Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Mol Pharm ; 21(5): 2081-2096, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38630656

ABSTRACT

Small interfering RNAs (siRNAs) are promising therapeutic strategies, and five siRNA drugs have been approved by the Food and Drug Administration (FDA) and the European Commission (EC). This marks a significant milestone in the development of siRNA for clinical applications. The approved siRNA agents can effectively deliver siRNAs to the liver and treat liver-related diseases. Currently, researchers have developed diverse delivery platforms for transporting siRNAs to different tissues such as the brain, lung, muscle, and others, and a large number of siRNA drugs are undergoing clinical trials. Here, these delivery technologies and the latest advancements in clinical applications are summarized, and this Review provides a concise overview of the strategies employed for siRNA delivery to both hepatic and extrahepatic tissues.


Subject(s)
RNA, Small Interfering , RNA, Small Interfering/administration & dosage , Humans , Animals , Drug Delivery Systems/methods , Gene Transfer Techniques , Liver/metabolism , RNA Interference , Nanoparticles/chemistry , United States Food and Drug Administration , Clinical Trials as Topic
2.
Article in English | MEDLINE | ID: mdl-38457025

ABSTRACT

Colorectal cancer (CRC) is the fourth most common cancer in the world, with the second highest incidence rate after lung cancer. Oxaliplatin (OXA) is a broad-spectrum anti-tumor agent with significant therapeutic efficacy in colorectal cancer, and as a divalent platinum analog, it is not selective in its distribution in the body and has systemic toxicity with continued use. Interleukin-12 (IL12) is an immunostimulatory cytokine with cytokine monotherapy that has made advances in the fight against cancer, limiting the clinical use of cytokines due to severe toxicity. Here, we introduced a long alkyl chain and N-methyl-2,2-diaminodiethylamine to the ligand of OXA to obtain OXA-LIP, which effectively reduces its toxicity and improves the uptake of the drug by tumor cells. We successfully constructed IL12 mRNA and used LNPs to deliver IL12 mRNA, and in vivo pharmacodynamic studies demonstrated that OXA-LIP combined with IL12 mRNA had better tumor inhibition and higher biosafety. In addition, it was investigated by pharmacokinetic experiments that the OXA-LIP drug could accumulate in nude mice at the tumor site, which prolonged the half-life and enhanced the anti-tumor efficiency of OXA. It is hoped that these results will provide an important reference for the subsequent research and development of OXA-LIP with IL12 mRNA, as well as provide new therapeutic approaches for the treatment of colon cancer.

3.
MedComm (2020) ; 5(2): e481, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38344397

ABSTRACT

Drug development is a long and costly process, with a high degree of uncertainty from the identification of a drug target to its market launch. Targeted drugs supported by human genetic evidence are expected to enter phase II/III clinical trials or be approved for marketing more quickly, speeding up the drug development process. Currently, genetic data and technologies such as genome-wide association studies (GWAS), whole-exome sequencing (WES), and whole-genome sequencing (WGS) have identified and validated many potential molecular targets associated with diseases. This review describes the structure, molecular biology, and drug development of human genetics-based validated beneficial loss-of-function (LOF) mutation targets (target mutations that reduce disease incidence) over the past decade. The feasibility of eight beneficial LOF mutation targets (PCSK9, ANGPTL3, ASGR1, HSD17B13, KHK, CIDEB, GPR75, and INHBE) as targets for drug discovery is mainly emphasized, and their research prospects and challenges are discussed. In conclusion, we expect that this review will inspire more researchers to use human genetics and genomics to support the discovery of novel therapeutic drugs and the direction of clinical development, which will contribute to the development of new drug discovery and drug repurposing.

