Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 193
Filter
1.
Drug Deliv Transl Res ; 14(8): 2062-2078, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38517568

ABSTRACT

To improve the therapeutic activity of inhaled glucocorticoids and reduce potential side effects, we designed a formulation combining the advantages of nanoparticles, which have an enhanced uptake by alveolar cells, allow targeted delivery and sustained drug release, as well as limited drug systemic passage, with those of microparticles, which display good alveolar deposition. Herein, a polymer-drug conjugate, poly(malic acid)-budesonide (PMAB), was first synthesized with either 11, 20, 33, or 43 mol% budesonide (drug:polymer from 1:8 to 3:4), the drug creating hydrophobic domains. The obtained conjugates self-assemble into nanoconjugates in water, yielding excellent drug loading of up to 73 wt%, with 80-100 nm diameters. In vitro assays showed that budesonide could be steadily released from the nanoconjugates, and the anti-inflammatory activity was preserved, as evidenced by reduced cytokine production in LPS-activated RAW 264.7 macrophages. Nanoconjugates were then embedded into microparticles through spray-drying with L-leucine, forming nano-embedded microparticles (NEMs). NEMs were produced with an aerodynamic diameter close to 1 µm and a density below 0.1 g.cm-3, indicative of a high alveolar deposition. NEMs spray-dried with the less hydrophobic nanoconjugates, PMAB 1:4, were readily dissolved in simulated lung fluid and were chosen for in vivo experiments to study pharmacokinetics in healthy rats. As it was released in vivo from NEMs, sustained distribution of budesonide was obtained for 48 h in lung tissue, cells, and lining fluid. With high loading rates, modulable release kinetics, and low cytotoxicity, these nanoconjugates delivered by NEMs are promising for the more efficient treatment of pulmonary inflammatory diseases.


Subject(s)
Budesonide , Lung , Nanoconjugates , Animals , Mice , Budesonide/administration & dosage , Budesonide/pharmacokinetics , Budesonide/chemistry , RAW 264.7 Cells , Lung/metabolism , Nanoconjugates/chemistry , Nanoconjugates/administration & dosage , Male , Polymers/chemistry , Polymers/administration & dosage , Malates/chemistry , Malates/administration & dosage , Malates/pharmacokinetics , Administration, Inhalation , Particle Size , Rats , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacokinetics , Drug Liberation , Rats, Sprague-Dawley
2.
BMC Cancer ; 21(1): 1176, 2021 Nov 03.
Article in English | MEDLINE | ID: mdl-34732161

ABSTRACT

BACKGROUND: According to the NAPOLI-1 trial, nanoliposomal irinotecan (nal-IRI) plus fluorouracil/folinic acid (5-FU/LV) showed improved overall survival compared to fluorouracil alone for patients with metastatic pancreatic cancer who were previously treated with gemcitabine-based therapy. In that trial, Asian patients had frequent dose modification due to haematological toxicity. There has been limited information on the clinical benefits and toxicity of this regimen in real-world settings. In this study, we assessed real-world experience of nal-IRI plus 5-FU/LV in patients with advanced pancreatic cancer after gemcitabine failure. METHODS: We conducted a single institution, retrospective analysis of response, survival and safety in patients who had been treated with nal-IRI with 5-FU/LV. Patients with metastatic pancreatic ductal adenocarcinoma previously treated with gemcitabine-based therapy received nal-IRI (80 mg/m2) with 5-FU/LV every 2 weeks. Kaplan-Meier analysis was performed to obtain median progression free survival and median overall survival. The hazard ratio and 95% confidence interval (CI) were estimated using a stratified Cox regression model. A multivariate Cox proportional hazards regression model was used to identify the effects of clinical factors. RESULTS: Fifty-one patients received nal-IRI plus 5-FU/LV between January 2015 and December 2020. The median age was 67 years, and males were 58.8%. A total of 40 (78.4%) and 11 (21.6%) patients had received one and two lines of prior chemotherapy before enrollment, respectively. Median progression-free survival was 2.8 months (95% CI 1.8-3.7) and median overall survival was 7.0 months (95% CI 6.0-7.9). Chemotherapy doses were reduced or delayed in 33 (64.7%) patients during the first 6 weeks and median relative dose intensity was 0.87. Thirty-six (70.6%) patients experienced grade 3 or 4 adverse events, most commonly neutropenia (58.8%). Most non-haematologic adverse events were under grade 2. Since the start of first-line chemotherapy, median overall survival was 16.3 months (95% CI 14.1-18.4). CONCLUSIONS: Nal-IRI plus 5-FU/LV seems to be effective, with manageable toxicities, following gemcitabine-based treatment in patients with metastatic pancreatic ductal adenocarcinoma. Nal-IRI plus 5-FU/LV following gemcitabine with nab-paclitaxel is a feasible sequential treatment option in patients with metastatic pancreatic cancer. TRIAL REGISTRATION: Retrospectively registered.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Carcinoma, Pancreatic Ductal/drug therapy , Fluorouracil/administration & dosage , Irinotecan/administration & dosage , Leucovorin/administration & dosage , Pancreatic Neoplasms/drug therapy , Aged , Albumins/therapeutic use , Antimetabolites, Antineoplastic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/secondary , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Drug Administration Schedule , Female , Fluorouracil/adverse effects , Humans , Irinotecan/adverse effects , Irinotecan/analogs & derivatives , Kaplan-Meier Estimate , Leucovorin/adverse effects , Liposomes , Male , Middle Aged , Nanoconjugates/administration & dosage , Nanoconjugates/adverse effects , Neutropenia/chemically induced , Paclitaxel/therapeutic use , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Progression-Free Survival , Proportional Hazards Models , Retrospective Studies , Gemcitabine
3.
Adv Sci (Weinh) ; 8(14): e2100549, 2021 07.
Article in English | MEDLINE | ID: mdl-34032027

