Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Acta Biomater ; 99: 339-349, 2019 11.
Article in English | MEDLINE | ID: mdl-31499197

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) is a highly fatal disease with 5-year survival of ∼8.5%. Nanoplatforms such as nab-paclitaxel and nanoliposomal irinotecan demonstrate superiority and utility in treating different progressions of PDA by prolonging the median overall survival by only a few months. Due to the dense surrounding stroma and the high autophagy in pancreatic cancer, integrin ɑvß3 targeting, acid environmental sensitive, TR peptide-modified liposomal platforms loaded with combined autophagy inhibiting hydroxychloroquine (HCQ), and cytotoxic paclitaxel (PTX) were designed (TR-PTX/HCQ-Lip) to accomplish the aim of synergistically killing tumor cells while inhibiting stroma fibrosis. The results showed that TR peptide-modified liposomes (TR-Lip) have superior targeting and penetrating effects both in vitro and in vivo. TR-PTX/HCQ-Lip efficiently inhibited autophagy in pancreatic cells and surrounding cancer-associated fibroblasts. The synergistic anti-fibrosis roles were also confirmed both in vitro and in vivo, all of which contributes to the enhanced curative effects of TR-PTX/HCQ-Lip in both heterogenetic and orthotopic pancreatic cancer models. STATEMENT OF SIGNIFICANCE: Autophagy plays a significant role in pancreatic ductal adenocarcinoma, especially in activating cancer associated fibroblasts which is also related to collagen generation that promotes the formation of dense stroma, which hinder the cytotoxic drugs to target and kill cancer cells. In this study, we designed integrin ɑvß3 targeting, acid environmental sensitive liposomal platforms to co-loaded paclitaxel and the autophagy inhibitor hydroxychloroquine. The results showed that the muti-functional liposomes can target to the pancreatic tumor and efficiently kill tumor cells and inhibit stroma fibrosis, thus improve the therapeutic effect in orthotopic pancreatic cancer models.


Subject(s)
Autophagy/drug effects , Cancer-Associated Fibroblasts/cytology , Carcinoma, Pancreatic Ductal/metabolism , Liposomes/chemistry , Pancreatic Neoplasms/metabolism , Peptides/chemistry , Animals , Antineoplastic Agents/pharmacology , Cancer-Associated Fibroblasts/metabolism , Disease Progression , Female , Fibrosis , Humans , Hydroxychloroquine/pharmacology , Irinotecan/pharmacology , Mice , Mice, Nude , NIH 3T3 Cells , Nanomedicine , Neoplasm Metastasis , Neoplasm Transplantation , Paclitaxel/pharmacology , Wound Healing
2.
Eur J Pharm Sci ; 124: 240-248, 2018 Nov 01.
Article in English | MEDLINE | ID: mdl-30071282

ABSTRACT

Effective chemotherapy for clinical glioma treatment is still lacking due to the poor penetration of blood-brain barrier (BBB) and the poor internalization into tumor cells. To facilitate the transmigration across the BBB as well as the glioma targeting of chemotherapeutics, we constructed cell penetrating peptide dNP2 and tumor microenvironment-cleavable folic acid (FA) dual modified, paclitaxel (PTX) loaded liposome for the targeted delivery of glioma. The modification of dNP2 significantly enhanced the transmigration across the BBB in an in vitro BBB model. The acid-cleavable cFd-Lip/PTX exhibited sensitive cleavage of FA at pH 6.8, which led to enhanced cellular uptake mediated by both cell penetrating peptide dNP2 and the interaction between FA and folate receptor (FR) on the glioma cells. After intravenous injection, compared with non-cleavable Fd-Lip and single modified liposomes, cFd-Lip enhanced the accumulation in orthotropic glioma and improved the anti-tumor effect of glioma-bearing mice. The dual modified liposomes also facilitated deep penetration into tumor cells and consequently enhanced the cytotoxicity of PTX-loaded liposomes. The acid-cleavable dual modified strategy retained the BBB penetrating and tumor targeting ability, meanwhile, the cleavage of FA further maximized the cell permeability of dNP2, exhibiting enhanced tumor targeting effect. The multi-targeting strategy provides a promising approach towards targeted chemotherapy for glioma.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Brain Neoplasms/drug therapy , Cell-Penetrating Peptides/administration & dosage , Folic Acid/administration & dosage , Glioma/drug therapy , Paclitaxel/administration & dosage , Animals , Antineoplastic Agents, Phytogenic/chemistry , Blood-Brain Barrier/metabolism , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell-Penetrating Peptides/chemistry , Female , Folate Receptors, GPI-Anchored/metabolism , Folic Acid/chemistry , Glioma/metabolism , Hydrogen-Ion Concentration , Liposomes , Mice, Inbred BALB C , Molecular Targeted Therapy , Paclitaxel/chemistry , Phosphatidylethanolamines/administration & dosage , Phosphatidylethanolamines/chemistry , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/chemistry
3.
Nanomedicine ; 14(6): 1833-1843, 2018 08.
Article in English | MEDLINE | ID: mdl-29800759

ABSTRACT

Cancer associated fibroblasts (CAFs) which shape the tumor microenvironment (TME) and the presence of blood brain barrier (BBB) remain great challenges in targeting breast cancer and its brain metastasis. Herein, we reported a strategy using PTX-loaded liposome co-modified with acid-cleavable folic acid (FA) and BBB transmigrating cell penetrating peptide dNP2 peptide (cFd-Lip/PTX) for enhanced delivery to orthotopic breast cancer and its brain metastasis. Compared with single ligand or non-cleavable Fd modified liposomes, cFd-Lip exhibited synergistic TME targeting and BBB transmigration. Moreover, upon arrival at the TME, the acid-cleavable cFd-Lip/PTX showed sensitive cleavage of FA, which reduced the hindrance effect and maximized the function of both FA and dNP2 peptide. Consequently, efficient targeting of folate receptor (FR)-positive tumor cells and FR-negative CAFs was achieved, leading to enhanced anti-tumor activity. This strategy provides a feasible approach to the cascade targeting of TME and BBB transmigration in orthotopic and metastatic cancer treatment.


Subject(s)
Blood-Brain Barrier/drug effects , Brain Neoplasms/drug therapy , Breast Neoplasms/drug therapy , Drug Delivery Systems , Paclitaxel/administration & dosage , Tumor Microenvironment/drug effects , Animals , Apoptosis/drug effects , Brain Neoplasms/metabolism , Brain Neoplasms/secondary , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cell-Penetrating Peptides/chemistry , Female , Folic Acid/chemistry , Humans , Hydrogen-Ion Concentration , Ligands , Liposomes/administration & dosage , Liposomes/chemistry , Mice , Mice, Inbred BALB C , Paclitaxel/chemistry , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Biomaterials ; 168: 1-9, 2018 06.
Article in English | MEDLINE | ID: mdl-29574131

ABSTRACT

In attempts to explore the role of autophagy in breast cancer metastasis, we here report a tumor-activatable particle (named as "D/PSP@CQ/CaP") with the ability of efficient autophagy inhibition. D/PSP@CQ/CaP was prepared by coprecipitating chloroquine phosphate (CQ) with calcium chloride, in the form of chloroquine-calcium phosphate coprecipitate (CQ/CaP), onto the surface of a deep-tumor-penetrating doxorubicine (DOX)-loading core particle (named as "D/PSP"). CQ/CaP could partly disintegrate and release CQ within tumor microenvironment and totally be dissolved within lysosomes. Paxillin is a key component of focal adhesion which functions to anchor tumors cells within the primary tumor for limiting cancer cells' detachment from the primary tumor. We tested that autophagy inhibition caused by CQ released from CQ/CaP could reduce the degradation of paxillin by 2.9 folds in vitro and 2.5 folds in vivo (vs. Control), respectively. Thus metastasis could be influenced by exploiting autophagy-dependent paxillin degradation. Data analysis together proved that D/PSP@CQ/CaP decreased the cancer metastatic extent by 7.5 folds (vs. Control) on mice model via inhibiting the autophagy-dependent disassembly of focal adhesion. At the same time, the growth rate of tumors treated by D/PSP@CQ/CaP was inhibited by 9.1 folds (vs. Control), which could be attributed to its effective tumor drug delivery.


Subject(s)
Autophagy/drug effects , Breast Neoplasms/drug therapy , Drug Carriers , Focal Adhesions/drug effects , Nanoparticles , Animals , Antineoplastic Agents/therapeutic use , Breast Neoplasms/pathology , Cell Adhesion/drug effects , Cell Line, Tumor , Doxorubicin/therapeutic use , Female , Mice , Mice, Inbred BALB C , Neoplasm Metastasis/pathology , Neoplasm Metastasis/prevention & control
5.
Pharm Dev Technol ; 23(1): 13-21, 2018 Jan.
Article in English | MEDLINE | ID: mdl-27884084

ABSTRACT

The dense collagen network in tumors restricts the penetration of drugs into tumors. Free losartan could inhibit collagen, but it would cause hypotension at the dosage of 10 mg/kg/d. In this study, losartan was encapsulated in liposomes (LST-Lip) and the collagen inhibition ability of LST-Lip was investigated. Our results showed the blood pressure was not affected by LST-Lip at the dosage of 2.5 mg/kg every other day. The amount of Evans Blue in tumor in LST-Lip group was 1.98 times of that in control group. Confocal laser scanning microscopy images showed that prior injection of LST-Lip could inhibit collagen and further improve the tumorous accumulation of liposomes modified with TH peptides (AGYLLGHINLHHLAHL(Aib)HHIL-NH2) (TH-Lip) in 4T1 tumors. Compared with control group, the tumor inhibition rate of combined strategy of LST-Lip and paclitaxel loaded TH-Lip (PTX-TH-Lip) was 41.73%, while that of group only treated with PTX-TH-Lip was 14.94%. Masson's trichrome staining confirmed that collagen was inhibited in LST-Lip group. Thus, the administration of LST-Lip in advance could inhibit the collagen in tumors effectively and did not affect the blood pressure, then PTX-TH-Lip injected subsequently could exert enhanced antitumor efficacy. In conclusion, this combined strategy might be promising for breast cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Collagen/antagonists & inhibitors , Liposomes/chemistry , Losartan/pharmacology , Paclitaxel/pharmacology , Animals , Antineoplastic Agents/chemistry , Breast Neoplasms/metabolism , Cell Line, Tumor , Drug Delivery Systems/methods , Female , Hydrogen-Ion Concentration , Losartan/chemistry , Mice , Mice, Inbred BALB C , Paclitaxel/chemistry , Peptides/chemistry
6.
J Drug Target ; 25(2): 179-187, 2017 02.
Article in English | MEDLINE | ID: mdl-27608834

