Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 33
1.
J Control Release ; 309: 72-81, 2019 09 10.
Article En | MEDLINE | ID: mdl-31306678

A photopolymerizable hydrogel-based local drug delivery system was developed for the postsurgical treatment of glioblastoma (GBM). We aimed for a local drug combination therapy with paclitaxel (PTX) and temozolomide (TMZ) within a hydrogel to synergistically inhibit tumor growth. The in vitro cytotoxicity of TMZ was assessed in U87MG cells. We demonstrated the synergistic effect of PTX and TMZ on U87MG cells by clonogenic assay. Treatment with TMZ did not induce O6-methylguanine-DNA methyltransferase related drug resistance in tumor-bearing mice. PTX had sustained release for at least 1 month in vivo in healthy mice brains. The drug combination was tolerable and suppressed tumor growth more efficiently than the single drugs in the U87MG orthotopic tumor model. The PTX and TMZ codelivery hydrogel showed superior antitumor effects and can be considered a promising approach for the postsurgical treatment of GBM.


Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Drug Delivery Systems , Glioblastoma/drug therapy , Paclitaxel/administration & dosage , Temozolomide/administration & dosage , Animals , Antineoplastic Agents/therapeutic use , Brain Neoplasms/pathology , Cell Line, Tumor , Drug Combinations , Female , Glioblastoma/pathology , Humans , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Mice, Nude , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/prevention & control , Paclitaxel/therapeutic use , Temozolomide/therapeutic use
2.
Int J Pharm ; 559: 220-227, 2019 Mar 25.
Article En | MEDLINE | ID: mdl-30703501

Lauroyl-gemcitabine lipid nanocapsules (GemC12-LNC) hydrogel, administered intratumorally or perisurgically in the tumor resection cavity, increases animal survival in several orthotopic GBM models. We hypothesized that GemC12-LNC can be used as nanodelivery platform for other drugs, to obtain a combined local therapeutic approach for GBM. Paclitaxel (PTX) was selected as a model molecule and PTX-GemC12-LNC formulation was evaluated in terms of physicochemical and mechanical properties. The PTX-GemC12-LNC hydrogel stability and drug release were evaluated over time showing no significant differences compared to GemC12-LNC. The drug combination was evaluated on several GBM cell lines showing increased cytotoxic activity compared to the original formulation and synergy between PTX and GemC12. Our results suggest that GemC12-LNC hydrogel can be used as nanodelivery platform for dual drug delivery to encapsulate active agents with different mechanisms of action to achieve a better antitumor efficacy against GBM or other solid tumors.


Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Hydrogels/pharmacology , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Deoxycytidine/chemistry , Deoxycytidine/pharmacology , Drug Carriers/chemistry , Drug Combinations , Drug Liberation/drug effects , Hydrogels/chemistry , Injections/methods , Lipids/chemistry , Lipids/pharmacology , Mice , Nanocapsules/chemistry , Nanomedicine/methods , Paclitaxel/chemistry , Paclitaxel/pharmacology , Rats , Gemcitabine
3.
J Drug Target ; 27(5-6): 614-623, 2019.
Article En | MEDLINE | ID: mdl-30633585

INTRODUCTION: We hypothesised that the active targeting of αvß3 integrin overexpressed in neoangiogenic blood vessels and glioblastoma (GBM) cells combined with magnetic targeting of paclitaxel- and SPIO-loaded PLGA-based nanoparticles could improve accumulation of nanoparticles in the tumour and therefore improve the treatment of GBM. METHODS: PTX/SPIO PLGA nanoparticles with or without RGD-grafting were characterised. Their in vitro cellular uptake and cytotoxicity was evaluated by fluorospectroscopy and MTT assay. In vivo safety and anti-tumour efficacy of different targeting strategies were evaluated in orthotopic U87MG tumour model over multiple intravenous injections. RESULTS: The nanoparticles of 250 nm were negatively charged. RGD targeted nanoparticles showed a specific and higher cellular uptake than untargeted nanoparticles by activated U87MG and HUVEC cells. In vitro IC50 of PTX after 48 h was ∼1 ng/mL for all the PTX-loaded nanoparticles. The median survival time of the mice treated with magnetic targeted nanoparticles was higher than the control (saline) mice or mice treated with other evaluated strategies. The 6 doses of PTX did not induce any detectable toxic effects on liver, kidney and heart when compared to Taxol. CONCLUSION: The magnetic targeting strategy resulted in a better therapeutic effect than the other targeting strategies (passive, active).


Glioblastoma/drug therapy , Nanoparticles/chemistry , Paclitaxel/chemistry , Paclitaxel/pharmacology , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line , Cell Line, Tumor , Drug Carriers/chemistry , Female , Human Umbilical Vein Endothelial Cells , Humans , Integrin alphaVbeta3/metabolism , Magnetics/methods , Mice , Mice, Nude , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Tissue Distribution/drug effects , Xenograft Model Antitumor Assays/methods
4.
Nanomedicine (Lond) ; 13(16): 1999-2013, 2018 08.
Article En | MEDLINE | ID: mdl-30204064

AIM: Anticancer drug-loaded hydrogels are a promising strategy for the local treatment of incurable brain tumors such as glioblastoma (GBM). Recently, we demonstrated the efficacy of lauroyl-gemcitabine lipid nanocapsule hydrogel (GemC12-LNC) in a U-87 MG xenograft orthotopic mouse model. In this study, we developed a reliable and reproducible surgical procedure to resect orthotopic GBM tumors in rats. GemC12-LNC hydrogel integrity was tested after brain administration in rats and its anti-tumor efficacy was tested on a 9L syngeneic orthotopic model. RESULTS: We demonstrated that LNC integrity is maintained at least for one week after local administration of GemC12-LNC. GemC12-LNC was able to delay the formation of recurrences in 9L tumor-bearing resected rats, demonstrating the efficacy of this nanomedicine hydrogel in this preclinical model. CONCLUSION: Our results confirm that GemC12-LNC, a hydrogel uniquely formed by a nanocarrier and a cytotoxic drug, could be a promising and safe delivery tool for the local treatment of operable GBM tumors.


Brain Neoplasms/drug therapy , Deoxycytidine/analogs & derivatives , Glioblastoma/drug therapy , Hydrogels/chemistry , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Deoxycytidine/administration & dosage , Deoxycytidine/chemistry , Deoxycytidine/therapeutic use , Female , Magnetic Resonance Imaging , Nanomedicine/methods , Rats , Rats, Sprague-Dawley , Gemcitabine
5.
Int J Pharm ; 551(1-2): 322-328, 2018 Nov 15.
Article En | MEDLINE | ID: mdl-30236645

Acriflavine (ACF) hydrochloride is currently repurposed as multimodal drug, inhibiting hypoxia-inducible factors (HIF) pathways and exerting cytotoxic properties. The aim of this study was to encapsulate ACF in reverse micelles and to incorporate this suspension in lipid nanocapsules (LNC). Designs of experiments were used to work under quality by design conditions. LNC were formulated using a phase-inversion temperature method, leading to an encapsulation efficiency around 80%. In vitro, the encapsulated drug presented similar cytotoxic activity and decrease in HIF activity in 4T1 cells compared to the free drug. In vivo, ACF-loaded nanoparticles (ACF dose of 5 mg/kg) demonstrated a higher antitumor efficacy compared to free ACF on an orthotopic model of murine breast cancer (4T1 cells). Moreover, the use of LNC allowed to drastically decrease the number of administrations compared to the free drug (2 versus 12 injections), suppressing the ACF-induced toxicity.


Acriflavine/administration & dosage , Drug Carriers/administration & dosage , Hypoxia-Inducible Factor 1/antagonists & inhibitors , Lipids/administration & dosage , Mammary Neoplasms, Experimental/drug therapy , Nanocapsules/administration & dosage , Animals , Cell Line, Tumor , Female , Mice , Mice, Inbred BALB C
6.
Int J Nanomedicine ; 13: 4509-4521, 2018.
Article En | MEDLINE | ID: mdl-30127603

INTRODUCTION: Glioblastoma (GBM) therapy is highly challenging, as the tumors are very aggressive due to infiltration into the surrounding normal brain tissue. Even a combination of the available therapeutic regimens may not debulk the tumor completely. GBM tumors are also known for recurrence, resulting in survival rates averaging <18 months. In addition, systemic chemotherapy for GBM has been challenged for its minimal desired therapeutic effects and unwanted side effects. PURPOSE: We hypothesized that paclitaxel (PTX) and superparamagnetic iron oxide (SPIO)-loaded PEGylated poly(lactic-co-glycolic acid) (PLGA)-based nanoparticles (NPs; PTX/SPIO-NPs) can serve as an effective nanocarrier system for magnetic targeting purposes, and we aimed to demonstrate the therapeutic efficacy of this system in an orthotopic murine GBM model. MATERIALS AND METHODS: PTX/SPIO-NPs were prepared by emulsion-diffusion-evaporation method and characterized for physicochemical properties. In vitro cellular uptake of PTX/SPIO-NPs was evaluated by fluorescence microscopy and Prussian blue staining. Orthotopic U87MG tumor model was used to evaluate blood-brain barrier disruption using T1 contrast agent, ex vivo biodistribution, in vivo toxicity and in vivo antitumor efficacy of PTX/SPIO-NPs. RESULTS: PTX/SPIO-NPs were in the size of 250 nm with negative zeta potential. Qualitative cellular uptake studies showed that the internalization of NPs was concentration dependent. Through magnetic resonance imaging, we observed that the blood-brain barrier was disrupted in the GBM area. An ex vivo biodistribution study showed enhanced accumulation of NPs in the brain of GBM-bearing mice with magnetic targeting. Short-term in vivo safety evaluation showed that the NPs did not induce any systemic toxicity compared with Taxol® (PTX). When tested for in vivo efficacy, the magnetic targeting treatment significantly prolonged the median survival time compared with the passive targeting and control treatments. CONCLUSION: Overall, PTX/SPIO-NPs with magnetic targeting could be considered as an effective anticancer targeting strategy for GBM chemotherapy.


Glioblastoma/drug therapy , Lactic Acid/chemistry , Magnetics , Nanoparticles/chemistry , Paclitaxel/therapeutic use , Polyglycolic Acid/chemistry , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/pathology , Cell Line, Tumor , Endocytosis/drug effects , Female , Glioblastoma/pathology , Humans , Mice, Nude , Paclitaxel/pharmacology , Polylactic Acid-Polyglycolic Acid Copolymer , Survival Analysis , Tissue Distribution/drug effects , Xenograft Model Antitumor Assays
7.
Int J Pharm ; 548(1): 522-529, 2018 Sep 05.
Article En | MEDLINE | ID: mdl-30017818

Glioblastoma multiforme (GBM) is the most common primary malignant brain tumor. Despite available therapeutic options, the prognosis for patients with GBM remains very poor. We hypothesized that the intra-operative injection of a photopolymerizable hydrogel into the tumor resection cavity could sustain the release of the anti-cancer drug paclitaxel (PTX) encapsulated in poly (lactic-co-glycolic acid) (PLGA) nanoparticles and prevent GBM recurrence. The tumor was resected 13 days after implantation and a pre-gel solution composed of polyethylene glycol dimethacrylate (PEG-DMA) polymer, a photoinitiator and PTX-loaded PLGA nanoparticles (PTX PLGA-NPs) was injected into the tumor resection cavity. A solid gel filling the whole cavity was formed immediately by photopolymerization using a 400 nm light. PTX in vitro release study showed a burst release (11%) in the first 8 h and a sustained release of 29% over a week. In vitro, U87 MG cells were sensitive to PTX PLGA-NPs with IC50 level of approximately 0.010 µg/mL. The hydrogel was well-tolerated when implanted in the brain of healthy mice for 2 and 4 months. Administration of PTX PLGA-NPs-loaded hydrogel into the resection cavity of GBM orthotopic model lead to more than 50% long-term survival mice (150 days) compared to the control groups (mean survival time 52 days). This significant delay of recurrence is very promising for the post-resection treatment of GBM.


Antineoplastic Agents, Phytogenic/administration & dosage , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Hydrogels/administration & dosage , Nanoparticles/administration & dosage , Paclitaxel/administration & dosage , Animals , Antineoplastic Agents, Phytogenic/chemistry , Apoptosis/drug effects , Brain Neoplasms/surgery , Cell Line, Tumor , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/chemistry , Drug Liberation , Female , Glioblastoma/surgery , Humans , Hydrogels/chemistry , Intraoperative Period , Lactic Acid/administration & dosage , Lactic Acid/chemistry , Methacrylates/administration & dosage , Methacrylates/chemistry , Mice , Nanoparticles/chemistry , Paclitaxel/chemistry , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/chemistry , Polyglycolic Acid/administration & dosage , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer
8.
J Control Release ; 281: 42-57, 2018 07 10.
Article En | MEDLINE | ID: mdl-29753958