ABSTRACT

Klebsiella pneumoniae has emerged as a severe opportunistic pathogen with multiple drug resistances. Finding effective vaccines against this pathogen is urgent. Although O-polysaccharides (OPS) of K. pneumoniae are suitable antigens for the preparation of vaccines given their low levels of diversity, the low immunogenicity (especially serotype O2) limit their application. In this study, a general Escherichia coli host system is developed to produce a nanoscale conjugate vaccine against K. pneumoniae using the Nano-B5 self-assembly platform. The experimental data illustrate that this nanoconjugate vaccine can induce an efficient humoral immune response in draining lymph nodes (dLNs) and elicit high titers of the IgG antibody against bacterial lipopolysaccharide (LPS). The ideal prophylactic effects of these nanoconjugate vaccines are further demonstrated in mouse models of both systemic and pulmonary infection. These results demonstrate that OPS with low immunogenicity can be changed into an effective antigen, indicating that other haptens may be applicable to this strategy in the future. To the knowledge, this is the first study to produce biosynthetic nanoconjugate vaccines against K. pneumoniae in E. coli, and this strategy can be applied to the development of other vaccines against pathogenic bacteria.


Subject(s)
Escherichia coli Infections/immunology , Klebsiella Infections/prevention & control , Klebsiella pneumoniae/immunology , Nanoconjugates/administration & dosage , Vaccines, Conjugate/administration & dosage , Animals , Disease Models, Animal , Female , Klebsiella Infections/immunology , Klebsiella pneumoniae/drug effects , Mice , Mice, Inbred BALB C , Vaccines, Conjugate/immunology
4.
Nanomedicine ; 34: 102376, 2021 06.
Article in English | MEDLINE | ID: mdl-33667725

ABSTRACT

Alcohol abuse induces the expression of inflammatory mediators by activating the immune receptors to trigger neuroinflammation and brain damage; however, therapies that reduce neuroimmune system activation may protect against alcohol's damaging effects. Curcuminoids possess anti-inflammatory properties but suffer from low bioavailability; therefore, we designed a new receptor-targeted biodegradable star-shaped crosslinked polypeptide polymer that bears propargylamine moieties and bisdemethoxycurcumin (StClPr-BDMC-ANG) as an enhanced anti-inflammatory therapeutic that penetrates the blood-brain-barrier and ameliorates alcohol-induced neuroinflammation. StClPr-BDMC-ANG administration maintains the viability of primary glia and inhibits the ethanol-induced upregulation of crucial inflammatory mediators in the prefrontal and medial cortex in a mouse model of chronic ethanol consumption. StClPr-BDMC-ANG treatment also suppresses the ethanol-mediated downregulation of microRNAs known to negatively modulate neuroinflammation in the brain cortex (miRs 146a-5p and let-7b-5p). In summary, our results demonstrate the attenuation of alcohol-induced neuroinflammation by an optimized and targeted polypeptide-based nanoconjugate of a curcuminoid.


Subject(s)
Alcohol Drinking/adverse effects , Curcumin/analogs & derivatives , Nanoconjugates/administration & dosage , Neuroinflammatory Diseases/drug therapy , Peptides/administration & dosage , Animals , Astrocytes/drug effects , Cells, Cultured , Curcumin/administration & dosage , Curcumin/chemistry , Mice , Nanoconjugates/chemistry , Neuroinflammatory Diseases/chemically induced , Peptides/chemistry
5.
Molecules ; 26(3)2021 Jan 22.
Article in English | MEDLINE | ID: mdl-33499047

ABSTRACT

Epidermal growth factor receptor (EGFR) is estimated to be overexpressed in 60~80% of colorectal cancer (CRC), which is associated with a poor prognosis. Anti-EGFR targeted monoclonal antibodies (cetuximab and panitumumab) have played an important role in the treatment of metastatic CRC. However, the therapeutic response of anti-EGFR monoclonal antibodies is limited due to multiple resistance mechanisms. With the discovery of new functions for gold nanoparticles (AuNPs), we hypothesize that cetuximab-conjugated AuNPs (cetuximab-AuNPs) will not only improve the cytotoxicity for cancer cells, but also introduce expression change of the related biomarkers on cancer cell surface. In this contribution, we investigated the size-dependent cytotoxicity of cetuximab-AuNPs to CRC cell line (HT-29), while also monitored the expression of cell surface biomarkers in response to treatment with cetuximab and cetuximab-AuNPs. AuNPs with the size of 60 nm showed the highest impact for cell cytotoxicity, which was tested by cell counting kit-8 (CCK-8) assay. Three cell surface biomarkers including epithelial cell adhesion molecule (EpCAM), melanoma cell adhesion molecule (MCAM), and human epidermal growth factor receptor-3 (HER-3) were found to be expressed at higher heterogeneity when cetuximab was conjugated to AuNPs. Both surface-enhanced Raman scattering/spectroscopy (SERS) and flow cytometry demonstrated the correlation of cell surface biomarkers in response to the drug treatment. We thus believe this study provides powerful potential for drug-conjugated AuNPs to enhance cancer prognosis and therapy.


Subject(s)
Antineoplastic Agents, Immunological/administration & dosage , Cetuximab/administration & dosage , Colorectal Neoplasms/drug therapy , Metal Nanoparticles/administration & dosage , Biomarkers, Tumor/metabolism , CD146 Antigen/metabolism , Cell Survival/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Epithelial Cell Adhesion Molecule/metabolism , Gold , HT29 Cells , Humans , Metal Nanoparticles/ultrastructure , Nanoconjugates/administration & dosage , Nanoconjugates/ultrastructure , Particle Size , Phenotype , Receptor, ErbB-3/metabolism , Signal Transduction/drug effects , Spectrum Analysis, Raman
6.
Cancer Res ; 81(3): 763-775, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33177062

ABSTRACT

New treatments are needed to address persistent unmet clinical needs for diffuse large B-cell lymphoma (DLBCL). Overexpression of transferrin receptor 1 (TFR1) is common across cancer and permits cell-surface targeting of specific therapies in preclinical and clinical studies of various solid tumors. Here, we developed novel nanocarrier delivery of chemotherapy via TFR1-mediated endocytosis, assessing this target for the first time in DLBCL. Analysis of published datasets showed novel association of increased TFR1 expression with high-risk DLBCL cases. Carbon-nitride dots (CND) are emerging nanoparticles with excellent in vivo stability and distribution and are adaptable to covalent conjugation with multiple substrates. In vitro, linking doxorubicin (Dox) and transferrin (TF) to CND (CND-Dox-TF, CDT) was 10-100 times more potent than Dox against DLBCL cell lines. Gain- and loss-of-function studies and fluorescent confocal microscopy confirmed dependence of these effects on TFR1-mediated endocytosis. In contrast with previous therapeutics directly linking Dox and TF, cytotoxicity of CDT resulted from nuclear entry by Dox, promoting double-stranded DNA breaks and apoptosis. CDT proved safe to administer in vivo, and when incorporated into standard frontline chemoimmunotherapy in place of Dox, it improved overall survival by controlling patient-derived xenograft tumors with greatly reduced host toxicities. Nanocarrier-mediated Dox delivery to cell-surface TFR1, therefore, warrants optimization as a potential new therapeutic option in DLBCL. SIGNIFICANCE: Targeted nanoparticle delivery of doxorubicin chemotherapy via the TRF1 receptor presents a new opportunity against high-risk DLBCL tumors using potency and precision.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Antigens, CD/metabolism , Doxorubicin/administration & dosage , Lymphoma, Large B-Cell, Diffuse/drug therapy , Nanoparticles/administration & dosage , Receptors, Transferrin/metabolism , Transferrin/administration & dosage , Animals , Antibiotics, Antineoplastic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis , Cell Line, Tumor , Cell Nucleus , Cell Survival/drug effects , Cyclophosphamide/administration & dosage , Cyclophosphamide/pharmacology , DNA Breaks, Double-Stranded , Doxorubicin/pharmacology , Endocytosis , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/mortality , Male , Mice , Mice, Inbred NOD , Mice, SCID , Nanoconjugates/administration & dosage , Prednisone/administration & dosage , Prednisone/pharmacology , Rituximab/administration & dosage , Rituximab/pharmacology , Transferrin/pharmacology , Vincristine/administration & dosage , Vincristine/pharmacology
7.
Int J Nanomedicine ; 15: 7013-7034, 2020.
Article in English | MEDLINE | ID: mdl-33061365

ABSTRACT

PURPOSE: Gastric cancer stem cells (CSCs) are important for the initiation, growth, recurrence, and metastasis of gastric cancer, due to their chemo-resistance and indefinite proliferation. Herein, to eliminate gastric CSCs, we developed novel CSC-targeting glioma-associated oncogene homolog 1 (Gli1) small interfering RNA (siRNA) nanoparticles that are specifically guided by a di-stearoyl-phosphatidyl-ethanolamine- hyaluronic-acid (DSPE-HA) single-point conjugate, as an intrinsic ligand of the CD44 receptor. We refer to these as targeting Gli1 siRNA nanoparticles. METHODS: We used the reductive amination reaction method for attaching amine groups of DSPE to aldehydic group of hyaluronic acid (HA) at the reducing end, to synthesize the DSPE-HA single-point conjugate. Next, targeting Gli1 siRNA nanoparticles were prepared using the layer-by-layer assembly method. We characterized the stem cellular features of targeting Gli1 siRNA nanoparticles, including their targeting efficiency, self-renewal capacity, the migration and invasion capacity of gastric CSCs, and the penetration ability of 3D tumor spheroids. Next, we evaluated the therapeutic efficacy of the targeting Gli1 siRNA nanoparticles by using in vivo relapsed tumor models of gastric CSCs. RESULTS: Compared with the multipoint conjugates, DSPE-HA single-point conjugates on the surface of nanoparticles showed significantly higher binding affinities with CD44. The targeting Gli1 siRNA nanoparticles significantly decreased Gli1 protein expression, inhibited CSC tumor spheroid and colony formation, and suppressed cell migration and invasion. Furthermore, in vivo imaging demonstrated that targeting Gli1 siRNA nanoparticles accumulated in tumor tissues, showing significant antitumor recurrence efficacy in vivo. CONCLUSION: In summary, our targeting Gli1 siRNA nanoparticles significantly inhibited CSC malignancy features by specifically blocking Hedgehog (Hh) signaling both in vitro and in vivo, suggesting that this novel siRNA delivery system that specifically eliminates gastric CSCs provides a promising targeted therapeutic strategy for gastric cancer treatment.


Subject(s)
Hyaluronan Receptors/genetics , Nanoparticles/chemistry , Neoplastic Stem Cells/drug effects , RNA, Small Interfering/administration & dosage , Stomach Neoplasms/drug therapy , Zinc Finger Protein GLI1/genetics , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Genetic Therapy/methods , Hedgehog Proteins/metabolism , Humans , Hyaluronic Acid/chemistry , Male , Mice, Inbred BALB C , Molecular Targeted Therapy/methods , Nanoconjugates/administration & dosage , Nanoconjugates/chemistry , Nanoparticles/administration & dosage , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Phosphatidylethanolamines/chemistry , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Xenograft Model Antitumor Assays
8.
Int J Nanomedicine ; 15: 6649-6658, 2020.
Article in English | MEDLINE | ID: mdl-32982224