ABSTRACT

Cabazitaxel (CBX) is an effective antineoplastic agent for the treatment of many kinds of cancers. However, the poor water solubility remains a serious deterrent to the utilization of CBX as a commercial drug. In this study, we designed a strategy that integrated CBX into albumin nanoparticles (ANs) formed with human serum albumin (HSA) to improve the water solubility and targeting ability. Meanwhile, we utilized a photothermal agent-indocyanine green (ICG), which could cooperate with CBX to enhance the antitumor effect. The obtained ANs containing ICG and CBX (AN-ICG-CBX) exhibited good mono-dispersity. In vitro cytotoxicity study showed the effectiveness of CBX and ICG, respectively, whereas AN-ICG-CBX with irradiation exhibited the most efficient antiproliferative ability (83.7%). In vivo safety evaluation studies demonstrated the safety of AN-ICG-CBX. Furthermore, the in vivo antitumor study indicated that the AN-ICG-CBX with irradiation achieved higher tumor inhibition rate (91.3%) compared with CBX-encapsulated AN (AN-CBX) (83.3%) or ICG-encapsulated AN (AN-ICG) plus irradiation (60.1%) in 4T1 tumor-bearing mice. To sum up, a safety and effective formulation AN-ICG-CBX was developed in this study and successfully reduced the drug toxicity, improved the targeting efficiency and enhanced the therapeutic effects, becoming a promising candidate for clinical application.


Subject(s)
Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Indocyanine Green/pharmacology , Taxoids/adverse effects , Taxoids/therapeutic use , Animals , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Drug Delivery Systems , Female , Indocyanine Green/administration & dosage , Mammary Neoplasms, Animal , Mice , Mice, Inbred BALB C , Nanoparticles , Neoplasms, Experimental/drug therapy , Random Allocation , Taxoids/administration & dosage
7.
Amino Acids ; 49(1): 75-88, 2017 01.
Article in English | MEDLINE | ID: mdl-27645217

ABSTRACT

Cell-penetrating peptides (CPPs) are widely used in the development of various drug delivery systems because of their ability of penetrating plasma membrane. However, the safety of their application remains largely unknown. In this study, we found that the incubation of two main kinds of CPPs with human normal liver cells could cause the occurrence of apoptosis and necrosis, then the detailed apoptosis-related protein were detected out. To discover the specific way which leads to these results, several methods were used in this study. Several cytokines, such as Caspase3 and Bcl-2, were detected to prove that the damage happened after treated with different CPPs. Then shielding the positive charge of TAT and R8, depletion of Na+ in culturing medium and addition of several inhibitors of specific ATPase site were used to investigate whether the cytotoxicity were charge-dependent and ATPase-related. Furthermore, the membrane potential of mitochondria and the leakage of mitochondrial cytochrome c were detected after treated with CPPs to investigate the damage on mitochondria. In general, our results assess the cytotoxicity caused by two main kinds of CPPs and reveal the clear mechanism of how it occurs. This study reveals the essence of cytotoxicity caused by CPPs, and the methods we followed can be used to evaluate the biocompatibility of new-designed CPPs, which makes the application of CPPs better and safer.


Subject(s)
Cell-Penetrating Peptides/pharmacology , Hepatocytes/drug effects , Mitochondria/drug effects , Oligopeptides/pharmacology , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , tat Gene Products, Human Immunodeficiency Virus/pharmacology , Amino Acid Sequence , Apoptosis/drug effects , Calcium/metabolism , Caspase 3/genetics , Caspase 3/metabolism , Cell Line , Cell Membrane/drug effects , Cell Survival/drug effects , Cell-Penetrating Peptides/chemical synthesis , Cytochromes c/metabolism , Fetus , Gene Expression Regulation , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Necrosis/chemically induced , Necrosis/metabolism , Oligopeptides/chemical synthesis , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/metabolism , tat Gene Products, Human Immunodeficiency Virus/chemical synthesis
8.
Autophagy ; 13(2): 359-370, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27911136

ABSTRACT

To further enhance the intensity of deep tumor drug delivery and integrate a combined therapy, we herein report on a core-shell nanocarrier that could simultaneously overcome the double barriers of the extracellular matrix (ECM) and multiple layers of tumor cells (MLTC). A pH-triggered reversible swelling-shrinking core and an MMP2 (matrix metallopeptidase 2) degradable shell were developed to encapsulate chemotherapeutics and macroautophagy/autophagy inhibitors, respectively. MMP2 degraded the shell, which was followed by the autophagy inhibitors' release. The exposed core could diffuse along the pore within the ECM to deliver chemotherapeutics into deep tumors, and it was able to swell in lysosomes and shrink back in the cytoplasm or ECM. The swelling of the core resulted in the rapid release of chemotherapeutics to kill autophagy-inhibited cells. After leaving the dead cells, the shrinking core could act on neighboring cells that were closer to the center of the tumor. The core thus could also cross MLTC layer by layer to deliver chemotherapeutics into the deep tumor.


Subject(s)
Adenine/analogs & derivatives , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Drug Carriers/chemistry , Drug Delivery Systems , Extracellular Matrix/metabolism , Nanoparticles/chemistry , Adenine/pharmacology , Animals , Cell Line, Tumor , Doxorubicin/pharmacology , Extracellular Matrix/drug effects , Gelatin/chemistry , Humans , MCF-7 Cells , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Particle Size , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Tissue Distribution/drug effects
9.
ACS Appl Mater Interfaces ; 9(3): 2083-2092, 2017 Jan 25.
Article in English | MEDLINE | ID: mdl-28025892

ABSTRACT

The nonselectivity of cell penetrating peptides had greatly limited the application in systemic administration. By conjugating a dGR motif to the C-terminal of octa-arginine, the formed tandem peptide R8-dGR had been proved to specifically recognize both integrin αvß3 and neuropilin-1 receptors. However, the positive charge of poly-arginine would still inevitably lead to rapid clearance in the circulation system. Therefore, in this study, we tried to reduce the positive charge of poly-arginine by decreasing the number of arginine, to thus achieve improved tumor targeting efficiency. We had designed several different Rx-dGR peptides (x = 4, 6, and 8) modified liposomes and investigated their tumor targeting and penetrating properties both in vitro and in vivo. Among all the liposomes, R6-dGR modified liposomes exhibited a long circulation time similar to that of PEGylated liposomes while they retained strong penetrating ability into both tumor cells and tumor tissues, and thus had displayed the most superior tumor targeting efficiency. Then, paclitaxel and indocyanine green coloaded liposomes were prepared, and R6-dGR modified coloaded liposomes also exhibited enhanced antitumor effect on C6 xenograft tumor bearing mice. Therefore, we suggest R6-dGR as a potential tumor targeting ligand with both strong penetrating ability and improved pharmacokinetic behavior, which could be further used for efficient antitumor therapy.


Subject(s)
Peptides/chemistry , Animals , Arginine , Cell Line, Tumor , Cell-Penetrating Peptides , Drug Delivery Systems , Liposomes , Mice , Oligopeptides , Polyethylene Glycols
10.
ACS Nano ; 10(11): 10086-10098, 2016 11 22.
Article in English | MEDLINE | ID: mdl-27934068

ABSTRACT

The treatment of brain tumors remains a challenge due to the limited accumulation of drugs and nanoparticles. Here, we triggered the aggregation of gold nanoparticles (AuNPs) using legumain to enhance the retention of chemotherapeutics in brain tumors. This nanoplatform, AuNPs-A&C, is comprised of Ala-Ala-Asn-Cys-Lys modified AuNPs (AuNPs-AK) and 2-cyano-6-aminobenzothiazole modified AuNPs (AuNPs-CABT). AuNPs-AK could be hydrolyzed to expose the 1,2-thiolamino groups on AuNPs-AK in the presence of legumain, which occurs by a click cycloaddition with the contiguous cyano group on AuNPs-CABT, resulting in formation of AuNPs aggregates. This strategy led to an enhanced retention of the AuNPs in glioma cells both in vitro and in vivo due to the blocking of nanoparticle exocytosis and minimizing nanoparticle backflow to the bloodstream. After conjugation of doxorubicin (DOX) via a pH-sensitive linker to AuNPs-A&C, the efficiency for treating glioma was improved. The median survival time for the DOX-linked AuNPs-A&C increased to 288% in comparison to the saline group. We further show the use of the AuNPs-A&C for optical imaging applications. In conclusion, we provide a strategy to increase nanoparticle tumor accumulation with the potential to improve therapeutic outcome.


Subject(s)
Brain Neoplasms/drug therapy , Gold , Metal Nanoparticles , Antimetabolites, Antineoplastic/pharmacokinetics , Cell Line, Tumor , Doxorubicin/pharmacokinetics , Humans
11.
Theranostics ; 6(12): 2141-2160, 2016.
Article in English | MEDLINE | ID: mdl-27698946

ABSTRACT

The enhanced permeability and retention (EPR) effect has been comfortably accepted, and extensively assumed as a keystone in the research on tumor-targeted drug delivery system. Due to the unsatisfied tumor-targeting efficiency of EPR effect being one conspicuous drawback, nanocarriers that merely relying on EPR effect are difficult to access the tumor tissue and consequently trigger efficient tumor therapy in clinic. In the present contribution, we break up the shackles of EPR effect on nanocarriers thanks to their universal distribution characteristic. We successfully design a paclitaxel (PTX) and alpha-galactosylceramide (αGC) co-loaded TH peptide (AGYLLGHINLHHLAHL(Aib)HHIL-Cys) -modified liposome (PTX/αGC-TH-Lip) and introduce a new concept of immuno-chemotherapy combination via accumulation of these liposomes at both spleen and tumor sites naturally and simultaneously. The PTX-initiated cytotoxicity attacks tumor cells at tumor sites, meanwhile, the αGC-triggered antitumor immune response emerges at spleen tissue. Different to the case that liposomes are loaded with sole drug, in this concept two therapeutic processes effectively reinforce each other, thereby elevating the tumor therapy efficiency significantly. The data demonstrates that the PTX/αGC-TH-Lip not only possess therapeutic effect against highly malignant B16F10 melanoma tumor, but also adjust the in vivo immune status and induce a more remarkable systemic antitumor immunity that could further suppress the growth of tumor at distant site. This work exhibits the capability of the PTX/αGC-TH-Lip in improving immune-chemotherapy against tumor after systemic administration.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Drug Carriers/pharmacokinetics , Immunotherapy/methods , Liposomes/pharmacokinetics , Melanoma/drug therapy , Nanostructures/chemistry , Paclitaxel/pharmacokinetics , Animals , Antineoplastic Agents/administration & dosage , Disease Models, Animal , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Galactosylceramides/metabolism , Liposomes/administration & dosage , Liposomes/chemistry , Melanoma/immunology , Mice, Inbred C57BL , Paclitaxel/administration & dosage , Treatment Outcome
12.
Sci Rep ; 6: 27669, 2016 06 08.
Article in English | MEDLINE | ID: mdl-27273770

ABSTRACT

In recent years, active targeting strategies by ligand modification have emerged to enhance tumor accumulation of NP, but their clinical application was strictly restricted due to the complex preparation procedures, poor stability and serious toxicity. An effective and clinical translational strategy is required to satisfy the current problems. Interestingly, the internalization of NP is intimately related with cell cycle and the expression of receptors is not only related with cancer types but also cell cycle progression. So the cellular uptake of ligand modified NP may be related with cell cycle. However, few investigations were reported about the relationship between cell cycle and the internalization of ligand modified NP. Herein, cellular uptake of folic acid (FA) modified NP after utilizing chemotherapeutic to retain the tumor cells in G2/M phase was studied and a novel strategy was designed to enhance the active targeting effect. In our study, docetaxel (DTX) notably synchronized cells in G2/M phase and pretreatment with DTX highly improved in vitro and in vivo tumor cell targeting effect of FA decorated NP (FANP). Since FA was a most common used tumor active targeting ligand, we believe that this strategy possesses broader prospects in clinical application for its simplicity and effectiveness.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , G2 Phase Cell Cycle Checkpoints/drug effects , Nanoparticles , Neoplasms/metabolism , Animals , Cell Cycle/drug effects , Cell Line, Tumor , Disease Models, Animal , Docetaxel , Folic Acid , Humans , Ligands , Male , Mice , Molecular Imaging , Molecular Targeted Therapy , Nanoparticles/chemistry , Nanoparticles/ultrastructure , Polyethylene Glycols , Polymers , Taxoids/administration & dosage , Taxoids/pharmacokinetics , Xenograft Model Antitumor Assays
13.
Autophagy ; 12(6): 949-62, 2016 06 02.
Article in English | MEDLINE | ID: mdl-27123811

ABSTRACT

Hydroxychloroquine (HCQ) inhibits autophagy and therefore can sensitize some cancer cells to chemotherapy, but the high doses required limit its clinical use. Here we show that loading HCQ into liposomes (HCQ/Lip) decorated with a pH-sensitive TH-RGD targeting peptide (HCQ/Lip-TR) can concentrate HCQ in B16F10 tumor cells and lysosomes. HCQ/Lip-TR was efficiently internalized as a result of its ability to bind ITGAV-ITGB3/integrin αvß3 receptors highly expressed on the tumor cell surface and to undergo charge reversal from anionic at pH 7.4 to cationic at pH 6.5. Studies in vitro at pH 6.5 showed that the intracellular HCQ concentration was 35.68-fold higher, and lysosomal HCQ concentration 32.22-fold higher, after treating cultures with HCQ/Lip-TR than after treating them with free HCQ. The corresponding enhancements observed in mice bearing B16F10 tumors were 15.16-fold within tumor cells and 14.10-fold within lysosomes. HCQ/Lip-TR was associated with milder anemia and milder myosuppressive reductions in white blood cell and platelet counts than free HCQ, as well as less accumulation in the small intestine, which may reduce risk of intestinal side effects. In addition, co-delivering HCQ/Lip-TR with either free doxorubicin (DOX) or liposomal DOX improved the ability of DOX to inhibit tumor growth. Biochemical, electron microscopy and immunofluorescence experiments confirmed that HCQ/Lip-TR blocked autophagic flux in tumor cells. Our results suggest that loading HCQ into Lip-TR liposomes may increase the effective concentration of the inhibitor in tumor cells, allowing less toxic doses to be used.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Doxorubicin/analogs & derivatives , Drug Delivery Systems , Hydroxychloroquine/administration & dosage , Hydroxychloroquine/pharmacology , Lysosomes/metabolism , Animals , Biophysical Phenomena , Doxorubicin/pharmacology , Female , Humans , Hydrogen-Ion Concentration , Lysosomes/drug effects , MCF-7 Cells , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Models, Biological , Peptides/pharmacology , Polyethylene Glycols/pharmacology
14.
Sci Rep ; 6: 19800, 2016 Feb 04.
Article in English | MEDLINE | ID: mdl-26842655

ABSTRACT

[D]-H6L9, as a pH-responsive anti-microbial peptide (AMP), has been evidenced by us to be an excellent choice in tumor microenvironment-responsive delivery as it could render liposomes responsive to the acidified tumor microenvironment. However, [D]-H6L9-modified liposomes could not actively target to tumor area. Therefore, integrin αvß3-targeted peptide RGD was co-modified with [D]-H6L9 onto liposomes [(R + D)-Lip] for improved tumor delivery efficiency. Under pH 6.3, (R + D)-Lip could be taken up by C26 cells and C26 tumor spheroids (integrin αvß3-positive) with significantly improved efficiency compared with other groups, which was contributed by both RGD and [D]-H6L9, while RGD did not increase the cellular uptake performance on MCF-7 cells (integrin αvß3-negative). Results showed that RGD could decrease cellular uptake of (R + D)-Lip while [D]-H6L9 could increase it, implying the role of both RGD and [D]-H6L9 in cellular internalization of (R + D)-Lip. On the other hand, (R + D)-Lip could escape the entrapment of lysosomes. PTX-loaded (R + D)-Lip could further increase the cellular toxicity against C26 cells compared with liposomes modified only with RGD and [D]-H6L9 respectively, and achieve remarkable tumor inhibition effect on C26 tumor models.


Subject(s)
Antimicrobial Cationic Peptides/chemistry , Liposomes/chemistry , Oligopeptides/chemistry , Animals , Antimicrobial Cationic Peptides/metabolism , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/toxicity , Cell Line, Tumor , Cell Survival/drug effects , Humans , Hydrogen-Ion Concentration , Integrin alphaVbeta3/metabolism , Liposomes/metabolism , MCF-7 Cells , Mice , Mice, Inbred BALB C , Microscopy, Confocal , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Oligopeptides/metabolism , Paclitaxel/administration & dosage , Paclitaxel/chemistry , Paclitaxel/toxicity , Particle Size , Polyethylene Glycols/chemistry , Tissue Distribution , Transplantation, Heterologous , Tumor Microenvironment
15.
Theranostics ; 6(2): 177-91, 2016.
Article in English | MEDLINE | ID: mdl-26877777

ABSTRACT

Cell penetrating peptides (CPPs) were widely used for drug delivery to tumor. However, the nonselective in vivo penetration greatly limited the application of CPPs-mediated drug delivery systems. And the treatment of malignant tumors is usually followed by poor prognosis and relapse due to the existence of extravascular core regions of tumor. Thus it is important to endue selective targeting and stronger intratumoral diffusion abilities to CPPs. In this study, an RGD reverse sequence dGR was conjugated to a CPP octa-arginine to form a CendR (R/KXXR/K) motif contained tandem peptide R8-dGR (RRRRRRRRdGR) which could bind to both integrin αvß3 and neuropilin-1 receptors. The dual receptor recognizing peptide R8-dGR displayed increased cellular uptake and efficient penetration ability into glioma spheroids in vitro. The following in vivo studies indicated the active targeting and intratumoral diffusion capabilities of R8-dGR modified liposomes. When paclitaxel was loaded in the liposomes, PTX-R8-dGR-Lip induced the strongest anti-proliferation effect on both tumor cells and cancer stem cells, and inhibited the formation of vasculogenic mimicry channels in vitro. Finally, the R8-dGR liposomal drug delivery system prolonged the medium survival time of intracranial C6 bearing mice by 2.1-fold compared to the untreated group, and achieved an exhaustive anti-glioma therapy including anti-tumor cells, anti-vasculogenic mimicry and anti-brain cancer stem cells. To sum up, all the results demonstrated that R8-dGR was an ideal dual receptor recognizing CPP with selective glioma targeting and efficient intratumoral diffusion, which could be further used to equip drug delivery system for effective glioma therapy.


Subject(s)
Brain Neoplasms/drug therapy , Cell-Penetrating Peptides/pharmacokinetics , Glioma/drug therapy , Integrin alphaVbeta3/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Cell-Penetrating Peptides/adverse effects , Cell-Penetrating Peptides/chemistry , HeLa Cells , Humans , Liposomes/adverse effects , Liposomes/pharmacokinetics , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Oligopeptides/adverse effects , Oligopeptides/physiology , Protein Binding
16.
ACS Appl Mater Interfaces ; 7(38): 21442-54, 2015 Sep 30.
Article in English | MEDLINE | ID: mdl-26371468

ABSTRACT

Glioma, one of the most common aggressive malignancies, has the highest mortality in the present world. Delivery of nanocarriers from the systemic circulation to the glioma sites would encounter multiple physiological and biological barriers, such as blood-brain barrier (BBB) and the poor penetration of nanocarriers into the tumor. To circumvent these hurdles, the paclitaxel-loaded liposomes were developed by conjugating with a TR peptide (PTX-TR-Lip), integrin αvß3-specific vector with pH-responsible cell-penetrating property, for transporting drug across the BBB and then delivering into glioma. Surface plasmon resonance (SPR) studies confirmed the very high affinity of TR-Lip and integrin αvß3. In vitro results showed that TR-Lip exhibited strong transport ability across BBB, killed glioma cells and brain cancer stem cells (CSCs), and destroyed the vasculogenic mimicry (VM) channels. In vivo results demonstrated that TR-Lip could better target glioma, and eliminated brain CSCs and the VM channels in tumor tissues. The median survival time of tumor-bearing mice after administering PTX-TR-Lip (45 days) was significantly longer than that after giving free PTX (25.5 days, p < 0.001) or other controls. In conclusion, PTX-TR-Lip would improve the therapeutic efficacy of brain glioma in vitro and in vivo.


Subject(s)
Blood-Brain Barrier/metabolism , Glioma/drug therapy , Integrin alphaVbeta3/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biological Transport/drug effects , Blood-Brain Barrier/drug effects , Brain Neoplasms/pathology , Cell Death/drug effects , Cell Line , Cell Survival/drug effects , Diagnostic Imaging , Electric Impedance , Endocytosis/drug effects , Epithelium/drug effects , Female , Glioma/pathology , Hydrogen-Ion Concentration , Liposomes , Mice, Inbred BALB C , Neoplastic Stem Cells/drug effects , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Particle Size , Peptides/chemistry , Proton Magnetic Resonance Spectroscopy , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Static Electricity , Surface Plasmon Resonance
17.
J Control Release ; 217: 138-50, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26368312

ABSTRACT

The use of pH-responsive cell-penetrating peptides (CPPs) is an attractive strategy for drug delivery in vivo, however, they still could not actively target to the desired sites. Here, we designed a pH-responsive CPP (TR) with the ability of active targeting to integrin αvß3, which was a tandem peptide consisted of active targeting ligand peptide (c(RGDfK)) and pH-responsive CPP (TH). The targeting efficiency of TR with integrin was evaluated by molecular simulation and docking studies. The affinity assays of TR peptide modified liposomes (TR-Lip) at pH7.4 and pH6.5 demonstrated adequately the pH-responsive binding efficacy of TR-Lip with integrin αvß3. The cellular uptake of CFPE-labeled TR-Lip on integrin αvß3-overexpressing B16F10 cells was 41.67-, 30.67-, and 11.90-fold higher than that of CFPE-labeled PEG-, RGD-, and TH-modified liposomes at pH6.5, respectively, suggesting that TR-Lip could not only actively target to αvß3-overexpressing cells compared to TH-Lip, but also significantly increased cellular uptake compared to RGD-Lip. At the concentration of 20µg/mL paclitaxel (PTX), the killing activity of PTX-loaded TR-Lip (PTX-TR-Lip) against B16F10 cells was 1.80-, 1.45-, 1.30-, 1.15-time higher than that of PTX-loaded PEG-, RGD-, TH-modified liposomes and free PTX at pH6.5, respectively. In vivo imaging displayed the maximum accumulation of DiD-labeled TR-Lip at tumor sites compared to the other groups. Tumor inhibition rate of B16F10 tumor-bearing mice treated with PTX-TR-Lip was 85.04%, relative to that of PBS. In B16F10 tumor-bearing mice, PTX-TR-Lip showed significantly higher survival rate compared with the other groups. Collectively, all the results in vitro and in vivo suggested that TR-Lip would be a potential delivery system for PTX to treat integrin αvß3-overexpressing tumor-bearing mice.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Cell-Penetrating Peptides/administration & dosage , Integrin alphaVbeta3/metabolism , Melanoma, Experimental/drug therapy , Paclitaxel/administration & dosage , Animals , Antineoplastic Agents, Phytogenic/blood , Antineoplastic Agents, Phytogenic/pharmacokinetics , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/therapeutic use , Coculture Techniques , Drug Liberation , Female , Humans , Hydrogen-Ion Concentration , Liposomes , Melanoma, Experimental/metabolism , Mice, Inbred C57BL , Paclitaxel/blood , Paclitaxel/pharmacokinetics , Phosphatidylethanolamines/chemistry , Polyethylene Glycols/chemistry
18.
J Biomed Nanotechnol ; 11(3): 382-91, 2015 Mar.
Article in English | MEDLINE | ID: mdl-26307822

ABSTRACT

Hybrid nanoparticles consisting of lipids and the biodegradable polymer, poly (D,L-lactide-co-glycolide) (PLGA), were developed for the targeted delivery of the anticancer drug, docetaxel. Transmission electron microscopic observations confirmed the presence of a lipid coating over the polymeric core. Using coumarin-6 as a fluorescent probe, the uptake efficacy of RGD conjugated lipid coated nanoparticles (RGD-L-P) by C6 cells was increased significantly, compared with that of lipid-polymer hybrid nanoparticles (L-P; 2.5-fold higher) or PLGA-nanoparticles (PLGA-P; 1.76-fold higher). The superior tumor spheroid penetration of RGD-L-P indicated that RGD-L-P could target effectively and specifically to C6 cells overexpressing integrin α(v)ß3. The anti-proliferative activity of docetaxel-loaded RGD-L-P against C6 cells was increased 2.69- and 4.13-fold compared with L-P and PLGA-P, respectively. Regarding biodistribution, the strongest brain-localized fluorescence signals were detected in glioblastoma multiforme (GBM)-bearing rats treated with 1,10-Dioctadecyl-3,3,30,30-tetramethylindotricarb-ocyanine iodide (DiR)-loaded RGD-L-P, compared to rats treated with DiR-loaded L-P or PLGA-P. The median survival time of GBM-bearing rats treated with docetaxel-loaded RGD-L-P was 57 days, a fold increase of 1.43, 1.78, 3.35, and 3.56 compared with animals given L-P (P < 0.05), PLGA-P (P < 0.05), Taxotere (P < 0.01) and saline (P < 0.01), respectively. Collectively, these results support RGD-L-P as a promising drug delivery system for the specific targeting and the treatment of GBM.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Glioblastoma/drug therapy , Glioblastoma/metabolism , Nanocapsules/chemistry , Taxoids/administration & dosage , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Diffusion , Docetaxel , Glioblastoma/pathology , Lipids/chemistry , Nanocapsules/ultrastructure , Nanocomposites/chemistry , Nanocomposites/ultrastructure , Oligopeptides/chemistry , Oligopeptides/pharmacokinetics , Polymers/chemistry , Rats , Rats, Sprague-Dawley , Survival Rate , Taxoids/chemistry , Treatment Outcome
19.
ACS Appl Mater Interfaces ; 7(30): 16792-801, 2015 Aug 05.
Article in English | MEDLINE | ID: mdl-26173814

ABSTRACT

The chemotherapy of aggressive glioma is usually accompanied by a poor prognosis because of the formation of vasculogenic mimicry (VM) and brain cancer stem cells (BCSCs). VM provided a transporting pathway for nutrients and blood to the extravascular regions of the tumor, and BCSCs were always related to drug resistance and the relapse of glioma. Thus, it is important to evaluate the inhibition effect of antiglioma drug delivery systems on both VM and BCSCs. In this study, paclitaxel-loaded liposomes modified with a multifunctional tandem peptide R8-c(RGD) (R8-c(RGD)-Lip) were used for the treatment of glioma. An in vitro cellular uptake study proved the strongest targeting ability to be that of R8-c(RGD)-Lip to glioma stem cells. Drug loaded R8-c(RGD)-Lip exhibited an efficient antiproliferation effect on BCSCs and could induce the destruction of VM channels in vitro. The following pharmacodynamics study demonstrated that R8-c(RGD)-modified drug-loaded liposomes achieved both anti-VM and anti-BCSC effects in vivo. Finally, no significant cytotoxicity of the blood system or major organs of the drug-loaded liposomes was observed under treatment dosage in the safety evaluation. In conclusion, all of the results proved that R8-c(RGD)-Lip was a safe and efficient antiglioma drug delivery system.


Subject(s)
Brain Neoplasms/drug therapy , Liposomes/chemistry , Neoplastic Stem Cells/drug effects , Neovascularization, Pathologic/drug therapy , Oligopeptides/pharmacokinetics , Paclitaxel/administration & dosage , Angiogenesis Inhibitors/administration & dosage , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/chemistry , Apoptosis/drug effects , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Glioma , Mice, Inbred BALB C , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Oligopeptides/chemistry , Paclitaxel/chemistry , Treatment Outcome
20.
J Pharm Sci ; 104(3): 1160-73, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25449709

ABSTRACT

To combine the advantage of poly(ethylene gylcol) (PEG) for longer circulation and cell-penetrating peptides (CPPs) for efficient cellular uptake, paclitaxel (PTX)-loaded liposomes functionalized with TAT, the most frequently used CPP, and cleavable PEG via a redox-responsive disulfide linker (PTX-C-TAT-LP) were successfully developed here. Under physiological conditions, TAT was shielded by PEG layer and liposomes exhibited a long blood circulation. At tumor site, PEG could be detached in the presence of exogenous reducing agent [glutathione (GSH)] and TAT was exposed to facilitate cell internalization. In the presence of GSH, the liposomal vesicle C-TAT-LP showed increased cellular uptake and improved three-dimensional tumor spheroids penetration in vitro compared with analogous stable shielded liposomes. C-TAT-LP achieved enhanced tumor distribution and demonstrated superior delivery efficiency in vivo. PTX-C-TAT-LP with GSH strongly inhibited the proliferation of murine melanoma B16F1 tumor cells in vitro and in vivo with the tumor inhibition rate being 69.4% on B16F1-bearing mice. In addition, the serum aspartate transaminase level, alanine transaminase level, and creatine kinase level were almost completely within normal range in the PTX-C-TAT-LP with GSH group, revealing PTX-C-TAT-LP with GSH had no obvious drug-related adverse events for liver and heart. Taken together, C-TAT-LP is a promising tumor-targeting drug carrier.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Cell-Penetrating Peptides/metabolism , Lipids/chemistry , Melanoma, Experimental/drug therapy , Paclitaxel/administration & dosage , Polyethylene Glycols/chemistry , tat Gene Products, Human Immunodeficiency Virus/metabolism , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/metabolism , Cell Line, Tumor , Cell Membrane Permeability , Cell Proliferation/drug effects , Cell-Penetrating Peptides/chemistry , Chemistry, Pharmaceutical , Dose-Response Relationship, Drug , Glutathione/metabolism , Liposomes , Male , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Nanotechnology , Oxidation-Reduction , Paclitaxel/chemistry , Paclitaxel/metabolism , Technology, Pharmaceutical/methods , tat Gene Products, Human Immunodeficiency Virus/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...