Glioblastoma (GBM) is one of the most aggressive and deadliest central nervous system tumors, and the current standard treatment is surgery followed by radiotherapy with concurrent chemotherapy. Nevertheless, the survival period is notably low. Although ample research has been performed to develop an effective therapeutic strategy for treating GBM, the success of extending patients' survival period and quality of life is limited. This review focuses on the strategies developed to address the challenges associated with drug delivery in GBM, particularly nanomedicine. The first part describes major obstacles to the development of effective GBM treatment strategies. The second part focuses on the conventional chemotherapeutic nanomedicine strategies, their limitations and the novel and advanced strategies of nanomedicine, which could be promising for GBM treatment. We also highlighted the prominence of nanomedicine clinical translation. The near future looks bright following the beginning of clinical translation of nanochemotherapy for GBM.


Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Drug Delivery Systems/methods , Glioblastoma/drug therapy , Nanoparticles/chemistry , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Blood-Brain Barrier/metabolism , Clinical Trials as Topic , Drug Liberation , Humans , Nanomedicine/methods , Permeability , Quality of Life , Treatment Outcome
9.
Int J Pharm ; 532(1): 229-240, 2017 Oct 30.
Article En | MEDLINE | ID: mdl-28867450

A substantial drug release from poly(lactic-co-glycolic) acid (PLGA) micro- and nanoparticles can occur in the first hours of immersion, which is referred to as burst release. A strong burst release (when not intentional) is to be avoided as it decreases the efficacy of the treatment and could be dangerous to the host. In this work we analyze the total amount of drug released during burst and respective kinetics in relation to formulations characteristics, experimental conditions and drug molecular properties in 154 drug release experiments with 41 different drugs by partial least squares (PLS) and decision tree regression. The model created enables to quantify to which degree the physicochemical parameters control the burst release from PLGA particles. Our analysis shows that the amount of drug released during burst is mostly influenced by the formulation characteristics and the synthesis parameters, whereas the drug release kinetics is also influenced by the molecular properties of the drug. The variables that significantly influence the amount and kinetics of the burst release are discussed in detail and compared with findings from other researchers. The final regression models are shown to predict the release profile of a new drug, opening the possibility to be applied to systematically manipulate the burst release by means of designing an optimized drug delivery system.


Drug Liberation , Lactic Acid/chemistry , Models, Theoretical , Nanoparticles/chemistry , Polyglycolic Acid/chemistry , Drug Compounding , Pharmaceutical Preparations/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Regression Analysis
10.
J Neurosci Methods ; 284: 96-102, 2017 Jun 01.
Article En | MEDLINE | ID: mdl-28472680

In vitro and in vivo models of experimental glioma are useful tools to gain a better understanding of glioblastoma (GBM) and to investigate novel treatment strategies. However, the majority of preclinical models focus on treating solid intracranial tumours, despite surgical resection being the mainstay in the standard care of patients with GBM today. The lack of resection and recurrence models therefore has undermined efforts in finding a treatment for this disease. Here we present a novel orthotopic tumour resection and recurrence model that has potential for the investigation of local delivery strategies in the treatment of GBM. The model presented is simple to achieve through the use of a biopsy punch, is reproducible, does not require specific or expensive equipment, and results in a resection cavity suitable for local drug delivery systems, such as the implantation or injection of hydrogels. We show that tumour resection is well tolerated, does not induce deleterious neurological deficits, and significantly prolongs survival of mice bearing U-87 MG GBM tumours. In addition, the resulting cavity could accommodate adequate amounts of hydrogels for local delivery of chemotherapeutic agents to eliminate residual tumour cells that can induce tumour recurrence.


Brain Neoplasms/pathology , Brain Neoplasms/surgery , Disease Models, Animal , Glioblastoma/pathology , Glioblastoma/surgery , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/surgery , Animals , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude
11.
Cell Mol Life Sci ; 74(13): 2451-2466, 2017 07.
Article En | MEDLINE | ID: mdl-28210785

Although brain tumours have been documented and recorded since the nineteenth century, 2016 marked 90 years since Percival Bailey and Harvey Cushing coined the term "glioblastoma multiforme". Since that time, although extensive developments in diagnosis and treatment have been made, relatively little improvement on prognosis has been achieved. The resilience of GBM thus makes treating this tumour one of the biggest challenges currently faced by neuro-oncology. Aggressive and robust development, coupled with difficulties of complete resection, drug delivery and therapeutic resistance to treatment are some of the main issues that this nemesis presents today. Current treatments are far from satisfactory with poor prognosis, and focus on palliative management rather than curative intervention. However, therapeutic research leading to developments in novel treatment stratagems show promise in combating this disease. Here we present a review on GBM, looking at the history and advances which have shaped neurosurgery over the last century that cumulate to the present day management of GBM, while also exploring future perspectives in treatment options that could lead to new treatments on the road to a cure.


Brain Neoplasms/therapy , Glioblastoma/therapy , Animals , Brain Neoplasms/pathology , Clinical Trials as Topic , Drug Resistance, Neoplasm , Glioblastoma/pathology , Humans , Neoplastic Stem Cells/pathology
12.
Expert Opin Drug Deliv ; 14(7): 851-864, 2017 07.
Article En | MEDLINE | ID: mdl-27730820

INTRODUCTION: Nanomedicine has emerged as a major field of academic research with direct impact on human health. While a first generation of products has been successfully commercialized and has significantly contributed to enhance patient's life, recent advances in material design and the emergence of new therapeutics are contributing to the development of more sophisticated systems. As the field matures, it is important to comprehend the challenges related to nanoparticle commercial development for a more efficient and predictable path to the clinic. Areas covered: The review provides an overview of nanoparticle-based delivery systems currently on the market and in clinical trials, and discuss the principal challenges for their commercial development, both from a manufacturing and regulatory perspective, to help gain understanding of the translational path for these systems. Expert opinion: Clinical translation of nanoparticle-based delivery systems remains challenging on account of their 3D nanostructure and requires robust nano-manufacturing process along with adequate analytical tools and methodologies. By identifying early enough in the development the product critical attributes and understanding their impact on the therapeutic performance, the developers of nanopharmaceuticals will be better equipped to develop efficient product pipelines. Second-generation products are expected to broaden nanopharmaceutical global market in the upcoming years.


Drug Delivery Systems , Nanoparticles/administration & dosage , Animals , Europe , Humans , Legislation, Drug , Nanomedicine , United States
13.
J Control Release ; 210: 95-104, 2015 Jul 28.
Article En | MEDLINE | ID: mdl-25982679

Glioblastoma is the most frequent primary malignant brain tumor in adults. Despite treatments including surgery, radiotherapy and chemotherapy by oral Temozolomide (TMZ), the prognosis of patients with glioblastoma remains very poor. We hypothesized that a polyethylene glycol dimethacrylate (PEG-DMA) injectable hydrogel would provide a sustained and local delivery of TMZ. The hydrogel photopolymerized rapidly (<2min) and presented a viscous modulus (≈10kPa). TMZ release kinetic presented two phases: a linear burst release of 45% of TMZ during the first 24h, followed by a logarithmic release of 20% over the first week. The in vivo tolerability study showed that the unloaded hydrogel did not induce apoptosis in mice brains nor increased microglial activation. In vivo, the anti-tumor efficacy of TMZ-hydrogel was evaluated on xenograft U87MG tumor-bearing nude mice. The tumor weight of mice treated with the photopolymerized TMZ hydrogel drastically decreased compared with all other groups. Higher apoptosis (located at the center of the tumor) was also observed. The present study demonstrates the potential of a photopolymerizable TMZ-loaded hydrogel to treat glioblastoma.


Antineoplastic Agents, Alkylating/administration & dosage , Brain Neoplasms/drug therapy , Dacarbazine/analogs & derivatives , Drug Delivery Systems , Glioblastoma/drug therapy , Hydrogels/administration & dosage , Animals , Antineoplastic Agents, Alkylating/chemistry , Antineoplastic Agents, Alkylating/radiation effects , Antineoplastic Agents, Alkylating/therapeutic use , Apoptosis/drug effects , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Dacarbazine/administration & dosage , Dacarbazine/chemistry , Dacarbazine/radiation effects , Dacarbazine/therapeutic use , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/radiation effects , Delayed-Action Preparations/therapeutic use , Female , Glioblastoma/pathology , Humans , Hydrogels/chemistry , Hydrogels/radiation effects , Hydrogels/therapeutic use , Light , Methacrylates/chemistry , Methacrylates/radiation effects , Mice, Nude , Microglia/drug effects , Polyethylene Glycols/chemistry , Polyethylene Glycols/radiation effects , Temozolomide , Tumor Burden/drug effects
14.
Int J Pharm ; 481(1-2): 154-61, 2015 Mar 15.
Article En | MEDLINE | ID: mdl-25644286

Glioblastoma is the most frequent primary malignant brain tumor in adults. Despite treatments including surgery, radiotherapy and chemotherapy by oral Temozolomide (TMZ), the prognosis of patients with glioblastoma remains very poor. This is partly due to the resistance of malignant cells to therapy particularly TMZ. Overexpression of epidermal growth factor receptor (EGFR) and Galectin-1 by tumor cells significantly contributes to TMZ resistance. The purpose of this study was to evaluate in vivo, the effect of local administration by convection enhanced delivery (CED) of the anti-EGFR and anti-Galectin-1 siRNAs administered separately or in combination on (i) the survival of nude mice-bearing orthotopic U87MG glioblastoma cells and on (ii) the EGFR and Galectin-1 expression in excised U87MG tumor tissue. Both siRNAs were carried by chitosan lipid nanocapsules (LNCs). Survival of mice treated 14 days after tumor implantation by the combination of anti-EGFR and anti-Galectin-1 siRNAs and TMZ (40 mg/kg) was significantly increased compared to animals treated by single anti-EGFR or anti-Galectin-1 siRNAs carried by chitosan-LNCs. This was confirmed by a decreased EGFR and Galectin-1 expression at the protein level in excised U87MG tumor tissue, 8 days post-transfection, visualized by immunofluorescence. This study demonstrates the potential of our strategy in glioblastoma therapy.


Antineoplastic Agents, Alkylating/administration & dosage , Dacarbazine/analogs & derivatives , ErbB Receptors/genetics , Galectin 1/genetics , Nanocapsules/administration & dosage , RNA, Small Interfering/administration & dosage , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Line, Tumor , Chitosan/chemistry , Dacarbazine/administration & dosage , Drug Delivery Systems , Drug Resistance, Neoplasm/drug effects , Female , Gene Silencing , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Lipids/chemistry , Mice, Nude , Nanocapsules/chemistry , Temozolomide , Tumor Burden/drug effects
15.
Int J Pharm ; 479(2): 399-407, 2015 Feb 20.
Article En | MEDLINE | ID: mdl-25578367

Paclitaxel (PTX)-loaded polymeric micelles (M-PTX) have been shown to enhance the blood flow and oxygenation of tumors 24h after treatment. We hypothesized that these changes in the tumor microenvironment could lead to an enhancement of the EPR (enhanced permeability and retention) effect. M-PTX, administered 24h before analysis, increased the accumulation of macromolecules, nanoparticles and polymeric micelles in tumors. This increased EPR effect could be linked to normalization of the tumor vasculature and decreased interstitial fluid pressure. M-PTX used as a pre-treatment allowed a more effective delivery of three nanomedicines into tumors: polymeric micelles, liposomes and nanoparticles. These experiments demonstrate an enhanced EPR effect after M-PTX treatment, which lead to better availability and enhanced efficacy of a subsequent treatment with nanomedicines.


Antineoplastic Agents, Phytogenic/administration & dosage , Nanoparticles , Neoplasms/drug therapy , Paclitaxel/administration & dosage , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Female , Liposomes , Male , Mice , Mice, Inbred BALB C , Micelles , Nanomedicine , Neoplasms/pathology , Paclitaxel/pharmacology , Permeability , Polymers/chemistry , Time Factors , Tumor Microenvironment/drug effects
16.
J Control Release ; 198: 35-54, 2015 Jan 28.
Article En | MEDLINE | ID: mdl-25481448

