Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
Microb Cell Fact ; 22(1): 143, 2023 Aug 03.
Article in English | MEDLINE | ID: mdl-37533061

ABSTRACT

Fungal producing potency of camptothecin (CPT) raise the hope for their usage to be a platform for industrial production of CPT, nevertheless, attenuation of their productivity of CPT with the subculturing and preservation is the challenge. So, screening for novel endophytic fungal isolates with a reliable CPT-biosynthetic stability was the objective. Among the isolated endophytic fungi from the tested medicinal plants, Aspergillus terreus OQ642314.1, endophyte of Cinnamomum camphora, exhibits the highest yield of CPT (89.4 µg/l). From the NMR, FT-IR and LC-MS/MS analyses, the extracted CPT from A. terreus gave the same structure and molecular mass fragmentation pattern of authentic CPT (349 m/z). The putative CPT had a significant activity against MCF7 (0.27 µM) and HEPG-2 (0.8 µM), with a strong affinity to inhibits the human Topoisomerase 1 activity (IC50 0.362 µg/ml) as revealed from the Gel-based DNA relaxation assay. The purified CPT displayed a strong antimicrobial activity for various bacterial (E. coli and B. cereus) and fungal (A. flavus and A. parasiticus) isolates, ensuring the unique tertiary, and stereo-structure of A. terreus for penetrating the microbial cell walls and targeting the topoisomerase I. The higher dual activity of the purified CPT as antimicrobial and antitumor, emphasize their therapeutic efficiency, especially with growth of the opportunistic microorganisms due to the suppression of human immune system with the CPT uses in vivo. The putative CPT had an obvious activity against the tumor cell (MCF7) metastasis, and migration as revealed from the wound healing assay. The overall yield of A. terreus CPT was maximized with the Blackett-Burman design by twofolds increment (164.8 µg/l). The CPT yield by A. terreus was successively diminished with the multiple fungal subculturing, otherwise, the CPT productivity of A. terreus was restored, and increased over the zero culture upon coculturing with C. camphora microbiome (1.5% w/v), ensuring the restoring of CPT biosynthetic potency of A. terreus by the plant microbiome-derived chemical signals "microbial communication". This is the first report exploring the feasibility of A. terreus "endophyte of C. camphora" to be a preliminary platform for commercial production of CPT with a reliable sustainability upon uses of indigenous C. camphora microbiome.


Subject(s)
Anti-Infective Agents , Cinnamomum camphora , Microbiota , Humans , Endophytes/chemistry , Chromatography, Liquid , Escherichia coli , Spectroscopy, Fourier Transform Infrared , Tandem Mass Spectrometry , Camptothecin/pharmacology , Camptothecin/chemistry
2.
Planta Med ; 89(13): 1250-1258, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37044129

ABSTRACT

Camptothecin (CPT) and its derivatives have attracted worldwide attention because of their notable anticancer activity. However, the growing demand for CPT in the global pharmaceutical industry has caused a severe shortage of CPT-producing plant resources. In this study, phytochemical analysis of Nothapodytes tomentosa results in the isolation and identification of CPT (13: ) and 16 analogues (1:  - 12, 14:  - 17: ), including a new (1: ) and five known (9, 10, 12, 15: , and 17: ) CPT analogues with an open E-ring. In view of the potential anticancer activity of CPT analogues with an open E-ring, the fragmentation pathways and mass spectra profiles of these six CPT analogues (1, 9, 10, 12, 15: , and 17: ) are investigated, providing a reference for the rapid detection of these compounds in other plants. Furthermore, based on the fragmentation patterns of CPT (13: ) and known analogues (2:  - 8, 11, 14, 16, 18:  - 26: ), the distribution and content of these compounds in different tissues of N. tomentosa, N. nimmoniana, Camptotheca acuminata, and Ophiorrhiza japonica are further studied. Our findings not only provide an alternative plant resource for further expanding the development and utilization of CPT and its analogues, but also lay a foundation for improving the utilization of known CPT-producing plant resources.


