Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 193
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Biol Chem ; 300(6): 107325, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38685532

ABSTRACT

Immune checkpoint blockade (ICB) using monoclonal antibodies against programmed cell death protein 1 (PD-1) or programmed death-ligand 1 (PD-L1) is the treatment of choice for cancer immunotherapy. However, low tissue permeability, immunogenicity, immune-related adverse effects, and high cost could be possibly improved using alternative approaches. On the other hand, synthetic low-molecular-weight (LMW) PD-1/PD-L1 blockers have failed to progress beyond in vitro studies, mostly due to low binding affinity or poor pharmacological characteristics resulting from their limited solubility and/or stability. Here, we report the development of polymer-based anti-human PD-L1 antibody mimetics (α-hPD-L1 iBodies) by attaching the macrocyclic peptide WL12 to a N-(2-hydroxypropyl)methacrylamide copolymer. We characterized the binding properties of iBodies using surface plasmon resonance, enzyme-linked immunosorbent assay, flow cytometry, confocal microscopy, and a cellular ICB model. We found that the α-hPD-L1 iBodies specifically target human PD-L1 (hPD-L1) and block the PD-1/PD-L1 interaction in vitro, comparable to the atezolizumab, durvalumab, and avelumab licensed monoclonal antibodies targeting PD-L1. Our findings suggest that iBodies can be used as experimental tools to target hPD-L1 and could serve as a platform to potentiate the therapeutic effect of hPD-L1-targeting small molecules by improving their affinity and pharmacokinetic properties.


Subject(s)
B7-H1 Antigen , Immune Checkpoint Inhibitors , Humans , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , B7-H1 Antigen/metabolism , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/chemistry , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacology , Polymers/chemistry , Cell Line, Tumor
2.
Mol Pharm ; 21(4): 1838-1847, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38413029

ABSTRACT

The extensive use of opioids for chronic pain management has contributed significantly to the current opioid epidemic. While many alternative nonopioid analgesics are available, opioids remain the most potent analgesics for moderate to severe pain management. In addition to the implementation of multimodal analgesia, there is a pressing need for the development of more effective and safer opioids. In this study, we developed a thermoresponsive N-(2-hydroxypropyl) methacrylamide (HPMA) copolymer-based hydromorphone (HMP) prodrug (ProGel-HMP, HMP content = 16.2 wt %, in base form). The aqueous solution of ProGel-HMP was free-flowing at 4 °C but became a hydrogel when the temperature was raised to ≥37 °C, allowing sustained local retention when administered in vivo. When tested in the destabilization of the medial meniscus (DMM) mouse model of osteoarthritis (OA), ProGel-HMP was retained after intra-articular injection in the OA knee joint for at least 2 weeks postinjection, with low extra-articular distribution. ProGel-HMP was not detected in the central nervous system (CNS). A single dose of ProGel-HMP produced rapid and sustained joint pain resolution for greater than 14 days when compared to saline and dose-equivalent HMP controls, likely mediated through peripheral µ-opioid receptors in the knee joint. Systemic analgesia effect was absent in the DMM mice treated with ProGel-HMP, as evident in the lack of difference in tail flick response between the ProGel-HMP-treated mice and the controls (i.e., Healthy, Saline, and Sham). Repeated dosing of ProGel-HMP did not induce tolerance. Collectively, these data support the further development of ProGel-HMP as a potent, safe, long-acting and nonaddictive analgesic for better clinical pain management.


Subject(s)
Analgesia , Drug-Related Side Effects and Adverse Reactions , Osteoarthritis , Prodrugs , Mice , Animals , Hydromorphone , Pain Management , Prodrugs/therapeutic use , Pain/drug therapy , Analgesics, Opioid/adverse effects , Analgesics/therapeutic use
3.
Nanomedicine ; 55: 102716, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38738529

ABSTRACT

Rheumatoid arthritis is a chronic inflammatory autoimmune disease caused by alteration of the immune system. Current therapies have several limitations and the use of nanomedicines represents a promising strategy to overcome them. By employing a mouse model of adjuvant induced arthritis, we aimed to evaluate the biodistribution and therapeutic effects of glucocorticoid dexamethasone conjugated to a nanocarrier based on biocompatible N-(2-hydroxypropyl) methacrylamide copolymers. We observed an increased accumulation of dexamethasone polymer nanomedicines in the arthritic mouse paw using non-invasive fluorescent in vivo imaging and confirmed it by the analysis of tissue homogenates. The dexamethasone conjugate exhibited a dose-dependent healing effect on arthritis and an improved therapeutic outcome compared to free dexamethasone. Particularly, significant reduction of accumulation of RA mediator RANKL was observed. Overall, our data suggest that the conjugation of dexamethasone to a polymer nanocarrier by means of stimuli-sensitive spacer is suitable strategy for improving rheumatoid arthritis therapy.


Subject(s)
Arthritis, Rheumatoid , Dexamethasone , Polymers , Animals , Dexamethasone/chemistry , Dexamethasone/pharmacokinetics , Dexamethasone/administration & dosage , Dexamethasone/pharmacology , Dexamethasone/therapeutic use , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/pathology , Mice , Tissue Distribution , Polymers/chemistry , Polymers/pharmacokinetics , Arthritis, Experimental/drug therapy , Arthritis, Experimental/pathology , Nanoparticles/chemistry , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics
4.
Nanomedicine ; 57: 102744, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38460653

ABSTRACT

We recently prepared pH-responsive HPMA copolymer conjugates of bradykinin (P-BK), which release BK in response to the acidic tumor microenvironment, and found that administration of P-BK increased the tumor accumulation and therapeutic efficacy of nanomedicine. Because the release of BK from P-BK determines its onset of action, P-BKs with different release rates were prepared, and their properties were evaluated. The release kinetics were significantly altered by substitution proximal to hydrazone bond, release constant of methyl-substituted P-BK (P-MeBK) was approximately 4- and 80-fold higher than that of cyclopropyl-substituted P-BK (P-CPBK) and phenyl-substituted P-BK (P-PhBK). None of the P-BKs were active, but the release of BK restored their BK-like activity. Pre-administration of the P-BKs increased the tumor accumulation of nanomedicine in C26 tumor-bearing mice by 2- and 1.4-fold for P-MeBK and P-PhBK at 3 and 6 h. Altogether, this study provides insights into the design of pH-responsive nanodrugs with the desired release properties to target acidic lesions such as cancer and inflammation.


Subject(s)
Neoplasms , Polymers , Animals , Mice , Polymers/chemistry , Doxorubicin/chemistry , Bradykinin , Nanomedicine , Hydrogen-Ion Concentration , Neoplasms/drug therapy , Tumor Microenvironment
5.
Nanomedicine ; 56: 102730, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38158146

ABSTRACT

We synthesized three novel STAT3 inhibitors (S3iD1-S3iD3) possessing oxoheptanoic residue enabling linkage to HPMA copolymer carrier via a pH-sensitive hydrazone bond. HPMA copolymer conjugates bearing doxorubicin (Dox) and our STAT3 inhibitors were synthesized to evaluate the anticancer effect of Dox and STAT3 inhibitor co-delivery into tumors. S3iD1-3 and their copolymer-bound counterparts (P-S3iD1-P-S3iD3) showed considerable in vitro cytostatic activities in five mouse and human cancer cell lines with IC50 ~0.6-7.9 µM and 0.7-10.9 µM, respectively. S3iD2 and S3iD3 were confirmed to inhibit the STAT3 signaling pathway. The combination of HPMA copolymer-bound Dox (P-Dox) and P-S3iD3 at the dosage showing negligible toxicity demonstrated significant antitumor activity in B16F10 melanoma-bearing mice and completely cured 2 out of 15 mice. P-Dox alone had a significantly lower therapeutic activity with no completely cured mice. Thus, polymer conjugates bearing STAT3 inhibitors may be used for the chemosensitization of chemorefractory tumors.


Subject(s)
Doxorubicin , Methacrylates , Neoplasms , Mice , Humans , Animals , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Neoplasms/drug therapy , Polymethacrylic Acids , Hydrogen-Ion Concentration , STAT3 Transcription Factor/metabolism
6.
Mol Pharm ; 20(3): 1670-1680, 2023 03 06.
Article in English | MEDLINE | ID: mdl-36724294

ABSTRACT

Osteosarcoma (OS) is the most common form of primary malignant bone cancer in adolescents. Over the years, OS prognosis has greatly improved due to adjuvant and neoadjuvant (preoperative) chemotherapeutic treatment, increasing the chances of successful surgery and reducing the need for limb amputation. However, chemotherapeutic treatment to treat OS is limited by off-target toxicities and requires improved localization at the tumor site. Collagen, the main constituent of bone tissue, is extensively degraded and remodeled in OS, leading to an increased availability of denatured (monomeric) collagen. Collagen hybridizing peptides (CHPs) comprise a class of peptides rationally designed to selectively bind to denatured collagen. In this work, we have conjugated CHPs as targeting moieties to water-soluble N-(2-hydroxypropyl)methacrylamide (HPMA) copolymers to target OS tumors. We demonstrated increased accumulation of collagen-targeted HPMA copolymer-CHP conjugates compared to nontargeted HPMA copolymers, as well as increased retention compared to both nontargeted copolymers and CHPs, in a murine intratibial OS tumor model. Furthermore, we used microcomputed tomography analysis to evaluate the bone microarchitecture and correlated bone morphometric parameters (porosity, bone volume, and surface area) with maximum accumulation (Smax) and accumulation at 168 h postinjection (S168) of the copolymers at the tumor. Our results provide the foundation for the use of HPMA copolymer-CHP conjugates as targeted drug delivery systems in OS tumors.


Subject(s)
Bone Neoplasms , Osteosarcoma , Mice , Humans , Animals , Adolescent , X-Ray Microtomography , Drug Delivery Systems/methods , Methacrylates , Peptides , Collagen , Polymers
7.
Regul Toxicol Pharmacol ; 141: 105404, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37105297

ABSTRACT

Occupational asthma resulting from workplace exposure to chemical respiratory allergens is an important disease. No widely accepted or formally validated tests for the identification of chemical respiratory sensitizers. Consequently, there is a heavy reliance on human data from clinical examinations. Unfortunately, however, although such investigations are critical for the diagnosis of occupational asthma, and in guiding remedial actions, they do not reliably identify specific chemicals within the workplace that are the causative agents. There are several reasons for this, including the fact that specific inhalation tests conducted as part of clinical investigations are frequently performed with complex mixtures rather than single substances, that sometimes inhalation challenges are conducted at concentrations above the OEL and STEL, where effects may be confounded by irritation, and that involvement of immune mechanisms cannot be assumed from the observation of late asthmatic reactions. Further, caution should be taken when implicating substances on lists of "recognised" asthmagens unless they have undergone a formal weight of evidence assessment. Here the limitations of clinical investigations as currently performed for the purposes of regulatory classification and decision making are explored by reference to previously published case studies that implicate 2-hydroxyethylmethacrylate (HEMA) and/or 2-hydroxypropylmethacrylate (HPMA) as respiratory allergens.


Subject(s)
Asthma, Occupational , Occupational Exposure , Humans , Allergens/toxicity , Methacrylates/toxicity , Inflammation , Occupational Exposure/adverse effects
8.
Nanomedicine ; 48: 102636, 2023 02.
Article in English | MEDLINE | ID: mdl-36549553

ABSTRACT

In this study, we developed a nanoformulation of 5-aminolevulinic acid (5-ALA) for tumor-targeted photodynamic therapy, in which 5-ALA was conjugated with a biocompatible polymer N-(2-hydroxypropyl)methacrylamide (HPMA) through the hydrazone bond, i.e., P-ALA. P-ALA behaves as the nano-sized molecule with an average size of 5.5 nm in aqueous solution. P-ALA shows a largely increased release rate in acidic pH than physiological pH, suggesting the rapid release profile in acidic tumor environment. P-ALA did not show apparent cytotoxicity up to 0.1 mg/ml, however, under light irradiation, remarkable cell death was induced with the IC50 of 20-30 µg/ml. More importantly, we found significantly higher tumor accumulation of P-ALA than 5-ALA which benefit from its nano-size by taking advantage of the enhanced permeability and retention (EPR) effect. Consequently, P-ALA exhibited much improved in vivo antitumor efficacy without any apparent side effects. We thus anticipate the application of P-ALA as a nano-designed photosensitizer for anticancer photodynamic therapy.


Subject(s)
Antineoplastic Agents , Neoplasms , Photochemotherapy , Humans , Aminolevulinic Acid/pharmacology , Aminolevulinic Acid/therapeutic use , Antineoplastic Agents/pharmacology , Doxorubicin/pharmacology , Neoplasms/pathology , Polymers/chemistry , Cell Line, Tumor
9.
J Biol Chem ; 297(5): 101342, 2021 11.
Article in English | MEDLINE | ID: mdl-34710374

ABSTRACT

Peptide display methods are a powerful tool for discovering new ligands of pharmacologically relevant targets. However, the selected ligands often suffer from low affinity. Using phage display, we identified a new bicyclic peptide binder of prostate-specific membrane antigen (PSMA), a metalloprotease frequently overexpressed in prostate cancer. We show that linking multiple copies of a selected low-affinity peptide to a biocompatible water-soluble N-(2-hydroxypropyl)methacrylamide copolymer carrier (iBody) improved binding of the conjugate by several orders of magnitude. Furthermore, using ELISA, enzyme kinetics, confocal microscopy, and other approaches, we demonstrate that the resulting iBody can distinguish between different conformations of the target protein. The possibility to develop stable, fully synthetic, conformation-selective antibody mimetics has potential applications for molecular recognition, diagnosis and treatment of many pathologies. This strategy could significantly contribute to more effective drug discovery and design.


Subject(s)
Biomimetic Materials/chemistry , Drug Carriers/chemistry , Peptide Library , Humans , Kallikreins/chemistry , Prostate-Specific Antigen/chemistry
10.
Int J Mol Sci ; 22(11)2021 Jun 01.
Article in English | MEDLINE | ID: mdl-34206141

ABSTRACT

The interaction of multi-LacNAc (Galß1-4GlcNAc)-containing N-(2-hydroxypropyl) methacrylamide (HPMA) copolymers with human galectin-1 (Gal-1) and the carbohydrate recognition domain (CRD) of human galectin-3 (Gal-3) was analyzed using NMR methods in addition to cryo-electron-microscopy and dynamic light scattering (DLS) experiments. The interaction with individual LacNAc-containing components of the polymer was studied for comparison purposes. For Gal-3 CRD, the NMR data suggest a canonical interaction of the individual small-molecule bi- and trivalent ligands with the lectin binding site and better affinity for the trivalent arrangement due to statistical effects. For the glycopolymers, the interaction was stronger, although no evidence for forming a large supramolecule was obtained. In contrast, for Gal-1, the results indicate the formation of large cross-linked supramolecules in the presence of multivalent LacNAc entities for both the individual building blocks and the polymers. Interestingly, the bivalent and trivalent presentation of LacNAc in the polymer did not produce such an increase, indicating that the multivalency provided by the polymer is sufficient for triggering an efficient binding between the glycopolymer and Gal-1. This hypothesis was further demonstrated by electron microscopy and DLS methods.


Subject(s)
Blood Proteins/chemistry , Galectin 1/chemistry , Galectins/chemistry , Methacrylates/chemistry , Polymers/chemistry , Acrylamides/chemistry , Acrylamides/pharmacology , Binding Sites/drug effects , Blood Proteins/genetics , Carbohydrates/chemistry , Cryoelectron Microscopy , Galectin 1/genetics , Galectins/genetics , Humans , Ligands , Methacrylates/pharmacology , Polymers/pharmacology , Protein Binding/drug effects
11.
Int J Mol Sci ; 22(5)2021 Feb 28.
Article in English | MEDLINE | ID: mdl-33670859

ABSTRACT

Adenovirus-based gene transfer vectors are the most frequently used vector type in gene therapy clinical trials to date, and they play an important role as genetic vaccine candidates during the ongoing SARS-CoV-2 pandemic. Immediately upon delivery, adenovirus-based vectors exhibit multiple complex vector-host interactions and induce innate and adaptive immune responses. This can severely limit their safety and efficacy, particularly after delivery through the blood stream. In this review article we summarize two strategies to modulate Ad vector-induced immune responses: extensive genomic and chemical capsid modifications. Both strategies have shown beneficial effects in a number of preclinical studies while potential synergistic effects warrant further investigations.


Subject(s)
Adenoviridae/genetics , Adenoviridae/immunology , Capsid/immunology , Genetic Vectors/genetics , Genetic Vectors/immunology , Animals , COVID-19 , COVID-19 Vaccines/immunology , Capsid Proteins/genetics , Humans , Immunity , Immunogenicity, Vaccine , SARS-CoV-2/genetics , SARS-CoV-2/immunology
12.
Adv Funct Mater ; 30(12)2020 Mar 17.
Article in English | MEDLINE | ID: mdl-33071706

ABSTRACT

Checkpoint blockade immunotherapies harness the host's own immune system to fight cancer, but only work against tumors infiltrated by swarms of pre-existing T cells. Unfortunately, most cancers to date are immune-deserted. Here, we report a polymer-assisted combination of immunogenic chemotherapy and PD-L1 degradation for efficacious treatment in originally non-immunogenic cancer. "Priming" tumors with backbone-degradable polymer-epirubicin conjugates elicits immunogenic cell death and fosters tumor-specific CD8+ T cell response. Sequential treatment with a multivalent polymer-peptide antagonist to PD-L1 overcomes adaptive PD-L1 enrichment following chemotherapy, biases the recycling of PD-L1 to lysosome degradation via surface receptor crosslinking, and produces prolonged elimination of PD-L1 rather than the transient blocking afforded by standard anti-PD-L1 antibodies. Together, these findings established the polymer-facilitated tumor targeting of immunogenic drugs and surface crosslinking of PD-L1 as a potential new therapeutic strategy to propagate a long-term antitumor immunity, which might broaden the application of immunotherapy to immunosuppressive cancers.

13.
Pharm Res ; 37(11): 229, 2020 Oct 23.
Article in English | MEDLINE | ID: mdl-33098043

ABSTRACT

PURPOSE: The development of diagnostic and therapeutic agents utilizing small peptides (e.g., bombesin (BBN)) to target the overexpression of the gastrin-releasing peptide receptor (GRPR) in cancers has been widely investigated. Herein, we examine the capabilities of BBN-modified HPMA copolymers to target the GRPR. METHODS: Four positive, four negative, and two zwitterionic BBN HPMA copolymer conjugates of varying peptide content and charge were synthesized. In vitro and in vivo studies were conducted in a GRPR-overexpressing prostate cancer cell line (PC-3) and a normal CF-1 mouse model, respectively. RESULTS: Cellular uptake of the conjugates were found to be charge and BBN density dependent. The positively-charged conjugates illustrated a direct relationship between the extent of cellular internalization, ranging from 0.7 to 20%, and BBN-incorporation density. The negative and zwitterionic conjugates showed low PC-3 uptake values. Blocking studies confirmed the GRPR-targeting effect of the positively-charged constructs. In vivo studies of the positively-charged copolymers resulted in rapid blood clearance by the mononuclear phagocyte system (MPS)-associated tissues (e.g., liver and spleen). CONCLUSION: Positively-charged BBN-HPMA copolymer conjugates demonstrated good GRPR-targeting and internalization in vitro. However, the impact of peptide density and charge on in vivo MPS recognition are parameters that must be optimized in future agent development.


Subject(s)
Methacrylates/metabolism , Polymers/metabolism , Prostatic Neoplasms/metabolism , Receptors, Bombesin/metabolism , Tissue Distribution/physiology , Animals , Cell Line, Tumor , Female , Humans , Male , Mice , PC-3 Cells
14.
Int J Mol Sci ; 21(17)2020 Aug 21.
Article in English | MEDLINE | ID: mdl-32825790

ABSTRACT

Stimulus-sensitive polymer drug conjugates based on high molecular weight N-(2-hydroxypropyl) methacrylamide (HPMA) copolymers carrying doxorubicin via a pH-dependent cleavable bond (pHPMA-Dox) were previously shown to be able to overcome multi-drug resistance. Nevertheless, a tumor type dependent differential response was observed. Although an improved and more selective tumor accumulation of pHPMA-Dox is generally achieved due to the enhanced permeability and retention (EPR) effect, little is known about the fate of these conjugates upon entering the tumor tissue, which could explain the different responses. In this study, we compared in vitro and in vivo accumulation and Dox-activation of pHPMA-Dox in three cancer cell line models (1411HP, A2780cis, HT29) and derived xenograft tumors using a near-infrared fluorescence-labeled pHPMA-Dox conjugate. Firstly, cytotoxicity assays using different pH conditions proved a stepwise, pH-dependent increase in cytotoxic activity and revealed comparable sensitivity among the cell lines. Using multispectral fluorescence microscopy, we were able to track the distribution of drug and polymeric carrier simultaneously on cellular and histological levels. Microscopic analyses of cell monolayers confirmed the assumed mechanism of cell internalization of the whole conjugate followed by intracellular cleavage and nuclear accumulation of Dox in all three cell lines. In contrast, intratumoral distribution and drug release in xenograft tumors were completely different and were associated with different tissue substructures and microenvironments analyzed by Azan- and Hypoxisense®-staining. In 1411HP tumors, large vessels and less hypoxic/acidic microenvironments were associated with a pattern resulting from consistent tissue distribution and cellular uptake as whole conjugate followed by intracellular drug release. In A2780cis tumors, an inconsistent pattern of distribution partly resulting from premature drug release was associated with a more hypoxic/acidic microenvironment, compacted tumor tissue with compressed vessels and specific pre-damaged tissue structures. A completely different distribution pattern was observed in HT29 tumors, resulting from high accumulation of polymer in abundant fibrotic structures, with small embedded vessels featuring this tumor type together with pronounced premature drug release due to the strongly hypoxic/acidic microenvironment. In conclusion, the pattern of intratumoral distribution and drug release strongly depends on the tumor substructure and microenvironment and may result in different degrees of therapeutic efficacy. This reflects the pronounced heterogeneity observed in the clinical application of nanomedicines and can be exploited for the future design of such conjugates.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Doxorubicin/pharmacokinetics , Drug Carriers/pharmacokinetics , Animals , Antineoplastic Agents/administration & dosage , Carbocyanines/chemistry , Cell Line, Tumor , Doxorubicin/administration & dosage , Doxorubicin/chemistry , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Drug Delivery Systems , Drug Liberation , Fluorescent Dyes/chemistry , HT29 Cells , Humans , Hydrogen-Ion Concentration , Male , Methacrylates/chemistry , Mice, Nude , Molecular Weight , Tissue Distribution , Tumor Microenvironment , Xenograft Model Antitumor Assays
15.
Mol Pharm ; 16(8): 3452-3459, 2019 08 05.
Article in English | MEDLINE | ID: mdl-31294568

ABSTRACT

N-(2-Hydroxypropyl)methacrylamide copolymer conjugates of pirarubicin (THP), P-THP, accumulates selectively in solid tumor tissue by the enhanced permeability and retention (EPR) effect. Despite of high accumulation in solid tumors, some macromolecular antitumor agents show poor therapeutic outcome because of poor tissue diffusion into the tumor as well as obstructed tumor blood flow. Here, we confirmed that cellular uptake of P-THP was 25 times less than that of free THP at 1-4 h incubation time in vitro. The passage of P-THP through the confluent tight-monolayer cells junction was 12 times higher than free THP, and P-THP penetrated deeper into the tumor cell spheroid (1.3-1.7-fold) than free THP in 4 h. In addition, P-THP showed cytotoxicity comparable to that of free THP to tumor-cells in spheroid form, despite of 7 times lower cytotoxicity of P-THP to the monolayer cells to that of free THP in vitro. These results indicate that P-THP administration can exhibit deeper diffusion into the tumor cell spheroid than free THP. As a consequence, P-THP exhibits more efficient antitumor activity than free THP in vivo, which is also supported by better pharmacokinetics and tumor accumulation of P-THP than free THP.


Subject(s)
Acrylamides/chemistry , Antineoplastic Agents/administration & dosage , Doxorubicin/analogs & derivatives , Drug Carriers/chemistry , Neoplasms/drug therapy , Antineoplastic Agents/pharmacokinetics , Doxorubicin/administration & dosage , Doxorubicin/pharmacokinetics , Drug Screening Assays, Antitumor , HCT116 Cells , Humans , Neoplasms/pathology , Spheroids, Cellular
16.
Mol Pharm ; 16(3): 1234-1244, 2019 03 04.
Article in English | MEDLINE | ID: mdl-30702897

ABSTRACT

The addictive potential of clinically used opioids as a result of their direct action on the dopaminergic reward system in the brain has limited their application. In an attempt to reduce negative side effects as well as to improve the overall effectiveness of these analgesics, we have designed, synthesized, and evaluated an N-(2-hydroxypropyl)methacrylamide (HPMA)-based macromolecular prodrug of hydromorphone (HMP), a commonly used opioid. To this end, P-HMP was synthesized via RAFT polymerization and a subsequent polymer analogous reaction. Its interaction with inflammatory cells in arthritic joints was evaluated in vitro using a RAW 264.7 cell culture, and subsequent confocal microscopy analysis confirmed that P-HMP could be internalized by the cells via endocytosis. In vivo imaging studies indicated that the prodrug can passively target the arthritic joint after systemic administration in a rodent model of monoarticular adjuvant-induced arthritis (MAA). The inflammatory pain-alleviating properties of the prodrug were assessed in MAA rats using the incapacitance test and were observed to be similar to dose-equivalent HMP. Analgesia through mechanisms at the spinal cord level was further measured using the tail flick test, and it was determined that the prodrug significantly reduced spinal cord analgesia versus free HMP, further validating the peripheral restriction of the macromolecular prodrug. Immunohistochemical analysis of cellular uptake of the P-HMP within the MAA knee joint proved the internalization of the prodrug by phagocytic synoviocytes, colocalized with HMP's target receptor as well as with pain-modulating ion channels. Therefore, it can be concluded that the novel inflammation-targeting polymeric prodrug of HMP (P-HMP) has the potential to be developed as an effective and safe analgesic agent for musculoskeletal pain.


Subject(s)
Acrylamides/chemistry , Analgesics/therapeutic use , Arthritis, Experimental/drug therapy , Hydromorphone/chemistry , Pain/drug therapy , Polymers/therapeutic use , Prodrugs/therapeutic use , Analgesics, Opioid/adverse effects , Animals , Arthritis, Rheumatoid/drug therapy , Drug Discovery , Endocytosis , Male , Mice , Phagocytosis , Polymers/chemical synthesis , Polymers/metabolism , Prodrugs/chemical synthesis , Prodrugs/metabolism , RAW 264.7 Cells , Rats , Rats, Inbred Lew , Tissue Distribution , Treatment Outcome
17.
J Pept Sci ; 25(2): e3144, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30588703

ABSTRACT

Mitochondrion plays an important role in executing cell programmed death pathway. Therefore, drugs designed to target mitochondria are supposed to make superior contributions to cancer therapy. However, the problem that drugs or drug delivery systems being sequestrated in endosomes/lysosomes needs to be solved for effective drug delivery. Here, mitochondrial targeting and nonendocytic cell entry peptide SS20 modified HPMA copolymer (P-FITC-SS20) was synthesized. With SS20 peptide modification, the uptake behavior of HPMA copolymers changed remarkably compared with unmodified ones. The internalization of P-FITC-SS20 was not influenced by endocytic inhibitors and temperature. Further, the internalized copolymers were not trapped in endosomes/lysosomes. Although cellular uptake of HPMA copolymer was decreased after SS20 peptide modification, SS20 peptide significantly improved mitochondrial accumulation of HPMA copolymers due to its outstanding mitochondrial targeting ability. Moreover, owing to lower susceptibility to macrophagocyte in blood, P-SS20-Cy5 showed longer blood circulation time and enhanced tumor accumulation. The current study validated that SS20 peptide modification is a promising strategy for mitochondrial targeting drug delivery systems and can be further applied to mitochondria associated diseases to improve therapeutic efficacy.


Subject(s)
Endocytosis , Methacrylates/pharmacokinetics , Mitochondria/metabolism , Peptides/pharmacokinetics , Polymers/pharmacokinetics , Animals , Cells, Cultured , Endocytosis/drug effects , HeLa Cells , Humans , Methacrylates/chemical synthesis , Methacrylates/chemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Mitochondria/drug effects , Peptides/chemistry , Polymers/chemical synthesis , Polymers/chemistry , RAW 264.7 Cells , Tissue Distribution
18.
Mol Cell Neurosci ; 88: 70-82, 2018 04.
Article in English | MEDLINE | ID: mdl-29414104

ABSTRACT

Growing evidence suggests that oxidative stress plays a critical role in neuronal destruction characteristic of Parkinson's disease (PD). However, the molecular mechanisms of oxidative stress-mediated dopaminergic cell death are far from clear. In the current investigation, we tested the hypothesis that acrolein, an oxidative stress and lipid peroxidation (LPO) product, is a key factor in the pathogenesis of PD. Using a combination of in vitro, in vivo, and cell free models, coupled with anatomical, functional, and behavioral examination, we found that acrolein was elevated in 6-OHDA-injected rats, and behavioral deficits associated with 6-OHDA could be mitigated by the application of the acrolein scavenger hydralazine, and mimicked by injection of acrolein in healthy rats. Furthermore, hydralazine alleviated neuronal cell death elicited by 6-OHDA and another PD-related toxin, rotenone, in vitro. We also show that acrolein can promote the aggregation of alpha-synuclein, suggesting that alpha-synuclein self-assembly, a key pathological phenomenon in human PD, could play a role in neurotoxic effects of acrolein in PD models. These studies suggest that acrolein is involved in the pathogenesis of PD, and the administration of anti-acrolein scavengers such as hydralazine could represent a novel strategy to alleviate tissue damage and motor deficits associated with this disease.


Subject(s)
Acrolein/pharmacology , Cell Death/drug effects , Dopaminergic Neurons/drug effects , Parkinson Disease/metabolism , alpha-Synuclein/metabolism , Animals , Cell Line , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Oxidative Stress/drug effects , Parkinson Disease/drug therapy , Rats , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/metabolism , Substantia Nigra/drug effects , Substantia Nigra/metabolism
19.
Mol Pharm ; 15(9): 3654-3663, 2018 09 04.
Article in English | MEDLINE | ID: mdl-29543465

ABSTRACT

Herein, the biodegradable micelle-forming amphiphilic N-(2-hydroxypropyl) methacrylamide (HPMA)-based polymer conjugates with the anticancer drug doxorubicin (Dox) designed for enhanced tumor accumulation were investigated, and the influence of their stability in the bloodstream on biodistribution, namely, tumor uptake, and in vivo antitumor efficacy were evaluated in detail. Dox was attached to the polymer carrier by a hydrazone bond enabling pH-controlled drug release. While the polymer-drug conjugates were stable in a buffer at pH 7.4 (mimicking bloodstream environment), Dox was released in a buffer under mild acidic conditions modeling the tumor microenvironment or cells. The amphiphilic polymer carriers differed in the structure of hydrophobic cholesterol derivative moieties bound to the HPMA copolymers via a hydrolyzable hydrazone bond, exhibiting different rates of micellar structure disintegration at various pH values. Considerable dependence of the studied polymer-drug conjugate biodistribution on the stability of the micellar structure was observed in neutral, bloodstream-mimicking, environment, showing that a faster rate of the micelle disintegration in pH 7.4 increased the conjugate blood clearance, decreased tumor accumulation, and significantly reduced the tumor:blood and tumor:muscle ratios. Similarly, the final therapeutic outcome was strongly affected by the stability of the micellar structure because the most stable micellar conjugate showed an almost similar therapeutic outcome as the water-soluble, nondegradable, high-molecular-weight starlike HPMA copolymer-Dox conjugate, which was highly efficient in the treatment of solid tumors in mice. Based on the results, we conclude that the bloodstream stability of micellar polymer-anticancer drug conjugates, in addition to their low side toxicity, is a crucial parameter for their efficient solid tumor accumulation and high in vivo antitumor activity.


Subject(s)
Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Polymers/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Doxorubicin/therapeutic use , Drug Carriers/chemistry , Drug Liberation , Female , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Lymphoma/blood , Lymphoma/drug therapy , Mice , Mice, Inbred C57BL , Micelles
20.
Pharm Res ; 36(1): 19, 2018 Dec 03.
Article in English | MEDLINE | ID: mdl-30511238

ABSTRACT

PURPOSE: Tuberculosis (TB) chemotherapy witnesses some major challenges such as poor water-solubility and bioavailability of drugs that frequently delay the treatment. In the present study, an attempt to enhance the aqueous solubility of rifampicin (RMP) was made via co-polymeric nanoparticles approach. HPMA (N-2-hydroxypropylmethacrylamide)-PLGA based polymeric nanoparticulate system were prepared and evaluated against Mycobacterium tuberculosis (MTB) for sustained release and bioavailability of RMP to achieve better delivery. METHODOLOGY: HPMA-PLGA nanoparticles (HP-NPs) were prepared by modified nanoprecipitation technique, RMP was loaded in the prepared NPs. Characterization for particle size, zeta potential, and drug-loading capacity was performed. Release was studied using membrane dialysis method. RESULTS: The average particles size, zeta potential, polydispersity index of RMP loaded HPMA-PLGA-NPs (HPR-NPs) were 260.3 ± 2.21 nm, -6.63 ± 1.28 mV, and 0.303 ± 0.22, respectively. TEM images showed spherical shaped NPs with uniform distribution without any cluster formation. Entrapment efficiency and drug loading efficiency of HPR-NPs were found to be 76.25 ± 1.28%, and 26.19 ± 2.24%, respectively. Kinetic models of drug release including Higuchi and Korsmeyer-peppas demonstrated sustained release pattern. Interaction studies with human RBCs confirmed that RMP loaded HP-NPs are less toxic in this model than pure RMP with (p < 0.05). CONCLUSIONS: The pathogen inhibition studies revealed that developed HPR-NPs were approximately four times more effective with (p < 0.05) than pure drug against sensitive Mycobacterium tuberculosis (MTB) stain. It may be concluded that HPR-NPs holds promising potential for increasing solubility and bioavailability of RMP.


Subject(s)
Methacrylates/administration & dosage , Methacrylates/chemistry , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/administration & dosage , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Rifampin/administration & dosage , Rifampin/chemistry , Biological Availability , Drug Carriers , Drug Delivery Systems , Drug Liberation , Methacrylates/chemical synthesis , Microbial Sensitivity Tests , Mycobacterium tuberculosis/drug effects , Particle Size , Rifampin/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL