Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
2.
Int J Pharm X ; 2: 100049, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32490374

ABSTRACT

Carfilzomib-loaded polymeric micelles (CFZ-PM) based on poly(ethylene glycol)-b-poly(N-2-benzoyloxypropyl methacrylamide) (mPEG-b-p(HPMA-Bz)) were prepared with the aim to improve the maximum tolerated dose of carfilzomib in a "humanized" bone marrow-like scaffold model. For this, CFZ-PM were prepared and characterized for their size, carfilzomib loading and cytotoxicity towards multiple myeloma cells. Further, circulation and tumor & tissue distribution of fluorescently labeled micelles were determined. Tolerability of CFZ-PM versus the clinical approved formulation - Kyprolis® - was assessed. CFZ-PM presented small diameter below 55 nm and low PDI < 0.1. Cy7-labeled micelles circulated for extended periods of time with over 80% of injected dose in circulation at 24 h after intravenous injection and 1.3% of the injected dose of Cy7-labeled micelles accumulated in myeloma tumor-bearing scaffolds. Importantly, CFZ-PM were well tolerated whereas Kyprolis® showed adverse effects. Kyprolis® dosed at the maximum tolerated dose, as well as CFZ-PM, did not show therapeutic benefit, while multiple myeloma cells showed sensitivity in vitro, underlining the importance of the bone marrow crosstalk in testing novel formulations. Overall, this work indicates that PM are potential drug carriers of carfilzomib.

3.
Cells ; 8(12)2019 11 25.
Article in English | MEDLINE | ID: mdl-31775322

ABSTRACT

Extracellular vesicles (EVs) are mediators of intercellular communication by transferring functional biomolecules from their originating cells to recipient cells. This intrinsic ability has gained EVs increased scientific interest in their use as a direct therapeutic in the field of regenerative medicine or as vehicles for drug delivery. EVs derived from stem cells or progenitor cells can act as paracrine mediators to promote repair and regeneration of damaged tissues. Despite substantial research efforts into EVs for various applications, their use remains limited by the lack of highly efficient and scalable production methods. Here, we present the biofabrication of cell-derived nanovesicles (NVs) as a scalable, efficient, and cost-effective production alternative to EVs. We demonstrate that NVs have a comparable size and morphology as EVs, but lack standard EV (surface) markers. Additionally, in vitro uptake experiments show that human fetal cardiac fibroblast, endothelial cells, and cardiomyocyte progenitor cells internalize NVs. We observed that cardiac progenitor cell-derived NVs and EVs are capable of activating mitogen-activated protein kinase 1/2 (MAPK1/2)-extracellular signal-regulated kinase, and that both NVs and EVs derived from A431 and HEK293 cells can functionally deliver Cre-recombinase mRNA or protein to other cells. These observations indicate that NVs may have similar functional properties as EVs. Therefore, NVs have the potential to be applied for therapeutic delivery and regenerative medicine purposes.


Subject(s)
Drug Carriers/pharmacology , Endothelial Cells/drug effects , Myocytes, Cardiac/drug effects , Nanoparticles/therapeutic use , Stem Cells/cytology , Endothelial Cells/cytology , HEK293 Cells , Humans , Myocytes, Cardiac/cytology , Proteins/administration & dosage , RNA, Messenger/administration & dosage , Regenerative Medicine/methods
4.
Int J Pharm X ; 1: 100011, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31517276

ABSTRACT

Bortezomib is a proteasome inhibitor used for the treatment of multiple myeloma. The poor pharmacokinetic profile and off-target adverse effects provide a strong incentive to develop drug delivery systems for bortezomib. In the past, liposomal encapsulation has been proven to improve the therapeutic index of a variety of anti-neoplastic therapeutics. Here, we developed and characterized liposomal bortezomib formulations in order to find the most optimal loading conditions. Polyols were used to entrap bortezomib inside the liposomes as boronate ester via a remote loading strategy. Effect of various polyols, incubation duration, temperature, and total lipid concentration on loading efficiency was examined. Moreover, the effect of drug/lipid ratio on the release kinetics was studied. Loading efficiency was maximal when using meglumine plus mannitol as entrapping agents. Loading at room temperature was better than at 60 °C and loading efficiency was increased with increasing total lipid concentrations. There was a positive correlation between drug/lipid ratio and released amount of bortezomib. In vitro release kinetics in HBS and human plasma showed time dependent release. In HBS, at 4 °C, only 20% of the drug was released in three weeks, whereas at 37 °C 85% of the drug was released in 24 h. In human plasma, 5% of the drug retained after 24 h indicating faster release. Taken together, the most favorable liposomal formulation of bortezomib should be further exploited to study in vitro and in vivo efficacy performance.

5.
J Control Release ; 296: 232-240, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30682443

ABSTRACT

Glucocorticoids are the cornerstone in the clinic for treatment of hematological malignancies, including multiple myeloma. Nevertheless, poor pharmacokinetic properties of glucocorticoids require high and frequent dosing with the off-target adverse effects defining the maximum dose. Recently, nanomedicine formulations of glucocorticoids have been developed that improve the pharmacokinetic profile, limit adverse effects and improve solid tumor accumulation. Multiple myeloma is a hematological malignancy characterized by uncontrolled growth of plasma cells. These tumors initiate increased angiogenesis and microvessel density in the bone marrow, which might be exploited using nanomedicines, such as liposomes. Nano-sized particles can accumulate as a result of the increased vascular leakiness at the bone marrow tumor lesions. Pre-clinical screening of novel anti-myeloma therapeutics in vivo requires a suitable animal model that represents key features of the disease. In this study, we show that fluorescently labeled long circulating liposomes were found in plasma up to 24 h after injection in an advanced human-mouse hybrid model of multiple myeloma. Besides the organs involved in clearance, liposomes were also found to accumulate in tumor bearing human-bone scaffolds. The therapeutic efficacy of liposomal dexamethasone phosphate was evaluated in this model showing strong tumor growth inhibition while free drug being ineffective at an equivalent dose (4 mg/kg) regimen. The liposomal formulation slightly reduced total body weight of myeloma-bearing mice during the course of treatment, which appeared reversible when treatment was stopped. Liposomal dexamethasone could be further developed as monotherapy or could fit in with existing therapy regimens to improve therapeutic outcomes for multiple myeloma.


Subject(s)
Antineoplastic Agents, Hormonal/administration & dosage , Dexamethasone/administration & dosage , Glucocorticoids/administration & dosage , Multiple Myeloma/drug therapy , Animals , Body Weight/drug effects , Bone and Bones/metabolism , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Liposomes , Mice, Knockout , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Tissue Distribution , Tumor Burden/drug effects
6.
Int J Nanomedicine ; 13: 8105-8118, 2018.
Article in English | MEDLINE | ID: mdl-30555229

ABSTRACT

PURPOSE: Liposomal drug delivery can improve the therapeutic index of treatments for multiple myeloma. However, an appropriate 3D model for the in vitro evaluation of liposomal drug delivery is lacking. In this study, we applied a previously developed 3D bone marrow (BM) myeloma model to examine liposomal drug therapy. MATERIAL AND METHODS: Liposomes of different sizes (~75-200 nm) were tested in a 3D BM myeloma model, based on multipotent mesenchymal stromal cells, endothelial progenitor cells, and myeloma cells cocultured in hydrogel. The behavior and efficacy of liposomal drug therapy was investigated, evaluating the feasibility of testing liposomal drug delivery in 3D in vitro. Intracellular uptake of untargeted and integrin α4ß1 (very late antigen-4) targeted liposomes was compared in myeloma and supporting cells, as well as the effectivity of free and liposome-encapsulated chemotherapy (bortezomib, doxorubicin). Either cocultured myeloma cell lines or primary CD138+ myeloma cells received the treatments. RESULTS: Liposomes (~75-110 nm) passively diffused throughout the heterogeneously porous (~80-850 nm) 3D hydrogel model after insertion. Cellular uptake of liposomes was observed and was increased by targeting very late antigen-4. Liposomal bortezomib and doxorubicin showed increased cytotoxic effects toward myeloma cells compared with the free drugs, using either a cell line or primary myeloma cells. Cytotoxicity toward supporting BM cells was reduced using liposomes. CONCLUSION: The 3D model allows the study of liposome-encapsulated molecules on multiple myeloma and supporting BM cells, looking at cellular targeting, and general efficacy of the given therapy. The advantages of liposomal drug delivery were demonstrated in a primary myeloma model, enabling the study of patient-to-patient responses to potential drugs and treatment regimes.


Subject(s)
Bone Marrow/drug effects , Doxorubicin/administration & dosage , Drug Delivery Systems , Liposomes/administration & dosage , Models, Biological , Multiple Myeloma/drug therapy , Antibiotics, Antineoplastic/administration & dosage , Bone Marrow/pathology , Cell Proliferation , Humans , In Vitro Techniques , Multiple Myeloma/pathology , Tumor Cells, Cultured
7.
J Control Release ; 287: 194-215, 2018 10 10.
Article in English | MEDLINE | ID: mdl-30165140

ABSTRACT

Hematological malignancies (HM) are a collection of malignant transformations originating from cells in the primary or secondary lymphoid organs. Leukemia, lymphoma, and multiple myeloma comprise the three major types of HM. Current treatment consists of bone marrow transplantation, radiotherapy, immunotherapy and chemotherapy. Although, many chemotherapeutic drugs are clinically available for the treatment of HM, the use of these agents is limited due to dose-related toxicity and lack of specificity to tumor tissue. Moreover, the poor pharmacokinetic profile of most of the chemotherapeutics requires high dosage and frequent administration to maintain therapeutic levels at the target site, both increasing adverse effects. This underlines an urgent need for a suitable drug delivery system to improve efficacy, safety, and pharmacokinetic properties of conventional therapeutics. Nanomedicines have proven to enhance these properties for anticancer therapeutics. The most extensively studied nanomedicine systems are lipid-based nanoparticles and polymeric nanoparticles. Typically, nanomedicines are small sub-micron sized particles in the size range of 20-200 nm. The biocompatible and biodegradable nature of nanomedicines makes them attractive vehicles to improve drug delivery. Their small size allows them to extravasate and accumulate at malignant sites passively by means of the enhanced permeability and retention (EPR) effect, resulting from rapid angiogenesis and inflammation. Moreover, the specificity to the target tissue can be further enhanced by surface modification of nanoparticles. This review describes currently available therapies as well as limitations and potential advantages of nanomedicine formulations for treatment of various types of HM. Additionally, recent investigational and approved nanomedicine formulations and their limited applications in HM are discussed.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems/methods , Hematologic Neoplasms/therapy , Animals , Antineoplastic Agents/therapeutic use , Bone Marrow/drug effects , Bone Marrow/pathology , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/pathology , Hematologic Neoplasms/radiotherapy , Hematopoietic Stem Cell Transplantation/methods , Humans , Immunotherapy/methods , Liposomes/chemistry , Micelles , Nanomedicine/methods , Tumor Microenvironment/drug effects
8.
J Control Release ; 243: 243-249, 2016 12 10.
Article in English | MEDLINE | ID: mdl-27773734

ABSTRACT

Cancers are abundantly infiltrated by inflammatory cells that are modulated by tumor cells to secrete mediators fostering tumor cell survival and proliferation. Therefore, agents that interfere with inflammatory signaling molecules or specific immune cell populations have been investigated as anticancer drugs. Corticosteroids are highly potent anti-inflammatory drugs, whose activity is intensified when targeted by nanocarrier systems. Liposome-targeted corticosteroids have been shown to inhibit tumor growth in different syngeneic murine tumor models as well as human xenograft mouse models, which is attributed to a switch in the tumor microenvironment from a pro-inflammatory to an anti-inflammatory state. Despite the recognized value of implantation tumor models in preclinical research, the "acute" inflammation induced by inoculation of tumor cells together with the exponential tumor growth in a relatively short period of time does not resemble slow progressive human disease that develops in situ. Therefore, in this study, the antitumor effect of liposomal corticosteroids was investigated in a clinically more relevant setting of transgenic mice developing spontaneous breast carcinomas. Here we show that liposomal prednisolone phosphate inhibits the growth of spontaneous breast carcinoma. Interestingly, the liposomal prednisolone was significantly more active than free drug. At 72h after injection of the liposomal formulation, 3µg prednisolone per gram of tumor tissue was recovered whereas no drug could be recovered after injection of the free agent. This indicates that, despite etiological and morphological differences between implanted and spontaneous tumor models, EPR-mediated accumulation of drug occurs to similar extent in this spontaneous mammary carcinoma model as in the syngeneic tumor models. Finally, we analyzed miRNA profiles in the MMTV/neu model and showed that the top 10 of miRNAs in the MMTV/neu tumor consisted of miRNAs with a known involvement in breast carcinoma proliferation and metastasis. The only exception was the appearance of miR-146b, a known inflammation-regulating miRNA species, after liposomal prednisolone treatment.


Subject(s)
Antineoplastic Agents, Hormonal/administration & dosage , Glucocorticoids/administration & dosage , Mammary Neoplasms, Experimental/drug therapy , Prednisolone/analogs & derivatives , Animals , Antineoplastic Agents, Hormonal/pharmacology , Female , Glucocorticoids/pharmacology , Humans , Liposomes , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , MicroRNAs/metabolism , Prednisolone/administration & dosage , Prednisolone/pharmacology , Time Factors , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...