Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Theranostics ; 11(16): 7896-7910, 2021.
Article in English | MEDLINE | ID: mdl-34335971

ABSTRACT

Rationale: Small-molecule prodrug nanoassembly is emerging as an efficient platform for chemotherapy. The self-assembly stability plays a vital role on the drug delivery efficiency of prodrug nanoassembly. It is reported that fluoroalkylation could improve the self-assembly stability of amphiphilic polymers by utilizing the unique fluorination effect. But the application of fluoroalkylation on small-molecule prodrug nanoassembly has never been reported. Methods: Here, fluoro-modified prodrug was developed by conjugating paclitaxel with perfluorooctanol (F8-SS-PTX), and the paclitaxel-octanol prodrug (C8-SS-PTX) was used as control. The fluoro-mediated self-assembly mechanisms were illustrated using molecular dynamics simulation. In addition, the impacts of fluoroalkylation on the pharmacy characters, in vivo fate and antitumor effect of small-molecule prodrug nanoassembly were investigated in details. Results: Fluoroalkylation significantly improved the self-assembly stability of F8-SS-PTX NPs both in vitro and in vivo, which could be attributed to the fluoro-mediated hydrophobic force and halogen bonds. The AUC0-24h and tumor accumulation of F8-SS-PTX NPs was 6-fold and 2-fold higher than that of C8-SS-PTX NPs, respectively. As a result, F8-SS-PTX NPs exhibited much better antitumor effect than C8-SS-PTX NPs and Abraxane. Conclusion: Fluoroalkylation could improve the self-assembly stability, in vivo fate, and antitumor efficacy of small-molecule prodrug nanoassemblies, which could be an effective strategy for the rational design of advanced nanomedicines.


Subject(s)
Fluorides/chemistry , Prodrugs/chemistry , Theranostic Nanomedicine/methods , Animals , Cell Line, Tumor , Drug Delivery Systems/methods , Drug Liberation/physiology , Fluoridation/methods , Humans , Mice , Molecular Dynamics Simulation , Nanomedicine/methods , Nanoparticles/chemistry , Paclitaxel/therapeutic use , Polyethylene Glycols/chemistry , Polymers/chemistry , Prodrugs/pharmacology
2.
Int J Pharm ; 606: 120923, 2021 Sep 05.
Article in English | MEDLINE | ID: mdl-34303822

ABSTRACT

A surge of platinum(IV) compounds are utilized or investigated in cancer treatment but their therapeutic outcomes have been greatly compromised by remaining adverse effects and limited antitumor performance, attributable to nonspecific distribution and insufficient activation in tumor site. Herein, we designed a series of disulfide bond introduced Pt(IV)-lipid prodrugs with different branch length, all of which are able to self-stabilize into nanomedicine and be activated by high intracellular glutathione (GSH) level. The impact of precise modification of these prodrugs on their assembly stability, pharmacokinetics and cytotoxicity was probed to establish a connection between chemical structure and antiproliferation efficiency. With optimal assembly manner and delivery efficacy, the longest axial branched Pt(IV) prodrug CSS18 exhibited the most impressive therapeutic outcome, providing a potential path to more efficient nanocarriers for chemotherapeutic agents by chemical modulation and, giving insights into the rational design of reduction responsive platinum delivery system.


Subject(s)
Antineoplastic Agents , Prodrugs , Cell Line, Tumor , Nanomedicine , Platinum
3.
Colloids Surf B Biointerfaces ; 203: 111766, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33866279

ABSTRACT

The platinum-based drugs prevail in the therapy of malignant tumors treatment. However, their clinical outcomes have been heavily restricted by severe systemic toxicities. To ensure biosafety and efficiency, herein, we constructed a disulfide bond inserted Pt(IV) self-assembled nanoplatform that is selectively activated by rich glutathione (GSH) in tumor site. Disulfide bond was introduced into the conjugates of oxaliplatin (IV) and oleic acid (OA) which conferred cascade reduction-responsiveness to nanoassemblies. Disulfide bond cleavage and reduction of Pt(IV) center occur sequentially as a cascade process. In comparison to oxaliplatin solution, Pt(IV) nanoparticles (NPs) achieved prolonged blood circulation and higher maximum tolerated doses. Furthermore, Oxa(IV)-SS-OA prodrug NPs exhibited potent anti-tumor efficiency against 4T1 cells and low toxicities in other normal tissues, which offers a promising nano-platform for potential clinical application.


Subject(s)
Antineoplastic Agents , Nanoparticles , Neoplasms , Prodrugs , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Containment of Biohazards , Disulfides , Humans , Neoplasms/drug therapy , Prodrugs/therapeutic use
4.
Drug Deliv Transl Res ; 11(5): 2186-2197, 2021 10.
Article in English | MEDLINE | ID: mdl-33452654

ABSTRACT

Onivyde is the first irinotecan (IRI) nanoliposome that could improve pharmacokinetics and tumor biodistribution of irinotecan. Although FDA approves Onivyde for the treatment of pancreatic cancer patients who are not effective for GEM, the gastrointestinal toxicity caused by Onivyde is still a problem to be solved in clinical application. Berberine (BER), an isoquinolone alkaloid extracted from several different plants, has been reported to exhibit beneficial effect in alleviating intestinal mucositis and generating synergistic anticancer effect in combination with cytotoxic drugs. However, its therapeutic effect is affected by the different pharmacokinetic behavior of two drugs. Therefore, we utilized triethylamine-sucrose octasulfate gradient to construct nanoliposomes for co-delivery of irinotecan and berberine, termed as lipBI. This co-delivery nanoliposomes remained the synergistic ratio in the body and improved tumor distribution of IRI and BER. The lipBI significantly inhibited tumor growth in the BXPC-3 pancreatic cancer model compared with Onivyde (p < 0.05) and reduced the gastrointestinal toxicity in mice caused by IRI. Overall, IRI/BER co-loaded liposomes possessed great potential in the treatment of pancreatic cancer.


Subject(s)
Berberine , Pancreatic Neoplasms , Animals , Humans , Irinotecan/pharmacokinetics , Liposomes/therapeutic use , Mice , Pancreatic Neoplasms/drug therapy , Tissue Distribution
5.
Acta Pharm Sin B ; 10(9): 1730-1740, 2020 Sep.
Article in English | MEDLINE | ID: mdl-33088692

ABSTRACT

The combination of paclitaxel (PTX) and doxorubicin (DOX) has been widely used in the clinic. However, it remains unsatisfied due to the generation of severe toxicity. Previously, we have successfully synthesized a prodrug PTX-S-DOX (PSD). The prodrug displayed comparable in vitro cytotoxicity compared with the mixture of free PTX and DOX. Thus, we speculated that it could be promising to improve the anti-cancer effect and reduce adverse effects by improving the pharmacokinetics behavior of PSD and enhancing tumor accumulation. Due to the fact that copper ions (Cu2+) could coordinate with the anthracene nucleus of DOX, we speculate that the prodrug PSD could be actively loaded into liposomes by Cu2+ gradient. Hence, we designed a remote loading liposomal formulation of PSD (PSD LPs) for combination chemotherapy. The prepared PSD LPs displayed extended blood circulation, improved tumor accumulation, and more significant anti-tumor efficacy compared with PSD NPs. Furthermore, PSD LPs exhibited reduced cardiotoxicity and kidney damage compared with the physical mixture of Taxol and Doxil, indicating better safety. Therefore, this novel nano-platform provides a strategy to deliver doxorubicin with other poorly soluble antineoplastic drugs for combination therapy with high efficacy and low toxicity.

6.
Int J Pharm ; 589: 119814, 2020 Nov 15.
Article in English | MEDLINE | ID: mdl-32877728

ABSTRACT

The value of the clinical application of chemotherapeutic drugs is dependent on both systemic toxicity and treatment efficacy. Dose intensification and high tolerability suggest the potential for clinical cancer therapy. In this study, we developed a novel strategy for reconstructing a drug molecule into remote-loading liposomes. Two weak-base cabazitaxel derivatives were synthesized, and named CN and CN2. CN exhibited higher cytotoxic effects compared to CN2, and was selected for further study. CN was remotely loaded into nano-size liposomes (CN-LPs) via an ammonium sulfate gradient with high loading and encapsulation efficiency. When compared to the commercial formulation of cabazitaxel, JEVTANA®, CN-LPs showed less systemic toxicity and enhanced tolerability, with at least a 24-fold increase in the tolerated dose. Furthermore, CN-LPs significantly inhibited tumor growth in mice bearing 4T1 and RM-1 xenograft tumors. After intravenous injection, CN-LPs exhibited an extremely high drug concentration in blood, with a 757-fold increase in the area under the curve (AUC). Moreover, 48 h after a single intravenous injection, CN-LPs promoted higher drug accumulation in tumors compared to JEVTANA®. In summary, our liposome delivery system exhibits favorable pharmacologic efficacy and an improved safety profile.


Subject(s)
Antineoplastic Agents , Liposomes , Animals , Cell Line, Tumor , Mice , Taxoids
7.
Int J Pharm ; 587: 119680, 2020 Sep 25.
Article in English | MEDLINE | ID: mdl-32712250

ABSTRACT

Along with the malignant proliferation of tumor requiring nutrients, the expression of L-type amino acid transporter 1(LAT1) and amino acid transporter B0,+ (ATB0,+) in cancer cells is up-regulated that can be used as new targets for active targeting of tumor. However, since normal cells also express amino acid transporters in small amounts, traditional ligand-exposure drug delivery systems are potentially toxic to the body. Therefore, we designed a smart-response drug delivery system that buries the tyrosine ligand in PEG hydration layer at normal tissues and exposes the ligand by cleaving the pH-sensitive bond of PEG at the tumor site. Irinotecan (CPT-11) is actively loaded into the inner aqueous phase of liposomes via a copper ion gradient mechanism which has high encapsulation efficiency and stable drug release profile. Smart-response liposomes showed the strongest cytotoxicity and the maximum cellular uptake in vitro, the largest amount of tumor site accumulation and the best antitumor effect in vivo, compared with non-targeted liposomes and non-sensitive liposomes. It is worth noting that smart-response liposomes not only achieved enhanced antitumor effect but also attenuated side effects compared to ligand-exposure liposomes. This provides a smart responsive drug delivery system for precise treatment and shows a good application prospect.


Subject(s)
Liposomes , Neoplasms , Cell Line, Tumor , Drug Delivery Systems , Humans , Hydrogen-Ion Concentration , Irinotecan , Ligands , Neoplasms/drug therapy
8.
RSC Adv ; 10(46): 27676-27687, 2020 Jul 21.
Article in English | MEDLINE | ID: mdl-35516912

ABSTRACT

Liposomes are among the most successful nanocarriers; several products have been marketed, all of which were prepared by active loading methods. However, poorly water-soluble drugs without ionizable groups are usually incorporated into the lipid bi-layer of liposomes by passive loading methods, with serious drug leakage during blood circulation. Furthermore, there have been few improvements in their anti-cancer activity and safety. Herein, we designed and synthesized three weak-acid modified paclitaxel (PTX) derivatives with a one-step reaction for the remote loading of liposomal formulations. By comparison, PTX-succinic acid liposomes (PTX-SA LPs) exhibited the highest encapsulation efficiency (97.2 ± 1.8%) and drug loading (8.84 ± 0.16%); meanwhile, there was almost no change in their particle size or zeta potential within one month. Furthermore, compared with Taxol®, the PTX-SA LPs showed a 4.35-fold prolonged half-time, enhanced tumor accumulation, and an increased maximum tolerated dose (MTD) of more than 30 mg kg-1. As a result, the PTX-SA LPs displayed significantly improved in vivo anti-cancer efficacy in comparison with Taxol®. Therefore, weak-acid modification is proved to be a simple and effective method to achieve remote loading and high encapsulation efficiency of poorly soluble drugs, showing great potential for clinical application.

9.
J Control Release ; 316: 22-33, 2019 12 28.
Article in English | MEDLINE | ID: mdl-31676386

ABSTRACT

As the demand for nutrients in malignant proliferation of tumors increases, the L-type amino acid transporter 1(LAT1) and amino acid transporter B0,+ (ATB0,+) of tumor cells are more highly expressed than normal cells which can be used as new targets for active targeting of cancer. However, drug delivery systems often require multi-target design to achieve simultaneous targeting of different receptors or transporters due to the heterogeneity of the tumor. Here we utilized triethylamine-sucrose octasulfate gradient to actively encapsulate irinotecan into the introliposomal aqueous phase. Targeted ability was achieved through inserting different amino acids modified polyethylene glycol monostearate into the liposomes, and found that glutamate-liposomes can be targeted to LAT1, lysine-liposomes can be targeted to ATB0,+, and inspiringly, tyrosine-liposomes can be simultaneously targeted to LAT1 and ATB0,+. The tyrosine-modified liposomes showed the highest cellular uptake in BxPC-3 and MCF-7 cells which were highly expressed both LAT1 and ATB0,+. Moreover, we validated their targeting capabilities and elucidated the transport mechanism of LAT1 and ATB0,+-mediated endocytosis. The tumor inhibition rate of tyrosine-modified liposomes greatly increased from 39% to 87% compared with commercially available liposomes loaded CPT-11(Onivyde®). Overall, it showed a good application prospect for efficient tumor therapy and industrial production.


Subject(s)
Antineoplastic Agents/administration & dosage , Irinotecan/administration & dosage , Neoplasms/drug therapy , Tyrosine/chemistry , Amino Acid Transport Systems/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Delivery Systems , Humans , Irinotecan/pharmacology , Large Neutral Amino Acid-Transporter 1/metabolism , Liposomes , MCF-7 Cells , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/pathology
10.
Int J Pharm ; 557: 264-272, 2019 Feb 25.
Article in English | MEDLINE | ID: mdl-30599233

ABSTRACT

The combination regimen of irinotecan (IRI) and doxorubicin (DOX) for cancer treatment has been frequently exploited in clinical studies, but face challenges in design of efficacious combination drug delivery systems. Here we demonstrate a novel nanoliposome constructed by triethylammonium sucrose octasulfate gradient loading method for co-delivering the two therapeutic agents. In vitro cytotoxicity of IRI, DOX and their combinations against breast cancer cells (4T-1), non-small cell lung cancer cells (A549) and colon cancer cells (HT-29) was evaluated to screen optimal synergistic ratio of the two drugs. The co-delivery nanocarrier maintained the synergistic ratio in vivo, and increased tumor distribution of both drugs (≈2.18-fold vs single drug-loaded formulations). IRI/DOX co-loaded liposomes, with exceedingly high drug-to-phospholipid ratio of 0.61: 1 (molar ratio), exhibit potent antitumor efficacy in the 4T-1 mammary carcinoma xenograft, compared to the mixture of single drug-loaded liposomes (P < 0.001). This co-encapsulated and co-delivered nanoliposome technology offers a promising strategy for cancer treatment.


Subject(s)
Antineoplastic Agents/administration & dosage , Doxorubicin/administration & dosage , Irinotecan/administration & dosage , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/chemistry , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Cell Line, Tumor , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Drug Liberation , Drug Synergism , Erythrocytes/drug effects , Female , Humans , Irinotecan/chemistry , Irinotecan/pharmacokinetics , Liposomes , Methylamines/chemistry , Mice, Inbred BALB C , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Rabbits , Rats, Sprague-Dawley , Sucrose/analogs & derivatives , Sucrose/chemistry , Tissue Distribution , Tumor Burden/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...