A major issue in current cancer therapies is the lack of selectivity, which leads to damage in healthy tissues. Therefore, researchers have focused on numerous innovative targeting strategies to address this problem with the goal of increasing selectivity to avoid or minimize accumulation in healthy tissues. These strategies include (i) passive targeting, (ii) active targeting and (iii) stimuli-mediated targeting. Moreover, due to the high intra- and inter-variability found in tumors, nanotheranostics, which is the combination of a therapeutic and an imaging agent in a single vector, have emerged as indispensable tools for personalized therapy. Superparamagnetic iron oxide (SPIO) are MRI contrast agents that produce predominant T2 relaxation effects with excellent sensitivity compared with other MRI agents. Therefore, they have received increased interest in the field of theranostics during the past decade. However, few studies have been successfully conducted in vivo. This review aims to provide an overview of the targeted SPIO-based nanotheranostics recently used in pre-clinical studies and the major obstacles to in vivo studies and clinical translation. In the first section, we discuss personalized therapy as a biomedical application of theranostics. Then, we summarize the different imaging agents that have been used for theranostic purposes, with a focus on SPIO. In the third section, we detail recent advances in targeted SPIO-based nanotheranostics that have been used in pre-clinical studies. In the final sections, we discuss the limitations for in vivo studies, clinical translation and the clinical perspectives of SPIO-based nanotheranostics.


Antineoplastic Agents , Contrast Media , Ferric Compounds , Nanostructures , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Contrast Media/administration & dosage , Contrast Media/therapeutic use , Ferric Compounds/administration & dosage , Ferric Compounds/therapeutic use , Humans , Nanostructures/administration & dosage , Nanostructures/therapeutic use
17.
Int J Pharm ; 476(1-2): 9-15, 2014 Dec 10.
Article En | MEDLINE | ID: mdl-25245548

The purpose of this study was to develop vitamin E-based micelles loaded with Doxorubicin (DOX) (DOX-TOS-TPGS), taking advantages of the anti-cancer activity of vitamin E derivatives: Tocopherol succinate (TOS) and D-α-tocopherol polyethylene2000 succinate (TPGS). Therefore, we developed micelles consisting in a mixture of TOS (as solubilizer) and TPGS2000 (as stabilizer) (1:1). DOX-TOS-TPGS micelles exhibited a size of 78 nm and a ζ potential of -7 mV. High drug loading (40% w/w) was achieved. The critical micellar concentration was determined at 14 µg/ml. In vitro, after 24 h, DOX-TOS-TPGS micelles exhibited higher cytotoxicity than free-DOX (IC50 on MCF-7 cells, at 24 h, 58 vs 5 µg/ml). In vivo anti-tumor efficacy, performed on two tumor models (CT26 and MCF-7), demonstrated a 100% long-term survival of mice when treated with DOX-TOS-TPGS compared to DOX-free. Interestingly, the survival time of mice treated with unloaded TOS-TPGS micelles was similar to DOX-free, indicating an anti-cancer activity of vitamin E derivatives. Based on these results, it can be concluded that the formulations developed in this work may be considered as an effective DOX delivery system for cancer chemotherapy.


Antibiotics, Antineoplastic/pharmacology , Doxorubicin/pharmacology , Drug Delivery Systems , Vitamin E/analogs & derivatives , Animals , Antibiotics, Antineoplastic/administration & dosage , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Doxorubicin/administration & dosage , Excipients/chemistry , Female , Humans , Inhibitory Concentration 50 , MCF-7 Cells , Mice , Mice, Nude , Micelles , Polyethylene Glycols/chemistry , Survival Rate , Vitamin E/chemistry , alpha-Tocopherol/chemistry
18.
J Control Release ; 194: 82-91, 2014 Nov 28.
Article En | MEDLINE | ID: mdl-25178270

Multifunctional nanoparticles combining therapy and imaging have the potential to improve cancer treatment by allowing personalized therapy. Herein, we aimed to compare in vivo different strategies in terms of targeting capabilities: (1) passive targeting via the EPR effect, (2) active targeting of αvß3 integrin via RGD grafting, (3) magnetic targeting via a magnet placed on the tumor and (4) the combination of magnetic targeting and active targeting of αvß3 integrin. For a translational approach, PLGA-based nanoparticles loaded with paclitaxel and superparamagnetic iron oxides were used. Electron Spin Resonance spectroscopy and Magnetic Resonance Imaging (MRI) were used to both quantify and visualize the accumulation of multifunctional nanoparticles into the tumors. We demonstrate that compared to untargeted or single targeted nanoparticles, the combination of both active strategy and magnetic targeting drastically enhanced (i) nanoparticle accumulation into the tumor tissue with an 8-fold increase compared to passive targeting (1.12% and 0.135% of the injected dose, respectively), (ii) contrast in MRI (imaging purpose) and (iii) anti-cancer efficacy with a median survival time of 22 days compared to 13 for the passive targeting (therapeutic purpose). Double targeting of nanoparticles to tumors by different mechanisms could be a promising translational approach for the management of therapeutic treatment and personalized therapy.


Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacokinetics , Drug Delivery Systems/methods , Ferric Compounds/chemistry , Magnetics , Neoplasms/drug therapy , Neoplasms/metabolism , Paclitaxel/administration & dosage , Paclitaxel/pharmacokinetics , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Lactic Acid , Magnetic Resonance Imaging , Metal Nanoparticles , Mice , Mice, Inbred BALB C , Paclitaxel/pharmacology , Particle Size , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , Tissue Distribution , Xenograft Model Antitumor Assays
19.
Eur J Pharm Biopharm ; 88(1): 252-60, 2014 Sep.
Article En | MEDLINE | ID: mdl-24859391

We hypothesized that nanosuspensions could be promising for the delivery of the poorly water soluble anti-cancer multi-targeted kinase inhibitor, MTKi-327. Hence, the aims of this work were (i) to evaluate the MTKi-327 nanosuspension for parenteral and oral administrations and (ii) to compare this nanosuspension with other nanocarriers in terms of anti-cancer efficacy and pharmacokinetics. Therefore, four formulations of MTKi-327 were studied: (i) PEGylated PLGA-based nanoparticles, (ii) self-assembling PEG750-p-(CL-co-TMC) polymeric micelles, (iii) nanosuspensions of MTKi-327; and (iv) Captisol solution (pH=3.5). All the nano-formulations presented a size below 200 nm. Injections of the highest possible dose of the three nano-formulations did not induce any side effects in mice. In contrast, the maximum tolerated dose of the control Captisol solution was 20-fold lower than its highest possible dose. The highest regrowth delay of A-431-tumor-bearing nude mice was obtained with MTKi-327 nanosuspension, administered intravenously, at a dose of 650 mg/kg. After intravenous and oral administration, the AUC0₋∞ of MTKi-327 nanosuspension was 2.4-fold greater than that of the Captisol solution. Nanosuspension may be considered as an effective anti-cancer MTKi-327 delivery method due to (i) the higher MTKi-327 maximum tolerated dose, (ii) the possible intravenous injection of MTKi-327, (iii) its ability to enhance the administered dose and (iv) its higher efficacy.


Antineoplastic Agents/administration & dosage , Drug Delivery Systems , Macrocyclic Compounds/administration & dosage , Protein Kinase Inhibitors/administration & dosage , beta-Cyclodextrins/chemistry , Administration, Oral , Animals , Antineoplastic Agents/chemistry , Area Under Curve , Drug Screening Assays, Antitumor , Humans , Hydrogen-Ion Concentration , Maximum Tolerated Dose , Mice , Mice, Nude , Micelles , Nanoparticles/chemistry , Neoplasm Transplantation , Neoplasms/drug therapy , Polymers/chemistry , Solubility , Suspensions
20.
J Control Release ; 182: 33-44, 2014 May 28.
Article En | MEDLINE | ID: mdl-24631865

This review aims to highlight the development of novel vitamin E conjugates for the vectorization of active pharmaceutical ingredients through nanotechnologies. The physico-chemical and biological properties of vitamin E derivatives offer multiple advantages in drug delivery like biocompatibility, improvement of drug solubility and anticancer activity. Nanomedicines have shown high potential in drug delivery since (i) they may offer better drug biopharmaceutical properties such as longer half-life or better bioavailability and (ii) they have shown benefits in cancer therapy by improving anticancer drug therapeutic index. Vitamin E-based nanomedicines were developed to combine the pharmaceutical properties of both vitamin E and nanomedicines for two purposes: (i) to improve water solubility of hydrophobic drugs and (ii) to enhance the therapeutic efficiency of anticancer agents. This review is divided into three parts: the first one describes the biology and the metabolic functions of vitamin E, the second one focuses on the anticancer activity of two vitamin E derivatives: vitamin E succinate (TOS) and vitamin E polyethylene glycol-succinate (TPGS). Finally, in the third part, we discuss vitamin E derivatives based-nanomedicines.


Antineoplastic Agents/administration & dosage , Drug Delivery Systems , Vitamin E/administration & dosage , Animals , Antineoplastic Agents/chemistry , Humans , Nanomedicine , Neoplasms/drug therapy , Vitamin E/chemistry
...