4.
Acta Pharmacol Sin ; 44(1): 244-254, 2023 Jan.
Article in English | MEDLINE | ID: mdl-35705687

ABSTRACT

The combination of vascular endothelial growth factor (VEGF) inhibitors and tyrosine kinase inhibitors (TKIs) is newly available for molecular targeted therapy against non-small cell lung cancer (NSCLC) in clinic. However, the therapeutic benefits remain unsatisfying due to the poor drug delivery to targets of interest. In this study, we developed bevacizumab-coated gefitinib-loaded nanoparticles (BCGN) with dual-responsive drug release for inhibiting tumor angiogenesis and phosphorylation of epidermal growth factor receptor (EGFR). Through an exogenous corona strategy, bevacizumab is easily coated on gefitinib-loaded nanoparticles via electrostatic interaction. After intravenous injection, BCGN are efficiently accumulated in NSCLC tumors as confirmed by dual-model imaging. Bevacizumab is released from BCGN upon oxidation in tumor microenvironment, whereas gefitinib is released after being internalized by tumor cells and disassembled in reduction cytoplasm. The dual-responsive release of bevacizumab and gefitinib significantly inhibits tumor growth in both A549 and HCC827 human NSCLC models. Our approach provides a promising strategy to improve combinational molecular targeted therapy of NSCLC with precisely controlled drug release.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/metabolism , Gefitinib , Bevacizumab/therapeutic use , Lung Neoplasms/pathology , Vascular Endothelial Growth Factor A , Molecular Targeted Therapy , Quinazolines/pharmacology , Drug Resistance, Neoplasm , Cell Line, Tumor , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Tumor Microenvironment
5.
Signal Transduct Target Ther ; 7(1): 386, 2022 12 02.
Article in English | MEDLINE | ID: mdl-36460660

ABSTRACT

Bioorthogonal chemistry reactions occur in physiological conditions without interfering with normal physiological processes. Through metabolic engineering, bioorthogonal groups can be tagged onto cell membranes, which selectively attach to cargos with paired groups via bioorthogonal reactions. Due to its simplicity, high efficiency, and specificity, bioorthogonal chemistry has demonstrated great application potential in drug delivery. On the one hand, bioorthogonal reactions improve therapeutic agent delivery to target sites, overcoming off-target distribution. On the other hand, nanoparticles and biomolecules can be linked to cell membranes by bioorthogonal reactions, providing approaches to developing multi-functional drug delivery systems (DDSs). In this review, we first describe the principle of labeling cells or pathogenic microorganisms with bioorthogonal groups. We then highlight recent breakthroughs in developing active targeting DDSs to tumors, immune systems, or bacteria by bioorthogonal chemistry, as well as applications of bioorthogonal chemistry in developing functional bio-inspired DDSs (biomimetic DDSs, cell-based DDSs, bacteria-based and phage-based DDSs) and hydrogels. Finally, we discuss the difficulties and prospective direction of bioorthogonal chemistry in drug delivery. We expect this review will help us understand the latest advances in the development of active targeting and multi-functional DDSs using bioorthogonal chemistry and inspire innovative applications of bioorthogonal chemistry in developing smart DDSs for disease treatment.


Subject(s)
Bacteriophages , Nanoparticles , Prospective Studies , Drug Delivery Systems , Cell Membrane
6.
Adv Mater ; 34(33): e2205462, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35759925

ABSTRACT

Lung metastasis is challenging in patients with triple-negative breast cancer (TNBC). Surgery is always not available due to the dissemination of metastatic foci and most drugs are powerless because of poor retention at metastatic sites. TNBC cells generate an inflamed microenvironment and overexpress adhesive molecules to promote invasion and colonization. Herein, "walking dead" TNBC cells are developed through conjugating anti-PD-1 (programmed death protein 1 inhibitor) and doxorubicin (DOX)-loaded liposomes onto cell corpses for temporal chemo-immunotherapy against lung metastasis. The walking dead TNBC cells maintain plenary tumor antigens to conduct vaccination effects. Anti-PD-1 antibodies are conjugated to cell corpses via reduction-activated linker, and DOX-loaded liposomes are attached by maleimide-thiol coupling. This anchor strategy enables rapid release of anti-PD-1 upon reduction conditions while long-lasting release of DOX at inflamed metastatic sites. The walking dead TNBC cells improve pulmonary accumulation and local retention of drugs, reprogram the lung microenvironment through damage-associated molecular patterns (DAMPs) and PD-1 blockade, and prolong overall survival of lung metastatic 4T1 and EMT6-bearing mice. Taking advantage of the walking dead TNBC cells for pulmonary preferred delivery of chemotherapeutics and checkpoint inhibitors, this study suggests an alternative treatment option of chemo-immunotherapy to augment the efficacy against lung metastasis.


Subject(s)
Lung Neoplasms , Triple Negative Breast Neoplasms , Animals , Cadaver , Cell Line, Tumor , Humans , Liposomes/therapeutic use , Lung Neoplasms/drug therapy , Mice , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Tumor Microenvironment
7.
Acta Pharm Sin B ; 10(11): 2246-2257, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33304789

ABSTRACT

Hypoxia is a serious impediment to current treatments of many malignant tumors. Catalase, an antioxidant enzyme, is capable of decomposing endogenous hydrogen peroxide (H2O2) into oxygen for tumor reoxygenation, but suffered from in vivo instability and limited delivery to deep interior hypoxic regions in tumor. Herein, a deep-penetrated nanocatalase-loading DiIC18 (5, DiD) and soravtansine (Cat@PDS) were provided by coating catalase nanoparticles with PEGylated phospholipids membrane, stimulating the structure and function of erythrocytes to relieve tumor hypoxia for enhanced chemo-photodynamic therapy. After intravenous administration, Cat@PDS preferentially accumulated at tumor sites, flexibly penetrated into the interior regions of tumor mass and remarkably relieved the hypoxic status in tumor. Notably, the Cat@PDS + laser treatment produced striking inhibition of tumor growth and resulted in a 97.2% suppression of lung metastasis. Thus, the phospholipids membrane-coated nanocatalase system represents an encouraging nanoplatform to relieve tumor hypoxia and synergize the chemo-photodynamic cancer therapy.

8.
J Control Release ; 323: 59-70, 2020 07 10.
Article in English | MEDLINE | ID: mdl-32289329

ABSTRACT

Oral chemotherapy offers a more convenient treatment option for cancer patients but the effectiveness is significantly hindered by the limited drug delivery efficiency. Herein, we designed legumain-activable melittin (LM) decorated polymeric nanovehicles loading IR-780 and sorafenib (LPN) to enhance their oral delivery to tumors, with efficient accumulation and penetration capacity, for combinational photothermal-chemotherapy of gastric cancer. The nanosized LPN displayed good stability in simulated gastrointestinal fluids. After oral administration, the oral bioavailability of sorafenib was remarkably improved (75.9-fold by LPN versus free drug suspension). Moreover, the orally administered LPN could preferentially accumulate at the tumor site and penetrate into the interior regions of the tumor mass. Upon combination with laser irradiation, LPN produced notable inhibition of tumor growth, which was more effective than the counterpart unmodified nanovehicles. Therefore, LPN represents an encouraging oral delivery nanoplatform with favorable tumor accumulation and penetration capability for oral combinational cancer therapy.


Subject(s)
Antineoplastic Agents , Neoplasms , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cysteine Endopeptidases , Drug Delivery Systems , Humans , Neoplasms/drug therapy
9.
Acta Pharm Sin B ; 10(3): 529-545, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32140397

ABSTRACT

The limited penetration of nanoparticles and their poor accessibility to cancer cell fractions in tumor remain essential challenges for effective anticancer therapy. Herein, we designed a targeting peptide-decorated biomimetic lipoprotein (termed as BL-RD) to enable their deep penetration and efficient accessibility to cancer cell fractions in a tumor, thereby improving the combinational chemo-photodynamic therapy of triple negative breast cancer. BL-RD was composed of phospholipids, apolipoprotein A1 mimetic peptide (PK22), targeting peptide-conjugated cytotoxic mertansine (RM) and photodynamic agents of DiIC18(5) (DiD). The counterpart biomimetic lipoprotein system without RM (termed as BL-D) was fabricated as control. Both BL-D and BL-RD were nanometer-sized particles with a mean diameter of less than 30 nm and could be efficiently internalized by cancer cells. After intravenous injection, they can be specifically accumulated at tumor sites. When comparing to the counterpart BL-D, BL-RD displayed superior capability to permeate across the tumor mass, extravasate from tumor vasculature to distant regions and efficiently access the cancer cell fractions in a solid tumor, thus producing noticeable depression of the tumor growth. Taken together, BL-RD can be a promising delivery nanoplatform with prominent tumor-penetrating and cancer cells-accessing capability for effective tumor therapy.

13.
Adv Sci (Weinh) ; 5(12): 1801012, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30581704

ABSTRACT

Cancer stem cells (CSCs) are proposed to account for the initiation of cancer metastasis, but their accessibility remains a great challenge. This study reports deep tumor-penetrated biomimetic nanocages to augment the accessibility to CSCs fractions in tumor for anti-metastasis therapy. The nanocages can load photothermal agent of 1,1-dioctadecyl-3,3,3,3-tetramethylindotricarbocyanine iodide (DBN) and chemotherapeutic epirubicin (EBN) to eradicate CSCs for photothermal-chemotherapy of breast cancer metastasis. In metastatic 4T1-indcued tumor model, both DBN and EBN can efficiently accumulate in tumor sites and feasibly permeate throughout the tumor mass. These biomimetic nanosystems can be preferentially internalized by cancer cells and effectively accessed to CSCs fractions in tumor. The DBN+laser/EBN treatment produces considerable depression of primary tumor growth, drastically eradicates around 80% of CSCs fractions in primary tumor, and results in 95.2% inhibition of lung metastasis. Thus, the biomimetic nanocages can be a promising delivery nanovehicle with preferential CSCs-accessibility for effective anti-metastasis therapy.

14.
Int J Pharm ; 553(1-2): 201-209, 2018 Dec 20.
Article in English | MEDLINE | ID: mdl-30339945

ABSTRACT

Cancer stem-like cells (CSCs) are proposed to be responsible for tumor metastasis, resistance and relapse after therapy, but are unable to be eliminated by many current therapies. Herein, we report that the apoferritin nanocages loading cytotoxic mertansine (M-AFN) can significantly improve their uptake in CSCs-enriched tumorspheres and effectively eradicate CSCs in tumorspheres for anticancer therapy. M-AFN were uniformly nanocage structures with the mean diameter of 11.26 ±â€¯2.58 nm and the loading capacity of 0.62%. In the CSCs-enriched tumorsphere model, M-AFN could be preferentially internalized by tumorsphere cells and the average half-inhibitory concentration (IC50) of M-AFN was obviously reduced by 5.46-fold when comparing to the parent 4T1 breast cancer cells. Moreover, both the already existing tumorspheres and the formation of secondary tumorspheres were drastically disrupted by M-AFN, but barely impacted by mertansine alone. The flow cytometer analysis showed the CSCs fractions in tumorspheres were considerably reduced by the M-AFN treatment. Therefore, the apoferritin nanocages represent an encouraging nanoplatform to eradicate CSCs for effective anticancer therapy.


Subject(s)
Apoferritins/chemistry , Maytansine/pharmacology , Nanostructures , Neoplastic Stem Cells/drug effects , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Female , Flow Cytometry , Inhibitory Concentration 50 , Mammary Neoplasms, Animal/drug therapy , Mammary Neoplasms, Animal/pathology , Maytansine/administration & dosage , Maytansine/chemistry , Mice , NIH 3T3 Cells , Particle Size
15.
Nano Lett ; 18(8): 4762-4770, 2018 08 08.
Article in English | MEDLINE | ID: mdl-30028623

ABSTRACT

Specific drug delivery to metastatic tumors remains a great challenge for antimetastasis therapy. We herein report a bioengineered macrophage-based delivery system (LD-MDS) that can be preferentially delivered to lung metastases and intelligently transformed into nanovesicles and secondary nanovesicles for antimetastasis therapy. LD-MDS was prepared by anchoring a legumain-specific propeptide of melittin (legM) and cytotoxic soravtansine (DM4) prodrug onto the membrane of living macrophages. LD-MDS is responsively activated by legumain protease and converted into DM4-loaded exosome-like nanovesicles (DENs), facilitating efficient internalization by metastatic 4T1 cancer cells and considerable cell death. Afterward, the damaged 4T1 cells can release secondary nanovesicles and free drug molecules to destroy neighboring cancer cells. In vivo, LD-MDS displays superior targeting efficiency for lung metastatic lesions with diameters less than 100 µm and remarkably inhibits lung metastasis. This study provides a new opportunity to explore endogenous macrophages as living drug delivery vehicles with controlled drug release to target metastatic lung tumors.


Subject(s)
Antineoplastic Agents/chemistry , Drug Carriers/chemistry , Lung Neoplasms/drug therapy , Macrophages/chemistry , Nanoparticles/chemistry , Animals , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Cell Survival/drug effects , Cysteine Endopeptidases/metabolism , Drug Liberation , Humans , Lung Neoplasms/pathology , Macrophages/cytology , Maytansine/administration & dosage , Maytansine/chemistry , Melitten/administration & dosage , Melitten/chemistry , Mice, Nude , Neoplasm Metastasis , Prodrugs/administration & dosage , Prodrugs/chemistry
16.
Nano Lett ; 17(9): 5546-5554, 2017 09 13.
Article in English | MEDLINE | ID: mdl-28758755

ABSTRACT

Metastasis causes high mortality of breast cancer, and the inability of drug delivery to metastatic sites remains a crucial challenge for antimetastasis therapy. Herein, we report that inflammatory monocytes loading legumain-activated nanoparticles can actively target lung metastases and initiate metastasis-specific intelligent drug release for antimetastasis therapy. The cytotoxic mertansine is conjugated to poly(styrene-co-maleic anhydride) with a legumain-sensitive peptide and self-assembled into nanoparticles (SMNs), and then loaded into inflammatory monocytes to prepare the SMNs-loaded monocytes delivery system (M-SMNs). M-SMNs would be in living state in circulation to ensure their active targeting to lung metastases, and responsively damaged at the metastatic sites upon the differentiation of monocytes into macrophages. The anticancer drugs are intelligently released from M-SMNs as free drug molecules and drug-loaded microvesicles, resulting in considerable inhibition on the proliferation, migration, and invasion activities of metastatic 4T1 breast cancer cells. Moreover, M-SMNs significantly improve the delivery to lung metastases and penetrate the metastatic tumors, thus producing a 77.8% inhibition of lung metastases. Taken together, our findings provide an intelligent biomimetic drug delivery strategy via the biological properties of inflammatory monocytes for effective antimetastasis therapy.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Breast Neoplasms/pathology , Cysteine Endopeptidases/administration & dosage , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Maytansine/administration & dosage , Monocytes/immunology , Neoplasm Invasiveness/prevention & control , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Cysteine Endopeptidases/therapeutic use , Drug Delivery Systems/methods , Drug Liberation , Female , Humans , Lung Neoplasms/immunology , Maytansine/therapeutic use , Mice , Monocytes/transplantation , Neoplasm Invasiveness/immunology
17.
Acta Pharmacol Sin ; 38(6): 924-930, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28504249

ABSTRACT

Cancer metastasis is the primary cause of high mortality in breast cancer patients. In this study, we loaded an anti-cancer drug, cabazitaxel (CTX), into polymeric micelles (CTX-loaded polymeric micelles, PCMs), and explored their therapeutic efficacy in breast cancer metastasis. The characteristics of PCMs were investigated, and their anti-metastatic efficacy was assessed using in vitro and in vivo evaluations. PCMs had an average diameter of 50.13±11.96 nm with a CTX encapsulation efficiency of 97.02%±0.97%. PCMs could be effectively internalized into metastatic 4T1 breast cancer cells in vitro. CTX (10 ng/mL) or an equivalent concentration in PCMs did not significantly affected the viability of 4T1 cells, but dramatically decreased the cell migration activities. In an orthotopic metastatic breast cancer model, intravenously administered PCMs could be efficiently delivered to the tumor sites, resulting in a 71.6% inhibition of tumor growth and a 93.5% reduction of lung metastases. Taken together, our results verify the anti-metastatic efficacy of PCMs, thus providing an encouraging strategy for treating breast cancer metastasis.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Lactates/chemistry , Polyethylene Glycols/chemistry , Taxoids/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Breast Neoplasms/pathology , Breast Neoplasms/secondary , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Humans , Lactates/administration & dosage , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/secondary , Mice , Mice, Inbred BALB C , Mice, Nude , Micelles , Particle Size , Polyethylene Glycols/administration & dosage , Structure-Activity Relationship , Surface Properties , Taxoids/administration & dosage , Taxoids/chemistry
18.
ACS Nano ; 10(8): 7738-48, 2016 08 23.
Article in English | MEDLINE | ID: mdl-27454827

ABSTRACT

Cancer metastasis leads to high mortality of breast cancer and is difficult to treat because of the poor delivery efficiency of drugs. Herein, we report the wrapping of a drug-carrying liposome with an isolated macrophage membrane to improve delivery to metastatic sites. The macrophage membrane decoration increased cellular uptake of the emtansine liposome in metastatic 4T1 breast cancer cells and had inhibitory effects on cell viability. In vivo, the macrophage membrane enabled the liposome to target metastatic cells and produced a notable inhibitory effect on lung metastasis of breast cancer. Our results provide a biomimetic strategy via the biological properties of macrophages to enhance the medical performance of a nanoparticle in vivo for treating cancer metastasis.


Subject(s)
Breast Neoplasms/pathology , Liposomes , Lung Neoplasms/secondary , Macrophages , Nanoparticles , Animals , Cell Line, Tumor , Humans , Mice , Mice, Inbred BALB C
19.
Theranostics ; 6(6): 762-72, 2016.
Article in English | MEDLINE | ID: mdl-27162548

ABSTRACT

Cancer metastasis accounts for the high mortality of many types of cancer. Owing to the unique advantages of high specificity and minimal invasiveness, photothermal therapy (PTT) has been evidenced with great potential in treating cancer metastasis. In this review, we outline the current approaches of PTT with respect to its application in treating metastatic cancer. PTT can be used alone, guided with multimodal imaging, or combined with the current available therapies for effective treatment of cancer metastasis. Numerous types of photothermal nanotherapeutics (PTN) have been developed with encouraging therapeutic efficacy on metastatic cancer in many preclinical animal experiments. We summarize the design and performance of various PTN in PTT alone and their combinational therapy. We also point out the lacking area and the most promising approaches in this challenging field. In conclusion, PTT or their combinational therapy can provide an essential promising therapeutic modality against cancer metastasis.


Subject(s)
Hyperthermia, Induced/methods , Nanomedicine/methods , Nanostructures/administration & dosage , Neoplasm Metastasis/therapy , Neoplasms/therapy , Phototherapy/methods , Humans
20.
Theranostics ; 6(3): 435-45, 2016.
Article in English | MEDLINE | ID: mdl-26909117

ABSTRACT

Cancer metastasis is the leading reason for the high mortality of breast cancer. Herein, we report on a pH-responsive host-guest nanosystem of succinobucol (PHN) with pH-stimuli controlled drug release behavior to improve the therapeutic efficacy on lung metastasis of breast cancer. PHN was composed of the host polymer of ß-cyclodextrin linked with multiple arms of N,N-diisopropylethylenediamine (ßCD-DPA), the guest polymer of adamantyl end-capped methoxy poly(ethylene glycol) (mPEG-Ad), and the active agent of succinobucol. PHN comprises nanometer-sized homogenous spherical particles, and exhibits specific and rapid drug release in response to the intracellular acidic pH-stimuli. Then, the anti-metastatic efficacy of PHN is measured in metastatic 4T1 breast cancer cells, which effectively confirms the superior inhibitory effects on cell migration and invasion activities, VCAM-1 expression and cell-cell binding of RAW 264.7 to 4T1 cells. Moreover, PHN can be specifically delivered to the sites of metastatic nodules in lungs, and result in an obviously improved therapeutic efficacy on lung metastasis of breast cancer. Thereby, the pH-responsive host-guest nanosystem can be a promising drug delivery platform for effective treatment of cancer metastasis.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Breast Neoplasms/complications , Drug Delivery Systems , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Probucol/analogs & derivatives , Animals , Antineoplastic Agents/administration & dosage , Cell Movement/drug effects , Disease Models, Animal , Female , Hydrogen-Ion Concentration , Mice, Inbred BALB C , Nanostructures/administration & dosage , Nanostructures/chemistry , Neoplasm Invasiveness , Polyethylene Glycols/metabolism , Probucol/administration & dosage , Probucol/pharmacokinetics , Treatment Outcome , beta-Cyclodextrins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...