Cell Line, Tumor , Coculture Techniques , Endopeptidases , Fluorescent Dyes/chemistry , Gelatinases/chemistry , Humans , Membrane Proteins/chemistry , Mice , Mice, Nude , Microscopy, Electron, Scanning , Neoplasms/diagnostic imaging , Peptides/chemical synthesis , Peptides/chemistry , Serine Endopeptidases/chemistry , Tissue Distribution , Transplantation, Heterologous
13.
ACS Nano ; 13(2): 2176-2189, 2019 02 26.
Article En | MEDLINE | ID: mdl-30676731

Rapidly growing cancer cells exhibit a strong dependence on iron for their survival. Thus, iron-removing drugs, iron chelators, have potential applications in cancer treatment. Deferoxamine (DFO) is an efficient iron chelator, but its short circulation half-life and ability to induce hypoxia-inducible factor 1α (HIF1α) overexpression restricts its use as an antitumor agent. In the present study, we first found that a pattern of iron-related protein expression favoring higher intracellular iron closely correlates with shorter overall and relapse-free survival in pancreatic cancer patients. We subsequently found that a combination of DFO and the HIF1α inhibitor, lificiguat (also named YC1), significantly enhanced the antitumor efficacy of DFO in vitro. We then employed transferrin receptor 1 (TFR1) targeting liposomes to codeliver DFO and YC1 to pancreatic tumors in a mouse model. The encapsulation of DFO prolonged its circulation time, improved its accumulation in tumor tissues via the enhanced permeability and retention (EPR) effect, and facilitated efficient uptake by cancer cells, which express high level of TFR1. After entering the tumor cells, the encapsulated DFO and YC1 were released to elicit a synergistic antitumor effect in subcutaneous and orthotopic pancreatic cancer xenografts. In summary, our work overcame two major obstacles in DFO-based cancer treatment through a simple liposome-based drug delivery system. This nanoencapsulation and targeting paradigm lays the foundation for future application of iron chelation in cancer therapy.


Antineoplastic Agents/pharmacology , Deferoxamine/pharmacology , Drug Delivery Systems , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Iron Chelating Agents/pharmacology , Pancreatic Neoplasms/drug therapy , Administration, Oral , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Deferoxamine/administration & dosage , Deferoxamine/chemistry , Drug Screening Assays, Antitumor , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Iron Chelating Agents/administration & dosage , Iron Chelating Agents/chemistry , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology
14.
Adv Mater ; 31(6): e1804395, 2019 Feb.
Article En | MEDLINE | ID: mdl-30549110

Recent advances in cancer immunotherapy have exploited the efficient potential of natural killer (NK) cells to kill tumor cells through antibody-dependent cell-mediated cytotoxicity (ADCC). However, this therapeutic strategy is seriously limited by tumor antigen heterogeneity since antibodies can only recognize specific antigens. In this work, modified antibodies or their Fc fragments that can target solid tumors without the necessity of specific antigen presentation on tumors are developed. Briefly, Fc fragments or therapeutic monoclonal antibodies are conjugated with the N-terminus of pH low insertion peptide so that they will selectively assemble onto the membrane of solid tumor cells via the conformational transformation of the peptide by responding to the acidic tumor microenvironment. The inserted Fc fragments or antibodies can efficiently activate NK cells, initiating ADCC and killing multiple types of tumor cells, including antigen-negative cancer cells. In vivo therapeutic results also exhibit significant efficacy on both primary solid tumors and tumor metastasis. These modified Fc fragments and antibodies present strong potential to overcome the limitation of tumor antigen heterogeneity, broadening the applications of NK cell immunotherapy on solid tumor treatment.


Antibodies, Monoclonal/pharmacokinetics , Immunoglobulin Fc Fragments/pharmacology , Killer Cells, Natural/drug effects , Animals , Antibody-Dependent Cell Cytotoxicity/drug effects , Cell Line, Tumor , Cell Membrane/drug effects , Cell Survival/drug effects , Humans , Hydrogen-Ion Concentration , Immunotherapy/methods , Mice , Protein Conformation , Protein Multimerization/drug effects , Signal Transduction , Tumor Microenvironment
15.
ACS Nano ; 12(11): 10785-10796, 2018 11 27.
Article En | MEDLINE | ID: mdl-30407790

Pancreatic cancer (PCa) is one of the most lethal malignancies, with a 5 year survival rate of less than 8%. Current treatment regiments have a low response rate in unselected patients. However, the subgroup of PCa patients with BRCA mutations may benefit from poly-ADP-ribose polymerase inhibitors (PARPi) due to their biological properties in DNA repair. Dose-limiting toxicity in normal tissues is frequently observed when PARPi are combined with other chemotherapies, and the co-delivery of two drugs to tumor sites at an adequate concentration is challenging. To address this issue, we have engineered an epidermal growth factor receptor (EGFR) targeting (with GE11 peptide) self-assembly amphiphilic peptide nanoparticle (GENP) to co-deliver gemcitabine and the PARPi olaparib to treat BRCA mutant PCa. The GENP was relatively stable, exhibited high encapsulation efficiency, and could coordinately release the two drugs in tumor milieu. Gemcitabine and olaparib showed strong synergistic actions in optimized conditions in vitro. The nanoparticle prolonged the half-life of both drugs and resulted in their tumor accumulation at the optimal therapeutic ratio in vivo. The drug-loaded nanoparticles were able to significantly suppress tumor growth in a murine PCa model with minimal side effects. Drug co-delivery of DNA damaging agents and PARP inhibitors via the GENP represents a promising approach for treatment of pancreatic cancers with molecular defects in the DNA repair pathway.


BRCA2 Protein/genetics , Deoxycytidine/analogs & derivatives , Drug Delivery Systems , Nanoparticles/chemistry , Pancreatic Neoplasms/drug therapy , Phthalazines/administration & dosage , Piperazines/administration & dosage , Animals , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , BRCA2 Protein/metabolism , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacokinetics , Deoxycytidine/therapeutic use , ErbB Receptors/metabolism , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Mutation , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Peptides/chemistry , Phthalazines/pharmacokinetics , Phthalazines/therapeutic use , Piperazines/pharmacokinetics , Piperazines/therapeutic use , Gemcitabine
16.
Cancer Lett ; 431: 171-181, 2018 09 01.
Article En | MEDLINE | ID: mdl-29870774

Mutant KRAS is a known driver oncogene in pancreatic cancer. However, this protein remains an "undruggable" therapeutic target. Inhibiting mutated KRAS expression at the mRNA level is a potentially effective strategy. Recently, a novel CRISPR-Cas effector, Cas13a has been reported to specifically knock down mRNA expression under the guidance of a single CRISPR-RNA in mammalian cells. Here we demonstrate that the CRISPR-Cas13a system can be engineered for targeted therapy of mutant KRAS in pancreatic cancer. In initial screening, we show that the bacterial Cas13a protein and crRNA significantly knock down mutant KRAS mRNA expression, identifying a CRISPR-Cas13a system that can induce up to a 94% knockdown efficiency. Introducing a single mismatch into the crRNA-target duplex enabled the CRISPR-Cas13a system to specifically recognize KRAS-G12D mRNA with no detectable effects on wild-type KRAS mRNA. More importantly, CRISPR-Cas13a-mediated KRAS-G12D mRNA knockdown potently induced apoptosis in vitro and elicited marked tumor shrinkage in mice. Our work describes an optimization strategy for the development of a CRISPR-Cas13a system to affect efficient and specific knockdown of the oncogenic mRNA, establishing the CRISPR-Cas13a system as a flexible, targeted therapeutic tool.


CRISPR-Cas Systems , Mutation , Oncogenes , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/therapy , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation , Cell Survival , Female , Humans , Mice , Mice, Nude , Neoplasm Transplantation , RNA Interference , RNA, Messenger/metabolism , Signal Transduction
17.
Biomaterials ; 158: 44-55, 2018 03.
Article En | MEDLINE | ID: mdl-29275122

Low chemosensitivity considerably restricts the therapeutic efficacy of gemcitabine (GEM) in pancreatic cancer treatment. Using immunohistochemical evaluation, we investigated that decreased expression of human equilibrative nucleoside transporter-1 (hENT1, which is the major GEM transporter across cell membranes) and increased expression of ribonucleotide reductase subunit 2 (RRM2, which decreases the cytotoxicity of GEM) was associated with low GEM chemosensitivity. To solve these problems, we employed a nanomedicine-based formulation of cationic liposomes for co-delivery of GEM along with siRNA targeting RRM2. Due to the specific endocytic uptake mechanism of nanocarriers and gene-silencing effect of RRM2 siRNA, this nanomedicine formulation significantly increased GEM chemosensitivity in tumor models of genetically engineered Panc1 cells with low hENT1 or high RRM2 expression. Moreover, in a series of patient-derived cancer cells, we demonstrated that the therapeutic benefits of the nanomedicine formulations were associated with the expression levels of hENT1 and RRM2. In summary, we found that the essential factors of GEM chemosensitivity were the expression levels of hENT1 and RRM2, and synthesized nanoformulations can overcome these problems. This unique design of nanomedicine not only provides a universal platform to enhance chemosensitivity but also contributes to the precision design and personalized treatment in nanomedicine.


Carcinoma, Pancreatic Ductal/drug therapy , Deoxycytidine/analogs & derivatives , Liposomes , Animals , Cell Line, Tumor , Deoxycytidine/metabolism , Deoxycytidine/pharmacology , Equilibrative Nucleoside Transporter 1/genetics , Equilibrative Nucleoside Transporter 1/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Liposomes/chemistry , Mice , Precision Medicine/methods , RNA, Small Interfering/genetics , Ribonucleotide Reductases/genetics , Ribonucleotide Reductases/metabolism , Gemcitabine
18.
ACS Appl Mater Interfaces ; 10(3): 2347-2353, 2018 Jan 24.
Article En | MEDLINE | ID: mdl-29286239

Aerobic glycolysis enables cancer cells to rapidly take up nutrients (e.g., nucleotides, amino acids, and lipids) and incorporate them into the biomass needed to produce a new cell. In contrast to existing chemotherapy/radiotherapy strategies, inhibiting aerobic glycolysis to limit the adenosine 5'-triphosphate (ATP) yield is a highly efficient approach for suppressing tumor cell proliferation. However, most, if not all, current inhibitors of aerobic glycolysis cause significant adverse effects because of their nonspecific delivery and distribution to nondiseased organs, low bioavailability, and a narrow therapeutic window. New strategies to enhance the biosafety and efficacy of these inhibitors are needed for moving them into clinical applications. To address this need, we developed a liposomal nanocarrier functionalized with a well-validated tumor-targeting peptide to specifically deliver the aerobic glycolysis inhibitor 3-bromopyruvate (3-BP) into the tumor tissue. The nanoparticles effectively targeted tumors after systemic administration into tumor-bearing mice and suppressed tumor growth by locally releasing 3-BP to inhibit the ATP production of the tumor cells. No overt side effects were observed in the major organs. This report demonstrates the potential utility of the nanoparticle-enabled delivery of an aerobic glycolysis inhibitor as an anticancer therapeutic agent.


Neoplasms , Adenosine Triphosphate , Animals , Cell Line, Tumor , Cell Proliferation , Glycolysis , Liposomes , Mice , Nanoparticles
19.
ACS Nano ; 11(9): 8668-8678, 2017 09 26.
Article En | MEDLINE | ID: mdl-28806504

During pancreatic tumor development, pancreatic stellate cells (PSCs) proliferate exuberantly to secrete extracellular matrix (ECM) in the tumor stroma, which presents major barriers for drug delivery and penetration in tumor tissue. Thus, down-regulating ECM levels via regulation of the PSCs may allow enhanced penetration of therapeutic drugs and thereby enhancing their therapeutic efficacy. To regulate the PSCs, a matrix metalloproteinase-2 (MMP-2) responsive peptide-hybrid liposome (MRPL) was constructed via coassembly of a tailor-designed MMP-2 responsive amphiphilic peptide and phospholipids. By utilizing the MMP-2-rich pathological environment, the pirfenidone (PFD) loaded MRPL (MRPL-PFD) can specifically release PFD at the pancreatic tumor site and down-regulate the multiple components of ECM expressed by the PSCs. This resulted in a significant increase in the penetration of gemcitabine into the tumor tissue and enhanced the efficacy of gemcitabine for pancreatic tumor. Our design tailored for antifibrosis of pancreatic cancer may provide a practical approach to build functional liposomes through supramolecular assembly, and regulation of ECM may be a promising adjuvant therapeutic strategy for pancreatic and other ECM-rich tumors.


Antineoplastic Agents/administration & dosage , Deoxycytidine/analogs & derivatives , Extracellular Matrix/drug effects , Liposomes/metabolism , Matrix Metalloproteinase 2/metabolism , Pancreatic Neoplasms/drug therapy , Pyridones/administration & dosage , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacokinetics , Deoxycytidine/therapeutic use , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Humans , Liposomes/chemistry , Mice, Nude , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/drug effects , Pancreatic Stellate Cells/metabolism , Pancreatic Stellate Cells/pathology , Peptides/chemistry , Peptides/metabolism , Pyridones/pharmacokinetics , Pyridones/therapeutic use , Tumor Cells, Cultured , Gemcitabine
20.
Theranostics ; 7(5): 1062-1071, 2017.
Article En | MEDLINE | ID: mdl-28435448

Extensive evidence has shown that platelets support tumor metastatic progression by inducing epithelial-mesenchymal transition of cancer cells and by shielding circulating tumor cells from immune-mediated elimination. Therefore, blocking platelet function represents a potential new avenue for therapy focused on eliminating metastasis. Here we show that liposomal nanoparticles bearing the tumor-homing pentapeptide CREKA (Cys-Arg-Glu-Lys-Ala) can deliver a platelet inhibitor, ticagrelor, into tumor tissues to specifically inhibit tumor-associated platelets. The drug-loaded nanoparticles (CREKA-Lipo-T) efficiently blocked the platelet-induced acquisition of an invasive phenotype by tumor cells and inhibited platelet-tumor cell interaction in vitro. Intravenously administered CREKA-Lipo-T effectively targeted tumors within 24 h, and inhibited tumor metastasis without overt side effects. Thus, the CREKA-Lipo formulation provides a simple strategy for the efficient delivery of anti-metastatic drugs and shows considerable promise as a platform for novel cancer therapeutics.


Adenosine/analogs & derivatives , Antineoplastic Agents/pharmacokinetics , Blood Platelets/drug effects , Liposomes/administration & dosage , Nanoparticles/administration & dosage , Neoplasm Metastasis/prevention & control , Oligopeptides/metabolism , Adenosine/administration & dosage , Adenosine/pharmacokinetics , Animals , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Mammary Neoplasms, Experimental/drug therapy , Mice, Inbred BALB C , Ticagrelor , Treatment Outcome
...