ABSTRACT

BACKGROUND: Nanotechnology is gaining emerging interest in advanced drug discovery therapeutics due to their tremendous properties including enhanced delivery of therapeutic payload, extensive surface to volume ratio, high permeability, retention behaviors, etc. The gold nanoparticles (AuNPs) are favored due to their advanced features, such as biogenic, tunable physiochemical response, ease in synthesis, and wide range of biomedical applications. The phytochemicals have been focused to design Au nano-carrier-based conjugation for active-targeting drug delivery due to their nano conjugation ability. AIM: The present study describes the facile synthesis of 20nm spherical AuNPs and their conjugation with reported anti-cancer phytocompound Withanolide-A (1). METHODS: The 20nm sAuNPs were synthesized chemically and characterized their phytochemical gold nanoconjugates through UV-visible spectroscopy, dynamic light scattering (DLS) and transmission electron microscopy (TEM) imaging techniques. The anti-cancer therapeutic potentials were tested with both nanoconjugates and pure WithanolideA (1) by using SKBR3 breast cancer cells line. RESULTS: The synthesized sAuNPs showed significant conjugation with Withanolide-A and showed stability. Furthermore, these Au nanoconjugates with Withanolide-A (1) significantly induce blockage of SKBR3 cell growth at half maximal active concentration that compared to pure Withanolide-A (1). CONCLUSION: Our findings provide a foundation to further progress how they can overcome cancer drug resistance by conjugating active drugs in combination with AuNPs through optimizing the effective drug concentration and removing the surface barrier.


Subject(s)
Breast Neoplasms/drug therapy , Metal Nanoparticles/chemistry , Nanoconjugates/chemistry , Withanolides/pharmacology , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Delivery Systems , Drug Stability , Dynamic Light Scattering , Female , Gold/chemistry , Humans , Metal Nanoparticles/administration & dosage , Microscopy, Electron, Transmission , Nanoconjugates/administration & dosage , Withanolides/administration & dosage , Withanolides/chemistry
9.
J Med Chem ; 63(13): 7410-7421, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32524814

ABSTRACT

This paper emphasizes the synthesis of novel hybrid drug nanoparticles (Hyb-D-AuNPs) based on gold-temozolomide (TMZ) complexes combined with gemcitabine (GEM) and decitabine (DAC) to improve the efficiency and reduce the resistance of U87 malignant glial cells against TMZ. All products were evaluated by several spectroscopic techniques (Raman, UV-Vis) and transmission electron microscopy (TEM). Besides, for therapeutic purposes, the effect of these nanoparticles on cell proliferation and toxicity was evaluated, which clearly showed a synergic action of TMZ and GEM. Through the analysis of the exometabolome by nuclear magnetic resonance (NMR), the metabolic changes in the culture medium were measured in glial cells. Moreover, these nanoparticles are especially appropriated to the thermal destruction of cancer in the case of photothermal therapy due to their photothermal heating properties. This study presents an original chemical approach that it could play a central role in the field of nanomedicine, with novel perspectives for the development of new drugs and active targeting in glioblastoma multiforme (GBM) cancer therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Decitabine/pharmacology , Deoxycytidine/analogs & derivatives , Glioblastoma/drug therapy , Nanoconjugates/administration & dosage , Temozolomide/pharmacology , Antineoplastic Combined Chemotherapy Protocols/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Decitabine/administration & dosage , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacology , Drug Delivery Systems , Drug Liberation , Drug Resistance, Neoplasm , Drug Synergism , Glioblastoma/metabolism , Glioblastoma/pathology , Gold/chemistry , Humans , Magnetic Resonance Spectroscopy , Metal Nanoparticles/chemistry , Nanoconjugates/chemistry , Proof of Concept Study , Spectrophotometry, Ultraviolet , Temozolomide/administration & dosage , Gemcitabine
10.
J Hematol Oncol ; 13(1): 36, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32295630

ABSTRACT

BACKGROUND: Current acute myeloid leukemia (AML) therapy fails to eliminate quiescent leukemic blasts in the bone marrow, leading to about 50% of patient relapse by increasing AML burden in the bone marrow, blood, and extramedullar sites. We developed a protein-based nanoparticle conjugated to the potent antimitotic agent Auristatin E that selectively targets AML blasts because of their CXCR4 receptor overexpression (CXCR4+) as compared to normal cells. The therapeutic rationale is based on the involvement of CXCR4 overexpression in leukemic blast homing and quiescence in the bone marrow, and the association of these leukemic stem cells with minimal residual disease, dissemination, chemotherapy resistance, and lower patient survival. METHODS: Monomethyl Auristatin E (MMAE) was conjugated with the CXCR4 targeted protein nanoparticle T22-GFP-H6 produced in E. coli. Nanoconjugate internalization and in vitro cell viability assays were performed in CXCR4+ AML cell lines to analyze the specific antineoplastic activity through the CXCR4 receptor. In addition, a disseminated AML animal model was used to evaluate the anticancer effect of T22-GFP-H6-Auristatin in immunosuppressed NSG mice (n = 10/group). U of Mann-Whitney test was used to consider if differences were significant between groups. RESULTS: T22-GFP-H6-Auristatin was capable to internalize and exert antineoplastic effects through the CXCR4 receptor in THP-1 and SKM-1 CXCR4+ AML cell lines. In addition, repeated administration of the T22-GFP-H6-Auristatin nanoconjugate (9 doses daily) achieves a potent antineoplastic activity by internalizing specifically in the leukemic cells (luminescent THP-1) to selectively eliminate them. This leads to reduced involvement of leukemic cells in the bone marrow, peripheral blood, liver, and spleen, while avoiding toxicity in normal tissues in a luminescent disseminated AML mouse model. CONCLUSIONS: A novel nanoconjugate for targeted drug delivery of Auristatin reduces significantly the acute myeloid leukemic cell burden in the bone marrow and blood and blocks its dissemination to extramedullar organs in a CXCR4+ AML model. This selective drug delivery approach validates CXCR4+ AML cells as a target for clinical therapy, not only promising to improve the control of leukemic dissemination but also dramatically reducing the severe toxicity of classical AML therapy.


Subject(s)
Aminobenzoates/therapeutic use , Antineoplastic Agents/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Nanoconjugates/therapeutic use , Oligopeptides/therapeutic use , Receptors, CXCR4/metabolism , Aminobenzoates/administration & dosage , Animals , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Drug Delivery Systems , Female , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred NOD , Nanoconjugates/administration & dosage , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , Oligopeptides/administration & dosage
11.
Int J Pharm ; 582: 119345, 2020 May 30.
Article in English | MEDLINE | ID: mdl-32311470

ABSTRACT

Despite the great advances accomplished in the treatment of pediatric cancers, recurrences and metastases still exacerbate prognosis in some aggressive solid tumors such as neuroblastoma and osteosarcoma. In view of the poor efficacy and toxicity of current chemotherapeutic treatments, we propose a single multitherapeutic nanotechnology-based strategy by co-assembling in the same nanodevice two amphiphilic antitumor agents: squalenoyl-gemcitabine and edelfosine. Homogeneous batches of nanoassemblies were easily formulated by the nanoprecipitation method. Their anticancer activity was tested in pediatric cancer cell lines and pharmacokinetic studies were performed in mice. In vitro assays revealed a synergistic effect when gemcitabine was co-administered with edelfosine. Squalenoyl-gemcitabine/edelfosine nanoassemblies were found to be capable of intracellular translocation in patient-derived metastatic pediatric osteosarcoma cells and showed a better antitumor profile than squalenoyl-gemcitabine nanoassemblies alone. The intravenous administration of this combinatorial nanomedicine in mice exhibited a controlled release behavior of gemcitabine and diminished edelfosine plasma peak concentrations. These findings make it a suitable pre-clinical candidate for childhood cancer therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Bone Neoplasms/drug therapy , Nanoconjugates/therapeutic use , Nanoparticles , Neuroblastoma/drug therapy , Osteosarcoma/drug therapy , Phospholipid Ethers/pharmacology , Squalene/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/chemistry , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Compounding , Drug Synergism , Female , Inhibitory Concentration 50 , Injections, Intravenous , Mice, Nude , Nanoconjugates/administration & dosage , Nanoconjugates/chemistry , Neuroblastoma/metabolism , Neuroblastoma/pathology , Osteosarcoma/metabolism , Osteosarcoma/pathology , Phospholipid Ethers/administration & dosage , Phospholipid Ethers/chemistry , Phospholipid Ethers/pharmacokinetics , Squalene/administration & dosage , Squalene/chemistry , Squalene/pharmacokinetics , Squalene/therapeutic use
12.
Pak J Pharm Sci ; 33(2): 705-714, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32276917

ABSTRACT

Human diseases like viral organisms for example, hepatitis, HIV and etc., attack the health and caused large mortality in populations by many years. So finding novel delivery vehicles based antiviral drugs employing nano-materials is of high universal interest. In current approach a very biocompatible biodegradable nano-biopolymer anionic linear globular dendrimer second generation G2 was elaborately conjugated to a well-known anti-HIV drug Azidovudine and thereafter was characterized by different analytical techniques like AFM, Zeta sizer, 1HNMR, FTIR and LC-Mass spectroscopy. Then, Anionic Linear Globular DendrimerG2-Zidovudine Nano-Conjugate was assessed on human normal cells (toxicity assay by XTT test) and also HIV cell model and the results showed that Anionic Linear Globular DendrimerG2-Zidovudine Nano-Conjugate Significantly Decreased Retroviral Activity without any human cell toxicity respectively. Based on current experimental data such nano-compositions is proposed for further in vivo anti-HIV assays as well.


Subject(s)
Anti-Retroviral Agents/administration & dosage , Cell Survival/drug effects , Dendrimers/administration & dosage , Drug Delivery Systems/methods , Nanoconjugates/administration & dosage , Zidovudine/administration & dosage , Anions , Anti-Retroviral Agents/chemistry , Cell Survival/physiology , Dendrimers/chemistry , Dose-Response Relationship, Drug , HEK293 Cells , HIV-1/drug effects , HIV-1/physiology , Humans , Nanoconjugates/chemistry , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/chemistry , Stearates/administration & dosage , Stearates/chemistry , Zidovudine/chemistry
13.
Theranostics ; 10(7): 3240-3253, 2020.
Article in English | MEDLINE | ID: mdl-32194865

ABSTRACT

Rationale: Of the regulatory microRNAs expressed in the wounded skin, microRNA-21 (miR21) plays a pivotal role in wound repair by stimulating re-epithelialization, an essential feature to facilitate healing and reduce scar formation. Despite their crucial roles in wound healing, synthetic exogenous microRNAs have limited applications owing to the lack of an appropriate delivery system. Herein, we designed an miR21 mimic nanocarrier system using facial amphipathic bile acid-conjugated polyethyleneimines (BA-PEI) for the intracellular and transdermal delivery of synthetic miR21 molecules to accelerate wound repair. Methods: To design miR21 mimic nanocarriers, BA-conjugated PEIs prepared from three different types of BA at molar feed ratios of 1 and 3 were synthesized. The intracellular uptake efficiency of synthetic miR21 mimics was studied using confocal laser scanning microscopy and flow cytometry analysis. The optimized miR21/BA nanocarrier system was used to evaluate the wound healing effects induced by miR21 mimics in human HaCaT keratinocytes in vitro and a murine excisional acute wound model in vivo. Results: The cell uptake efficiency of miR21 complexed with BA-conjugated PEI was dramatically higher than that of miR21 complexed with PEI alone. Deoxycholic acid (DA)-modified PEI at a molar feed ratio of 3:1 (DA3-PEI) showed the highest transfection efficiency for miR21 without any increase in toxicity. After effective transdermal and intracellular delivery of miR21/DA3 nanocarriers, miR21 mimics promoted cell migration and proliferation through the post-transcriptional regulation of programmed cell death protein 4 (PDCD4) and matrix metalloproteinases. Thus, miR21 mimic nanocarriers improved both the rate and quality of wound healing, as evident from enhanced collagen synthesis and accelerated wound re-epithelialization. Conclusion: Our miRNA nanocarrier systems developed using DA3-PEI conjugates may be potentially useful for the delivery of synthetic exogenous miRNAs in various fields.


Subject(s)
Bile Acids and Salts/administration & dosage , Drug Carriers/administration & dosage , MicroRNAs/administration & dosage , Nanoconjugates/administration & dosage , Polyethyleneimine/administration & dosage , Skin/injuries , Wound Healing/drug effects , Administration, Cutaneous , Animals , Bile Acids and Salts/chemistry , Bone Morphogenetic Proteins/antagonists & inhibitors , Bone Morphogenetic Proteins/metabolism , Cell Division/drug effects , Cell Line , Cell Movement/drug effects , Drug Design , Drug Liberation , Gene Expression Profiling , Humans , Hydrophobic and Hydrophilic Interactions , Keratinocytes , Male , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred BALB C , MicroRNAs/therapeutic use , Molecular Mimicry , Signal Transduction/drug effects , Skin Absorption
14.
Int J Nanomedicine ; 15: 991-1003, 2020.
Article in English | MEDLINE | ID: mdl-32103952

ABSTRACT

INTRODUCTION AND OBJECTIVE: Pancreatic cancer (PC) is characterized by a robust desmoplastic environment, which limits the uptake of the standard first-line chemotherapeutic drug gemcitabine. Enhancing gemcitabine delivery to the complex tumor microenvironment (TME) is a major clinical challenge. Molecular crosstalk between pancreatic cancer cells (PCCs) and pancreatic stellate cells (PSCs) plays a critical role in desmoplastic reaction in PCs. Herein, we report the development of a targeted drug delivery system to inhibit the proliferation of PCCs and PSCs in vitro. Using gold nanoparticles as the delivery vehicle, the anti-EGFR antibody cetuximab (C225/C) as a targeting agent, gemcitabine as drug and polyethylene glycol (PEG) as a stealth molecule, we created a series of targeted drug delivery systems. METHODS: Fabricated nanoconjugates were characterized by various physicochemical techniques such as UV-Visible spectroscopy, transmission electron microscopy, HPLC and instrumental neutron activation analysis (INAA). RESULTS AND CONCLUSION: Targeted gemcitabine delivery systems containing mPEG-SH having molecular weights of 550 Da or 1000 Da demonstrated superior efficacy in reducing the viability of both PCCs and PSCs as compared to their non-targeted counterparts. EGFR-targeted pathway was further validated by pre-treating cells with C225 followed by determining cellular viability. Taken together, in our current study we have developed a PEGylated targeted nanoconjugate ACG44P1000 that showed enhanced selectivity towards pancreatic cancer cells and pancreatic stellate cells, among others, for gemcitabine delivery. We will investigate the ability of these optimized conjugates to inhibit desmoplasia and tumor growth in vivo in our future studies.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems/methods , Nanoconjugates/chemistry , Pancreatic Neoplasms/drug therapy , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Cetuximab/administration & dosage , Cetuximab/chemistry , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Deoxycytidine/chemistry , Drug Carriers/chemistry , Gold/chemistry , Humans , Metal Nanoparticles/administration & dosage , Metal Nanoparticles/chemistry , Metal Nanoparticles/toxicity , Nanoconjugates/administration & dosage , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/pathology , Polyethylene Glycols/chemistry , Tumor Microenvironment/drug effects , Gemcitabine
15.
Nano Lett ; 19(11): 7573-7587, 2019 11 13.
Article in English | MEDLINE | ID: mdl-31518145

ABSTRACT

Despite untiring efforts to develop therapies for pancreatic ductal adenocarcinoma (PDAC), survival statistics remain dismal, necessitating distinct approaches. Photodynamic priming (PDP), which improves drug delivery and combination regimens, as well as tumor photodestruction are key attributes of photodynamic therapy (PDT), making it a distinctive clinical option for PDAC. Localized, high-payload nanomedicine-assisted delivery of photosensitizers (PSs), with molecular specificity and controlled photoactivation, thus becomes critical in order to reduce collateral toxicity during more expansive photodynamic activation procedures with curative intent. As such, targeted photoactivable lipid-based nanomedicines are an ideal candidate but have failed to provide greater than two-fold cancer cell selectivity, if at all, due to their extensive multivariant physical, optical, and chemical complexity. Here, we report (1) a systematic multivariant tuning approach to engineer (Cet, anti-EGFR mAb) photoimmunonanoconjugates (PINs), and (2) stroma-rich heterotypic PDAC in vitro and in vivo models incorporating patient-derived pancreatic cancer-associated fibroblasts (PCAFs) that recapitulate the desmoplasia observed in the clinic. These offer a comprehensive, disease-specific framework for the development of Cet-PINs. Specificity-tuning of the PINs, in terms of PS lipid anchoring, electrostatic modulation, Cet orientation, and Cet surface densities, achieved ∼16-fold binding specificities and rapid penetration of the heterotypic organoids within 1 h, thereby providing a ∼16-fold enhancement in molecular targeted NIR photodestruction. As a demonstration of their inherent amenability for multifunctionality, encapsulation of high payloads of gemcitabine hydrochloride, 5-fluorouracil, and oxaliplatin within the Cet-PINs further improved their antitumor efficacy in the heterotypic organoids. In heterotypic desmoplastic tumors, the Cet-PINs efficiently penetrated up to 470 µm away from blood vessels, and photodynamic activation resulted in substantial tumor necrosis, which was not elicited in T47D tumors (low EGFR) or when using untargeted constructs in both tumor types. Photodynamic activation of the Cet-PINs in the heterotypic desmoplastic tumors resulted in collagen photomodulation, with a 1.5-fold reduction in collagen density, suggesting that PDP may also hold potential for conquering desmoplasia. The in vivo safety profile of photodynamic activation of the Cet-PINs was also substantially improved, as compared to the untargeted constructs. While treatment using the Cet-PINs did not cause any detriment to the mice's health or to healthy proximal tissue, photodynamic activation of untargeted constructs induced severe acute cachexia and weight loss in all treated mice, with substantial peripheral skin necrosis, muscle necrosis, and bowel perforation. This study is the first report demonstrating the true value of molecular targeting for NIR-activable PINs. These constructs integrate high payload delivery, efficient photodestruction, molecular precision, and collagen photomodulation in desmoplastic PDAC tumors in a single treatment using a single construct. Such combined PIN platforms and heterocellular models open up an array of further multiplexed combination therapies to synergistically control desmoplastic tumor progression and extend PDAC patient survival.


Subject(s)
Carcinoma, Pancreatic Ductal/drug therapy , Immunoconjugates/therapeutic use , Nanoconjugates/therapeutic use , Pancreatic Neoplasms/drug therapy , Photosensitizing Agents/therapeutic use , Animals , Antibodies, Monoclonal/therapeutic use , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/pathology , Carcinoma, Pancreatic Ductal/pathology , Drug Delivery Systems/methods , ErbB Receptors/antagonists & inhibitors , Humans , Immunoconjugates/administration & dosage , Mice , Nanoconjugates/administration & dosage , Nanomedicine/methods , Organoids/drug effects , Organoids/pathology , Pancreatic Neoplasms/pathology , Photochemotherapy/methods , Photosensitizing Agents/administration & dosage
16.
Nano Lett ; 19(8): 5515-5523, 2019 08 14.
Article in English | MEDLINE | ID: mdl-31362507

ABSTRACT

Designing simple-structured nanomedicine without lacking key functionalities, thereby avoiding incomplete damage or relapse of tumor with the administration of a safe dose, is pivotal for successful cancer nanotherapy. We herein presented a nanomedicine of photodynamic therapy (PDT) that simply assembled amphiphilic macromolecules of poly-l-lysine conjugating with photosensitizers onto hydrophobic upconverting nanoparticles. We demonstrated that the nanoformulation, despite its simple structure and synthesis, simultaneously possesses multiple features, including substantial payload of photosensitizers, avid cellular internalization both in vitro and in vivo, efficient diffusion and broad distribution in tumor lesion, and potent fatality for cancer stem cells that are refractory to other therapy modalities. Because of the combination of these functionalities, the tumors in mice were eradicated and no relapse was observed after at least 40 days, just with an extremely low intraperitoneal injection dose of 5.6 mg/kg. Our results suggested a strategy for designing multifunctional nanomedicines with simple construct and efficacious therapeutic response and presented the promising potential of PDT for a radical cure of cancer.


Subject(s)
Nanoconjugates/therapeutic use , Neoplasms/drug therapy , Photosensitizing Agents/therapeutic use , Animals , Cell Line, Tumor , HeLa Cells , Humans , Mice , Nanoconjugates/administration & dosage , Nanoconjugates/chemistry , Neoplasm Recurrence, Local/prevention & control , Neoplastic Stem Cells/drug effects , Photochemotherapy , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/chemistry , Polylysine/administration & dosage , Polylysine/analogs & derivatives , Polylysine/therapeutic use
17.
Drug Dev Ind Pharm ; 45(9): 1496-1505, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31241372

ABSTRACT

Nanomaterial based anticancer treatment is promising nowadays because of their small size that can penetrate and interact both inside and outside the cell surface. In this study, a simple protocol was followed for the conjugation of the biologically synthesized selenium nanoparticles (SeNPs) and short chain synthetic peptide. SeNPs was synthesized by using the culture supernatant of Streptomyces griseoruber, actinomycetes isolated from the soil. The short chain peptide Boc-L-F-OMe was synthesized by the conventional solution phase chemistry using a racemization-free fragment condensation strategy. Peptide interaction with different anticancer receptors was preliminarily studied by docking studies. Biosynthesized SeNPs was conjugated with short chain synthetic peptides by means of cysteine conjugation. Characterization of SeNPs with peptide was done by UV-visible spectroscopy and DLS that showed the red shift in the peak and increase in average particle size and zeta potential, respectively. Bioconjugated SeNPs- peptide was tested for its cytotoxicity against the colon cancer cell line HT-29. Bioconjugated SeNPs-peptide showed enhanced cytotoxic activity when compared to the peptide and nanoparticle alone that was tested at 10-50 µg/ml concentration. Further apoptotic studies were done by AO/PI staining and DNA fragmentation assay that confirms the cytotoxicity of the conjugates. Novel peptide-SeNPs conjugates tested in our study has a significant anticancer activity that can be potentially used for targeting the cancer cells.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/administration & dosage , Colorectal Neoplasms/drug therapy , Nanoconjugates/administration & dosage , Adenocarcinoma/pathology , Antineoplastic Agents/chemistry , Colorectal Neoplasms/pathology , Dipeptides/administration & dosage , Drug Evaluation, Preclinical , HT29 Cells , Humans , Molecular Docking Simulation , Nanoconjugates/chemistry , Selenium
18.
ACS Appl Mater Interfaces ; 11(18): 16336-16346, 2019 May 08.
Article in English | MEDLINE | ID: mdl-30986026

ABSTRACT

Gold nanoparticles (AuNPs) have emerged as promising drug delivery candidates that can be leveraged for cancer therapy. Lung cancer (LC) is a heterogeneous disease that imposes a significant burden on society, with an unmet need for new therapies. Chemotherapeutic drugs such as afatinib (Afb), which is clinically approved for the treatment of epidermal growth factor receptor positive LC, is hydrophobic and has low bioavailability leading to spread around the body, causing severe side effects. Herein, we present a novel afatinib-AuNP formulation termed Afb-AuNPs, with the aim of improving drug efficacy and biocompatibility. This was achieved by synthesis of an alkyne-bearing Afb derivative and reaction with azide-functionalized lipoic acid using copper-catalyzed click chemistry, then conjugation to AuNPs via alkylthiol-gold bond formation. The Afb-AuNPs were found to possess up to 3.7-fold increased potency when administered to LC cells in vitro and were capable of significantly inhibiting cancer cell proliferation, as assessed by MTT assay and electric cell-substrate impedance sensing, respectively. Furthermore, when exposed to Afb-AuNPs, human alveolar epithelial type I-like cells, a model of the healthy lung epithelium, maintained viability and were found to release less proinflammatory cytokines when compared to free drug, demonstrating the biocompatibility of our formulation. This study provides a new platform for the development of nontraditional AuNP conjugates which can be applied to other molecules of therapeutic or diagnostic utility, with potential to be combined with photothermal therapy in other cancers.


Subject(s)
Afatinib/chemistry , Carcinoma, Non-Small-Cell Lung/drug therapy , Nanoconjugates/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Afatinib/administration & dosage , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Delivery Systems , Humans , Materials Testing , Metal Nanoparticles/administration & dosage , Metal Nanoparticles/chemistry , Nanoconjugates/chemistry , Polyethylene Glycols/chemistry , Protein Kinase Inhibitors/chemistry , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/chemistry
19.
J Spinal Cord Med ; 42(6): 725-734, 2019 11.
Article in English | MEDLINE | ID: mdl-30843479

ABSTRACT

Context: Following a spinal cord hemisection at the second cervical segment the ipsilateral hemidiaphragm is paralyzed due to the disruption of the rostral ventral respiratory group (rVRG) axons descending to the ipsilateral phrenic motoneurons (PN). Systemically administered theophylline activates a functionally latent crossed phrenic pathway (CPP) which decussates caudal to the hemisection and activates phrenic motoneurons ipsilateral to the hemisection. The result is return of function to the paralyzed hemidiaphragm. Unfortunately, in humans, systemically administered theophylline at a therapeutic dose produces many unwanted side effects.Design and setting: A tripartite nanoconjugate was synthesized in which theophylline was coupled to a neuronal tracer, wheat germ agglutinin conjugated to horseradish peroxidase (WGA-HRP), using gold nanoparticles as the coupler. Following intradiaphragmatic injection of the nanoconjugate, WGA-HRP selectively targets the theophylline-bound nanoconjugate to phrenic motoneurons initially, followed by neurons in the rVRG by retrograde transsynaptic transport.Participants: (N/A)Interventions: (N/A)Outcome Measures: Immunostaining, Electromyography (EMG).Results: Delivery of the theophylline-coupled nanoconjugate to the nuclei involved in respiration induces a return of respiratory activity as detected by EMG of the diaphragm and a modest return of phrenic nerve activity.Conclusion: In addition to the modest return of phrenic nerve activity, there were many difficulties using the theophylline nanoconjugate because of its chemical instability, which suggests that the theophylline nanoconjugate should not be developed for clinical use as explained herein.


Subject(s)
Cervical Cord/injuries , Diaphragm/drug effects , Diaphragm/innervation , Metal Nanoparticles/administration & dosage , Nanoconjugates/administration & dosage , Recovery of Function/drug effects , Spinal Cord Injuries/drug therapy , Animals , Disease Models, Animal , Electromyography , Gold , Medulla Oblongata/drug effects , Motor Neurons/drug effects , Phrenic Nerve/drug effects , Pyramidal Tracts/drug effects , Rats , Theophylline/administration & dosage
20.
Nano Lett ; 19(3): 1719-1727, 2019 03 13.
Article in English | MEDLINE | ID: mdl-30724087

ABSTRACT

Protein nanocages are promising multifunctional platforms for nanomedicine owing to the ability to decorate their surfaces with multiple functionalities through genetic and/or chemical modification to achieve desired properties for therapeutic and diagnostic purposes. Here, we describe a model antigen (OVA peptide) that was conjugated to the surface of a naturally occurring hepatitis B core protein nanocage (HBc NC) by genetic modification. The engineered OVA-HBc nanocages (OVA-HBc NCs), displaying high density repetitive array of epitopes in a limited space by self-assembling into symmetrical structure, not only can induce bone marrow derived dendritic cells (BMDC) maturation effectively but also can be enriched in the draining lymph nodes. Naïve C57BL/6 mice immunized with OVA-HBc NCs are able to generate significant and specific cytotoxic T lymphocyte (CTL) responses. Moreover, OVA-HBc NCs as a robust nanovaccine can trigger preventive antitumor immunity and significantly delay tumor growth. When combined with a low-dose chemotherapy drug (paclitaxel), OVA-HBc NCs could specifically inhibit progression of an established tumor. Our findings support HBc-based nanocages with modularity and scalability as an attractive nanoplatform for combination cancer immunotherapy.


Subject(s)
Antigens, Neoplasm/administration & dosage , Hepatitis B Core Antigens/immunology , Nanoconjugates/administration & dosage , Neoplasms/therapy , Animals , Antigens, Neoplasm/immunology , Bioengineering/methods , Cell Proliferation/drug effects , Dendritic Cells/drug effects , Dendritic Cells/immunology , Epitopes/genetics , Epitopes/immunology , Hepatitis B/immunology , Hepatitis B Core Antigens/administration & dosage , Humans , Immunotherapy/methods , Mice, Inbred C57BL , Neoplasms/immunology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL