Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Mol Pharm ; 15(7): 2594-2605, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29763568

ABSTRACT

We previously developed a new zinc(II) phthalocyanine (ZnPc) derivative (Pc 1) conjugated to poly-L-glutamic acid (PGA) (1-PG) to address the limitations of ZnPc as part of an antitumor photodynamic therapy approach, which include hydrophobicity, phototoxicity, and nonselectivity in biodistribution and tumor targeting. During this study, we discovered that 1-PG possessed high near-infrared (NIR) light absorptivity (λmax = 675 nm), good singlet oxygen generation efficiency in an aqueous environment, and enhanced photocytotoxic efficacy and cancer cell uptake in vitro. In the current study, we discovered that 1-PG accumulated in 4T1 mouse mammary tumors, with a retention time of up to 48 h. Furthermore, as part of an antitumor PDT, low dose 1-PG (2 mg of Pc 1 equivalent/kg) induced a greater tumor volume reduction (-74 ± 5%) when compared to high dose ZnPc (8 mg/kg, -50 ± 12%). At higher treatment doses (8 mg of Pc 1 equivalent/kg), 1-PG reduced tumor volume maximally (-91 ± 6%) and suppressed tumor size to a minimal level for up to 15 days. The kidney, liver, and lungs of the mice treated with 1-PG (both low and high doses) were free from 4T1 tumor metastasis at the end of the study. Telemetry-spectral-echocardiography studies also revealed that PGA (65 mg/kg) produced insignificant changes to the cardiovascular physiology of Wistar-Kyoto rats when administered in vivo. Results indicate that PGA displays an excellent cardiovascular safety profile, underlining its suitability for application as a nanodrug carrier in vivo. These current findings indicate the potential of 1-PG as a useful photosensitizer candidate for clinical PDT.


Subject(s)
Indoles/administration & dosage , Nanoconjugates/chemistry , Neoplasms/drug therapy , Organometallic Compounds/administration & dosage , Photochemotherapy/methods , Photosensitizing Agents/administration & dosage , Animals , Cardiotoxicity/diagnostic imaging , Cardiotoxicity/etiology , Cell Line, Tumor/transplantation , Disease Models, Animal , Drug Screening Assays, Antitumor , Echocardiography , Humans , Indoles/adverse effects , Indoles/pharmacokinetics , Male , Mice , Mice, Inbred BALB C , Neoplasms/pathology , Organometallic Compounds/adverse effects , Organometallic Compounds/pharmacokinetics , Photochemotherapy/adverse effects , Photosensitizing Agents/adverse effects , Photosensitizing Agents/pharmacokinetics , Polyglutamic Acid/chemistry , Rats , Rats, Inbred WKY , Tissue Distribution
2.
Nanomedicine ; 13(4): 1447-1458, 2017 05.
Article in English | MEDLINE | ID: mdl-28214608

ABSTRACT

In photodynamic therapy (PDT), the low absorptivity of photosensitizers in an aqueous environment reduces singlet oxygen generation efficiency and thereby decreases photosensitizing efficacy in biological conditions. To circumvent this problem, we designed a phthalocyanine-poly-L-glutamic acid conjugate (1-PG) made from a new phthalocyanine (Pc 1) monofunctionalized to allow adequate conjugation to PGA. The resulting 1-PG conjugate retained high absorptivity in the near-infrared (NIR) region at its λmax 675nm in an aqueous environment. The 1-PG conjugate demonstrated good singlet oxygen generation efficiency, increased uptake by 4 T1 breast cancer cells via clathrin-mediated endocytosis, and enhanced photocytotoxic efficacy. The conjugate also displayed a high light-dark toxicity ratio, approximately 1.5-fold greater than zinc phthalocyanine at higher concentration (10 µM), an important feature for the reduction of dark toxicity and unwanted side effects. These results suggest that the 1-PG conjugate could be a useful alternative for deep tissue treatment with enhanced anti-cancer (PDT) efficacy.


Subject(s)
Glutamic Acid/chemistry , Indoles/chemistry , Nanoconjugates/chemistry , Photochemotherapy , Photosensitizing Agents/chemistry , Animals , Cell Line, Tumor , Endocytosis , Isoindoles , Light , Mice , Molecular Structure , Singlet Oxygen/chemistry
3.
Biomacromolecules ; 16(7): 1924-37, 2015 Jul 13.
Article in English | MEDLINE | ID: mdl-25988940

ABSTRACT

New pH-responsive polymersomes for active anticancer oligonucleotide delivery were prepared from triblock copolymers. The delivery systems were formed by two terminal hydrophilic blocks, PEG and polyglycerolmethacrylate (poly-GMA), and a central weakly basic block, polyimidazole-hexyl methacrylate (poly-ImHeMA), which can complex with oligonucleotides and control vesicle formation/disassembly via pH variations. Targeted polymersomes were prepared by mixing folate-derivatized and underivatized copolymers. At pH 5, ds-DNA was found to complex with the pH-responsive copolymers at a N/P molar ratio above ∼2:1, which assisted the encapsulation of ds-DNA in the polymersomes, while low association was observed at pH 7.4. Cytotoxicity studies performed on folate receptor overexpressing KB and B16-F10 cells and low folate receptor expressing MCF-7 cells showed high tolerance of the polymersomes at up to 3 mg/mL concentration. Studies performed with red blood cells showed that at pH 5.0 the polymersomes have endosomolytic properties. Cytofluorimetric studies showed a 5.5-fold higher uptake of ds-DNA loaded folate-functional polymersomes in KB cells compared to nontargeted polymersomes. In addition, ds-DNA was found to be localized both in the nucleus and in the cytosol. The incubation of luciferase transfected B16-F10 cells with targeted polymersomes loaded with luciferase and Hsp90 expression silencing siRNAs yielded 31 and 23% knockdown in target protein expression, respectively.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Nucleus/genetics , Polyethylene Glycols/chemistry , RNA, Small Interfering/pharmacology , Cytosol/metabolism , Drug Carriers/chemistry , Folate Receptors, GPI-Anchored/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , KB Cells , MCF-7 Cells , Methacrylates/chemical synthesis , Methacrylates/chemistry , Nanoparticles/chemistry
4.
Biomaterials ; 301: 122280, 2023 10.
Article in English | MEDLINE | ID: mdl-37598440

ABSTRACT

Modifying biological agents with polymers such as polyethylene glycol (PEG) has demonstrated clinical benefits; however, post-market surveillance of PEGylated derivatives has revealed PEG-associated toxicity issues, prompting the search for alternatives. We explore how conjugating a poly-l-glutamic acid (PGA) to an anti-insulin growth factor 1 receptor antibody (AVE1642) modulates the bio-nano interface and anti-tumor activity in preclinical prostate cancer models. Native and PGA-modified AVE1642 display similar anti-tumor activity in vitro; however, AVE1642 prompts IGF-1R internalization while PGA conjugation prompts higher affinity IGF-1R binding, thereby inhibiting IGF-1R internalization and altering cell trafficking. AVE1642 attenuates phosphoinositide 3-kinase signaling, while PGA-AVE1642 inhibits phosphoinositide 3-kinase and mitogen-activated protein kinase signaling. PGA conjugation also enhances AVE1642's anti-tumor activity in an orthotopic prostate cancer mouse model, while PGA-AVE1642 induces more significant suppression of cancer cell proliferation/angiogenesis than AVE1642. These findings demonstrate that PGA conjugation modulates an antibody's bio-nano interface, mechanism of action, and therapeutic activity.


Subject(s)
Glutamic Acid , Prostatic Neoplasms , Animals , Mice , Male , Humans , Phosphatidylinositol 3-Kinases , Prostatic Neoplasms/drug therapy , Cell Proliferation , Phosphatidylinositol 3-Kinase , Polyethylene Glycols
5.
Adv Healthc Mater ; 11(7): e2101544, 2022 04.
Article in English | MEDLINE | ID: mdl-34706167

ABSTRACT

Prostate cancer (PCa), one of the leading causes of cancer-related deaths, currently lacks effective treatment for advanced-stage disease. Paclitaxel (PTX) is a highly active chemotherapeutic drug and the first-line treatment for PCa; however, conventional PTX formulation causes severe hypersensitivity reactions and limits PTX use at high concentrations. In the pursuit of high molecular weight, biodegradable, and pH-responsive polymeric carriers, one conjugates PTX to a polyacetal-based nanocarrier to yield a tert-Ser-PTX polyacetal conjugate. tert-Ser-PTX conjugate provides sustained release of PTX over 2 weeks in a pH-responsive manner while also obtaining a degree of epimerization of PTX to 7-epi-PTX. Serum proteins stabilize tert-Ser-PTX, with enhanced stability in human serum versus PBS (pH 7.4). In vitro efficacy assessments in PCa cells demonstrate IC50 values above those for the free form of PTX due to the differential cell trafficking modes; however, in vivo tolerability assays demonstrate that tert-Ser-PTX significantly reduces the systemic toxicities associated with free PTX treatment. tert-Ser-PTX also effectively inhibits primary tumor growth and hematologic, lymphatic, and coelomic dissemination, as confirmed by in vivo and ex vivo bioluminescence imaging and histopathological evaluations in mice carrying orthotopic LNCaP tumors. Overall, the results suggest the application of tert-Ser-PTX as a robust antitumor/antimetastatic treatment for PCa.


Subject(s)
Antineoplastic Agents, Phytogenic , Prostatic Neoplasms , Acetals , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Cell Line, Tumor , Drug Carriers/chemistry , Humans , Male , Mice , Mice, Inbred BALB C , Paclitaxel/chemistry , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Polymers/chemistry , Prostatic Neoplasms/drug therapy
6.
Adv Drug Deliv Rev ; 119: 101-119, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28502767

ABSTRACT

Targeting angiogenesis-related pathologies, which include tumorigenesis and metastatic processes, has become an attractive strategy for the development of efficient guided nanomedicines. In this respect, integrins are cell-adhesion molecules involved in angiogenesis signaling pathways and are overexpressed in many angiogenic processes. Therefore, they represent specific biomarkers not only to monitor disease progression but also to rationally design targeted nanomedicines. Arginine-glycine-aspartic (RGD) containing peptides that bind to specific integrins have been widely utilized to provide ligand-mediated targeting capabilities to small molecules, peptides, proteins, and antibodies, as well as to drug/imaging agent-containing nanomedicines, with the final aim of maximizing their therapeutic index. Within this review, we aim to cover recent and relevant examples of different integrin-assisted nanosystems including polymeric nanoconstructs, liposomes, and inorganic nanoparticles applied in drug/gene therapy as well as imaging and theranostics. We will also critically address the overall benefits of integrin-targeting.


Subject(s)
Integrins/metabolism , Nanoparticles/administration & dosage , Neovascularization, Pathologic/drug therapy , Animals , Drug Delivery Systems/methods , Humans , Nanomedicine/methods , Neovascularization, Pathologic/metabolism
7.
Eur J Pharm Biopharm ; 83(3): 346-57, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23201051

ABSTRACT

A new acid-sensitive drug-delivery nanocarrier has been developed for tumour targeting. The self-assembling co-polymer stearoyl-PEG-poly-sulfadimethoxine methacrylate (stearoyl-PEG-polySDM) was prepared to obtain micelles with responsive behaviour in the physiopathologic pH range. Stearoyl-PEG-polySDM was synthesised using a multi-step procedure that includes pH-sensitive sulfadimethoxine methacrylate polymerisation by AGET-ATRP at the amino terminal side of stearoyl-PEG-NH2. Chemical analysis showed that the stearoyl-PEG-polySDM co-polymer contained a mean of seven methacryloyl sulfadimethoxines per molecule. Potentiometric and turbidimetric analyses showed that stearoyl-PEG-polySDM has an apparent pKa of 7.2 and a cloud point at pH 7.0. In water at pH 7.4, the co-polymer assembled spontaneously into 13.2±3.1 nm micelles with a critical micelle concentration (CMC) of 36 µM. Cell-culture studies showed that the material was more biocompatible with respect to the control Brij-700®. The paclitaxel loading capacity of the micelles was 3.25±0.25% (w/w, %). The colloidal formulations were stable at pH 7.4 for several hours, while at pH 6.5, they rapidly rearranged and aggregated. Fluorescence spectroscopic and cytofluorimetric studies showed that the incubation of MCF-7 tumour cells with fluorescein-labelled stearoyl-PEG-polySDM at pH 6.5 resulted in massive time-dependent cell association, while the incubation at pH 7.4 showed significantly lower cell interaction. Confocal microscopy confirmed that at pH 6.5, the micelles are taken up by cells and that the fluorescein-labelled stearoyl-PEG-polySDM is distributed into the cytosol. At pH 6.5, paclitaxel-loaded stearoyl-PEG-polySDM micelles had a higher cytotoxic effect than the micelles incubated at pH 7.4. The former displayed similar cytotoxic activity to free paclitaxel.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Drug Delivery Systems , Hydrogen-Ion Concentration , Lipids/chemistry , Micelles , Neoplasms/drug therapy , Paclitaxel/therapeutic use , Antineoplastic Agents, Phytogenic/administration & dosage , Flow Cytometry , Humans , MCF-7 Cells , Microscopy, Confocal , Paclitaxel/administration & dosage , Particle Size , Spectrometry, Fluorescence , Spectrometry, Mass, Electrospray Ionization
SELECTION OF CITATIONS
SEARCH DETAIL