Subject(s)
Antineoplastic Agents, Phytogenic , Camptotheca , Magnoliopsida , Camptothecin/chemistry , Camptothecin/metabolism , Antineoplastic Agents, Phytogenic/chemistry , Magnoliopsida/chemistry , Camptotheca/chemistry , Camptotheca/metabolism
3.
Int J Nanomedicine ; 18: 1-16, 2023.
Article in English | MEDLINE | ID: mdl-36632237

ABSTRACT

Introduction: Combination therapy is a promising approach to promote the efficacy and reduce the systemic toxicity of cancer therapy. Herein, we examined the potency of a combined chemo-phototherapy approach by constructing a hyaluronidase- and reactive oxygen species-responsive hyaluronic acid nanoparticle carrying a chemotherapy drug and a photosensitizer in a tumor-bearing mouse model. We hypothesized that following decomposition, the drugs inside the nanocomplex will be released in the tumors to provide effective tumor treatment. We aimed to design a smart drug delivery system that can improve traditional chemotherapy drug delivery and enhance the therapeutic efficacy in combination with photodynamic therapy. Methods: Hydrophilic hyaluronic acid (HA) was covalently modified with a hydrophobic 5ß-cholanic acid (CA) via an ROS-cleavable thioketal (tk) linker for a targeted co-deliver of 10-Hydroxy camptothecin (HCPT) and Chlorin e6 (Ce6) into tumors to improve the efficiency of combined chemo-photodynamic therapy. Results: The obtained HA-tk-CA nanoparticle carrying HCPT and Ce6, named HTCC, accumulated in the tumor through the enhanced permeable response (EPR) effect and HA-mediated CD44 targeting after intravenous administration. Upon laser irradiation and hyaluronidase degradation, HTCC was disrupted to release HCPT and Ce6 into the tumors. Compared to the monotherapy approach, HTCC demonstrated enhanced tumor growth inhibition and minimized systemic toxicity in a tumor-bearing mouse model. Conclusion: Our results suggested that controlled dual-drug release not only improved tumor drug delivery efficacy, but also reduced systemic side effects. In addition to HCPT and Ce6 delivery, the HA-tk-CA nanocomplex can be used to deliver other drugs in synergistic cancer therapy. Since most current combined therapy uses free drugs with distinct spatiotemporal distributions, the simultaneous co-delivery of dual drugs with a remote on-demand drug delivery nanosystem provides an alternative strategy for drug delivery design.


Subject(s)
Drug Delivery Systems , Nanoparticles , Neoplasms , Photochemotherapy , Photosensitizing Agents , Porphyrins , Animals , Mice , Camptothecin/chemistry , Cell Line, Tumor , Drug Delivery Systems/methods , Hyaluronic Acid/chemistry , Hyaluronoglucosaminidase , Nanoparticles/chemistry , Neoplasms/drug therapy , Photochemotherapy/methods , Photosensitizing Agents/administration & dosage , Porphyrins/chemistry , Reactive Oxygen Species
4.
J Colloid Interface Sci ; 630(Pt A): 385-394, 2023 Jan 15.
Article in English | MEDLINE | ID: mdl-36265340

ABSTRACT

Design of nanovectors inspired by nature is a short cut to improve the efficacy and bioavailability of chemotherapeutic agents, while reduce the toxicity. In this work, strongly hydrophobic camptothecin (CPT) was modified with different chain length of fatty acid (C4, C9 and C18) to synthesize CPT4C, CPT9C and CPT18C, respectively. CPT4C, CPT9C and CPT18C could complex with human serum albumin (HSA) readily to prepare CPT4C-HSA, CPT9C-HSA and CPT18C-HSA nanoagents. In vitro MTT assays demonstrated CPT18C-HSA possessed the highest cell killing capacity, due to the elevated cellular uptake that resulted from albumin-mediated transportation. In vivo tumor inhibition experiments verified that CPT18C-HSA had the most remarkable antitumor efficiency with distinctly lowered physiological toxicity. It could be used in large doses without obvious side effects. We believe this albumin-mediated transportation mode has great potential for efficient delivery of hydrophobic and/or physiologically unstable drugs.


Subject(s)
Antineoplastic Agents , Camptothecin , Humans , Camptothecin/pharmacology , Camptothecin/chemistry , Biomimetics , Fatty Acids , Serum Albumin, Human , Antineoplastic Agents/pharmacology
5.
Biomaterials ; 290: 121814, 2022 11.
Article in English | MEDLINE | ID: mdl-36194953

ABSTRACT

Drug-carrier compatibility impacts drug delivery efficiency and resulting therapeutic efficacy and tolerability. Although numerous biodegradable carrier materials have been pursued over the past decades, chemical strategies that are sought to tailor therapeutic structures and their carriers together in a concerted effort remain rare yet may be powerful. Based on the principle of improving the structural similarity between these central components, we developed an omega-3 fatty acid-conjugated poly(ethylene glycol) (PEG) nanocarrier host that is capable of supramolecular assembly of a cytotoxic prodrug guest. To demonstrate the proof of concept, we ligated two docosahexaenoic acid (DHA) molecules and one PEG chain via a d-lysine linkage to produce an amphiphilic matrix DHA2-PEG, which is suited for the encapsulation of active compounds, including a DHA monoconjugated camptothecin prodrug. The resulting DHA2-PEG-cloaked nanoassemblies show superior stability and rapid cellular uptake compared with those formulated in clinically approved materials. In a chemically induced mouse model of colitis-associated colorectal cancer, administration of the camptothecin nanoassemblies demonstrated notable inhibition of colon tumor growth. Furthermore, this new delivery platform has low systemic toxicity and immunotoxicity in animals and is appealing for further investigation and clinical translation. Thus, through rational engineering of the carrier biomaterials and drug derivatization, the in vivo performance of drug delivery systems can be improved. This approach also establishes a methodology for leveraging synthetic chemistry tools to optimize delivery systems for a broad range of drug classes.


Subject(s)
Colorectal Neoplasms , Fatty Acids, Omega-3 , Nanoparticles , Prodrugs , Mice , Animals , Prodrugs/chemistry , Nanoparticles/chemistry , Drug Delivery Systems/methods , Drug Carriers/chemistry , Camptothecin/pharmacology , Camptothecin/therapeutic use , Camptothecin/chemistry , Polyethylene Glycols/chemistry , Colorectal Neoplasms/drug therapy
6.
ACS Appl Mater Interfaces ; 14(36): 40546-40558, 2022 Sep 14.
Article in English | MEDLINE | ID: mdl-36059107

ABSTRACT

Hypoxia-activated prodrugs (HAPs) have drawn increasing attention for improving the antitumor effects while minimizing side effects. However, the heterogeneous distribution of the hypoxic region in tumors severely impedes the curative effect of HAPs. Additionally, most HAPs are not amenable to optical imaging, and it is difficult to precisely trace them in tissues. Herein, we carefully designed and synthesized a multifunctional therapeutic BAC prodrug by connecting the chemotherapeutic drug camptothecin (CPT) and the fluorescent photothermal agent boron dipyrromethene (BODIPY) via hypoxia-responsive azobenzene linkers. To enhance the solubility and tumor accumulation, the prepared BAC was further encapsulated into a human serum albumin (HSA)-based drug delivery system to form HSA@BAC nanoparticles. Since the CPT was caged by a BODIPY-based molecule at the active site, the BAC exhibited excellent biosafety. Importantly, the activated CPT could be quickly released from BAC and could perform chemotherapy in hypoxic cancer cells, which was ascribed to the cleavage of the azobenzene linker by overexpressed azoreductase. After irradiation with a 730 nm laser, HSA@BAC can efficiently generate hyperthermia to achieve irreversible cancer cell death by oxygen-independent photothermal therapy. Under fluorescence imaging-guided local irradiation, both in vitro and in vivo studies demonstrated that HSA@BAC exhibited superior antitumor effects with minimal side effects.


Subject(s)
Hyperthermia, Induced , Nanoparticles , Neoplasms , Prodrugs , Azo Compounds , Boron , Boron Compounds , Camptothecin/chemistry , Cell Line, Tumor , Humans , Hypoxia , Nanoparticles/chemistry , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Phototherapy , Photothermal Therapy , Porphobilinogen/analogs & derivatives , Prodrugs/chemistry
7.
J Nat Med ; 76(4): 748-755, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35902551

ABSTRACT

Ophiorrhiza plants (Family Rubiaceae) are known to produce diverse monoterpenoid indole alkaloids including camptothecin with potent antitumor activity. This review contains a summary of recent chemical studies reported over the past 10 years regarding alkaloids (monoterpenoid indole and tetrahydroisoquinoline alkaloids, and cyclopeptide) in Ophiorrhiza plants. In addition, the alkaloid biosynthetic pathways based on their reported structures were proposed.


Subject(s)
Alkaloids , Rubiaceae , Alkaloids/chemistry , Biosynthetic Pathways , Camptothecin/chemistry , Camptothecin/metabolism , Rubiaceae/chemistry , Rubiaceae/metabolism
8.
Mol Pharm ; 19(7): 2518-2534, 2022 07 04.
Article in English | MEDLINE | ID: mdl-35549267

ABSTRACT

Limited chemotherapeutic efficiency, drug resistance, and side effects are primary obstacles for cancer treatment. The development of co-delivery systems with synergistic treatment modes should be a promising strategy. Here, we fabricated a multifunctionalized nanocarrier with a combination of chemotherapeutic agents and gold nanoparticles (AuNPs), which could integrate chemo-photothermal therapy, thus enhancing overall anticancer efficacy, sensitizing drug-resistant cancer cells, and diminishing cancer stem cells (CSCs). To be specific, camptothecin nanocrystals (CPT NCs) were prepared as a platform, on the surface of which AuNPs were decorated and a hyaluronic acid layer acted as capping, stabilizing, targeting, and hydrophilic agents for CPT NCs, and reducing agents for AuNPs, providing a bridge connecting AuNPs to CPT. These AuNP-decorated CPT NCs exhibited good physico-chemical properties such as optimal sizes, payload, stability, and photothermal efficiency. Compared to other CPT formulations, they displayed considerably improved biocompatibility, selectivity, intracellular uptake, cytotoxicity, apoptosis induction activity, Pgp inhibitory capability, and anti-CSC activity, owing to a synergistic/cooperative effect from AuNPs, CPT, near-infrared treatment, pH/photothermal-triggered drug release, and nanoscaled structure. A mitochondrial-mediated signaling pathway is the underlying mechanism for cytotoxic and apoptotic effects from AuNP-decorated CPT NCs, in terms of mitochondrial dysfunction, intensified oxidative stress, DNA fragmentation, caspase 3 activation, upregulation of proapoptotic genes such as p53, Bax, and caspase 3, and lower levels of antiapoptotic Bcl-2.


Subject(s)
Antineoplastic Agents , Metal Nanoparticles , Nanoparticles , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Camptothecin/chemistry , Camptothecin/pharmacology , Caspase 3 , Cell Line, Tumor , Drug Delivery Systems , Drug Liberation , Drug Resistance , Gold/chemistry , Metal Nanoparticles/chemistry , Nanoparticles/chemistry , Phototherapy , Photothermal Therapy
9.
Int J Mol Sci ; 22(22)2021 Nov 13.
Article in English | MEDLINE | ID: mdl-34830168

ABSTRACT

Cancer cells are able to proliferate in an unregulated manner. There are several mechanisms involved that propel such neoplastic transformations. One of these processes involves bypassing cell death through changes in gene expression and, consequently, cell growth. This involves a complex epigenetic interaction within the cell, which drives it towards oncogenic transformations. These epigenetic events augment cellular growth by potentially altering chromatin structures and influencing key gene expressions. Therapeutic mechanisms have been developed to combat this by taking advantage of the underlying oncogenic mechanisms through chemical modulation. Camptothecin (CPT) is an example of this type of drug. It is a selective topoisomerase I inhibitor that is effective against many cancers, such as colorectal cancer. Previously, we successfully formulated a magnetic nanocarrier-conjugated CPT with ß-cyclodextrin and iron NPs (Fe3O4) cross-linked using EDTA (CPT-CEF). Compared to CPT alone, it boasts higher efficacy due to its selective targeting and increased solubility. In this study, we treated HT29 colon cancer cells with CPT-CEF and attempted to investigate the cytotoxic effects of the formulation through an epigenetic perspective. By using RNA-Seq, several differentially expressed genes were obtained (p < 0.05). Enrichr was then used for the over-representation analysis, and the genes were compared to the epigenetic roadmap and histone modification database. The results showed that the DEGs had a high correlation with epigenetic modifications involving histone H3 acetylation. Furthermore, a subset of these genes was shown to be associated with the Wnt/ß-catenin signaling pathway, which is highly upregulated in a large number of cancer cells. These genes could be investigated as downstream therapeutic targets against the uncontrolled proliferation of cancer cells. Further interaction analysis of the identified genes with the key genes of the Wnt/ß-catenin signaling pathway in colorectal cancer identified the direct interactors and a few transcription regulators. Further analysis in cBioPortal confirmed their genetic alterations and their distribution across patient samples. Thus, the findings of this study reveal that colorectal cancer could be reversed by treatment with the CPT-CEF nanoparticle-conjugated nanocarrier through an epigenetic mechanism.


Subject(s)
Camptothecin , Colorectal Neoplasms , Genes, Neoplasm , Histones , Nanocapsules , Neoplasm Proteins , Wnt Signaling Pathway/drug effects , Camptothecin/chemistry , Camptothecin/pharmacology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , HT29 Cells , Histones/genetics , Histones/metabolism , Humans , Nanocapsules/chemistry , Nanocapsules/therapeutic use , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism
10.
J Nanobiotechnology ; 19(1): 350, 2021 Oct 30.
Article in English | MEDLINE | ID: mdl-34717646

ABSTRACT

BACKGROUND: Nanoscale drug delivery systems have emerged as broadly applicable approach for chemo-photothermal therapy. However, these nanoscale drug delivery systems suffer from carrier-induced toxicity, uncontrolled drug release and low drug carrying capacity issues. Thus, to develop carrier-free nanoparticles self-assembled from amphiphilic drug molecules, containing photothermal agent and anticancer drug, are very attractive. RESULTS: In this study, we conjugated camptothecin (CPT) with a photothermal agent new indocyanine green (IR820) via a redox-responsive disulfide linker. The resulting amphiphilic drug-drug conjugate (IR820-SS-CPT) can self-assemble into nanoparticles (IR820-SS-CPT NPs) in aqueous solution, thus remarkably improving the membrane permeability of IR820 and the aqueous solubility of CPT. The disulfide bond in the IR820-SS-CPT NPs could be cleaved in GSH rich tumor microenvironment, leading to the on demand release of the conjugated drug. Importantly, the IR820-SS-CPT NPs displayed an extremely high therapeutic agent loading efficiency (approaching 100%). Besides, in vitro experimental results indicated that IR820-SS-CPT NPs displayed remarkable tumor cell killing efficiency. Especially, the IR820-SS-CPT NPs exhibited excellent anti-tumor effects in vivo. Both in vitro and in vivo experiments were conducted, which have indicated that the design of IR820-SS-CPT NPs can provide an efficient nanotherapeutics for chemo-photothermal therapy. CONCLUSION: A novel activatable amphiphilic small molecular prodrug IR820-SS-CPT has been developed in this study, which integrated multiple advantages of GSH-triggered drug release, high therapeutic agent content, and combined chemo-photothermal therapy into one drug delivery system.


Subject(s)
Camptothecin/administration & dosage , Camptothecin/chemistry , Drug Delivery Systems/methods , Nanoparticles/chemistry , Photothermal Therapy/methods , Prodrugs/chemistry , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Liberation , Female , Humans , Indocyanine Green , Mice , Mice, Inbred BALB C , Mice, Nude , Phototherapy , Solubility
11.
Biomolecules ; 11(8)2021 07 27.
Article in English | MEDLINE | ID: mdl-34439774

ABSTRACT

The interaction of immune checkpoint molecules in the tumor microenvironment reduces the anti-tumor immune response by suppressing the recognition of T cells to tumor cells. Immune checkpoint inhibitor (ICI) therapy is emerging as a promising therapeutic option for cancer treatment. However, modulating the immune system with ICIs still faces obstacles with severe immunogenic side effects and a lack of response against many cancer types. Plant-derived natural compounds offer regulation on various signaling cascades and have been applied for the treatment of multiple diseases, including cancer. Accumulated evidence provides the possibility of efficacy of phytochemicals in combinational with other therapeutic agents of ICIs, effectively modulating immune checkpoint-related signaling molecules. Recently, several phytochemicals have been reported to show the modulatory effects of immune checkpoints in various cancers in in vivo or in vitro models. This review summarizes druggable immune checkpoints and their regulatory factors. In addition, phytochemicals that are capable of suppressing PD-1/PD-L1 binding, the best-studied target of ICI therapy, were comprehensively summarized and classified according to chemical structure subgroups. It may help extend further research on phytochemicals as candidates of combinational adjuvants. Future clinical trials may validate the synergetic effects of preclinically investigated phytochemicals with ICI therapy.


Subject(s)
Immune Checkpoint Inhibitors/metabolism , Neoplasms/drug therapy , Neoplasms/immunology , Phytochemicals/chemistry , Programmed Cell Death 1 Receptor/metabolism , Animals , Antigens, CD/metabolism , Antineoplastic Agents/pharmacology , B7 Antigens/metabolism , B7-H1 Antigen/metabolism , CTLA-4 Antigen/metabolism , Camptothecin/chemistry , Diterpenes/chemistry , Epoxy Compounds/chemistry , Flavonoids/chemistry , Hepatitis A Virus Cellular Receptor 2/metabolism , Humans , Immunotherapy , Isothiocyanates/chemistry , Mice , Phenanthrenes/chemistry , Phytochemicals/pharmacology , Plant Extracts/pharmacology , Receptors, Immunologic/metabolism , Saponins/chemistry , Sulfoxides/chemistry , Terpenes/chemistry , Tumor Microenvironment/drug effects , Lymphocyte Activation Gene 3 Protein
12.
Carbohydr Polym ; 269: 118345, 2021 Oct 01.
Article in English | MEDLINE | ID: mdl-34294352

ABSTRACT

This work reports novel chitosan functionalized graphene oxide (GO) nanocomposites combined fluorescence imaging and therapeutic functions in one agent, which can serve as a promising alternative to alleviate related diseases caused hyperinflammation. Briefly, GO was designed to be conjugated with chitosan, fluorescein-labeled peptide, toll-like receptor 4 antibody and hydroxycamptothecin/aloe emodin. We have demonstrated that such nanocomposites could effectively achieve active targeted delivery of pro-apoptotic and anti-inflammatory drugs into inflammatory cells and cause cells apoptosis by acid-responsive drug release. Moreover, confocal fluorescence imaging confirms that the drug-induced inflammatory cells apoptosis could be visualized the light-up fluorescence of fluorescein activated by caspase-3. Meanwhile, inflammatory-related biomarkers have down-regulated after the nanocomposites' treatment in both vitro and vivo experiments consistent with the results in histological sections. In summary, the bifunctional nanocomposites that possess anti-inflammation and fluorescence imaging could serve as a promising therapeutic agent for reducing hyperinflammation caused by numerous diseases.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Apoptosis/physiology , Drug Carriers/chemistry , Inflammation/drug therapy , Nanocomposites/chemistry , Animals , Anti-Inflammatory Agents/chemistry , Antibodies/immunology , Camptothecin/analogs & derivatives , Camptothecin/chemistry , Camptothecin/therapeutic use , Cattle , Cell Line , Chitosan/chemistry , Drug Liberation , Emodin/chemistry , Emodin/therapeutic use , Fluorescent Dyes/chemistry , Graphite/chemistry , Humans , Lipopolysaccharides , Mammary Glands, Human/drug effects , Mammary Glands, Human/pathology , Mastitis/chemically induced , Mastitis/drug therapy , Mastitis/pathology , Mice , Toll-Like Receptor 4/immunology
13.
ACS Appl Mater Interfaces ; 13(28): 32690-32702, 2021 Jul 21.
Article in English | MEDLINE | ID: mdl-34229434

ABSTRACT

The synergistic nanotheranostics of reactive oxygen species (ROS) augment or phototherapy has been a promising method within synergistic oncotherapy. However, it is still hindered by sophisticated design and fabrication, lack of a multimodal synergistic effect, and hypoxia-associated poor photodynamic therapy (PDT) efficacy. Herein, a kind of porous shuttle-shape platinum (IV) methylene blue (Mb) coordination polymer nanotheranostics-loaded 10-hydroxycamptothecin (CPT) is fabricated to address the abovementioned limitations. Our nanoreactors possess spatiotemporally controlled O2 self-supply, self-sufficient singlet oxygen (1O2), and outstanding photothermal effect. Once they are taken up by tumor cells, nanoreactors as a cascade catalyst can efficiently catalyze degradation of the endogenous hydrogen peroxide (H2O2) into O2 to alleviate tumor hypoxia. The production of O2 can ensure enhanced PDT. Subsequently, under both stimuli of external red light irradiation and internal lysosomal acidity, nanoreactors can achieve the on-demand release of CPT to augment in situ mitochondrial ROS and highly efficient tumor ablation via phototherapy. Moreover, under the guidance of near-infrared (NIR) fluorescent imaging, our nanoreactors exhibit strongly synergistic potency for treatment of hypoxic tumors while reducing damages against normal tissues and organs. Collectively, shuttle-shape platinum-coordinated nanoreactors with augmented ROS capacity and enhanced phototherapy efficiency can be regarded as a novel tumor theranostic agent and further promote the research of synergistic oncotherapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Camptothecin/analogs & derivatives , Drug Carriers/chemistry , Nanostructures/chemistry , Neoplasms/drug therapy , Tumor Hypoxia/drug effects , Animals , Antineoplastic Agents/chemistry , Camptothecin/chemistry , Camptothecin/therapeutic use , Catalysis/radiation effects , Cell Line, Tumor , Drug Carriers/radiation effects , Drug Liberation , Female , Humans , Hydrogen Peroxide/chemistry , Hydrogen Peroxide/metabolism , Light , Methylene Blue/analogs & derivatives , Methylene Blue/radiation effects , Mice, Inbred BALB C , Nanostructures/radiation effects , Neoplasms/metabolism , Oxygen/metabolism , Photosensitizing Agents/chemistry , Photosensitizing Agents/therapeutic use , Photothermal Therapy , Platinum/chemistry , Platinum/radiation effects , Polymers/chemical synthesis , Polymers/chemistry , Polymers/radiation effects , Porosity , Singlet Oxygen/metabolism , Theranostic Nanomedicine
14.
J Nanobiotechnology ; 19(1): 84, 2021 Mar 25.
Article in English | MEDLINE | ID: mdl-33766058

ABSTRACT

BACKGROUND: The development of nano delivery systems is rapidly emerging area of nanotechnology applications where nanomaterials (NMs) are employed to deliver therapeutic agents to specific site in a controlled manner. To accomplish this, green synthesis of NMs is widely explored as an eco-friendly method for the development of smart drug delivery system. In the recent times, use of green synthesized NMs, especially metallic NMs have fascinated the scientific community as they are excellent carriers for drugs. This work demonstrates optimized green, biogenic synthesis of gold nanoparticles (AuNPs) for functionalization with quercetin (QT) and camptothecin (CPT) to enhance potential anti-inflammatory, anti-cancer and anti-angiogenic activities of these drugs. RESULTS: Gold nanoparticles were optimally synthesized in 8 min of reaction at 90 °C, pH 6, using 4 mM of HAuCl4 and 4:1 ratio of extract: HAuCl4. Among different capping agents tested, capping of AuNPs with polyethylene glycol 9000 (PG9) was found best suited prior to functionalization. PG9 capped AuNPs were optimally functionalized with QT in 1 h reaction at 70 °C, pH 7, using 1200 ppm of QT and 1:4 ratio of AuNPs-PG9:QT whereas, CPT was best functionalized at RT in 1 h, pH 12, AuNPs-PG9:CPT ratio of 1:1, and 0.5 mM of CPT. QT functionalized AuNPs showed good anti-cancer activity (IC50 687.44 µg/mL) against MCF-7 cell line whereas test of anti-inflammatory activity also showed excellent activity (IC50 287.177 mg/L). The CAM based assessment of anti-angiogenic activity of CPT functionalized AuNPs demonstrated the inhibition of blood vessel branching confirming the anti-angiogenic effect. CONCLUSIONS: Thus, present study demonstrates that optimally synthesized biogenic AuNPs are best suited for the functionalization with drugs such as QT and CPT. The functionalization of these drugs with biogenic AuNPs enhances the potential anti-inflammatory, anti-cancer and anti-angiogenic activities of these drugs, therefore can be used in biomedical application.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents/pharmacology , Camptothecin/chemistry , Gold/pharmacology , Metal Nanoparticles/chemistry , Polyethylene Glycols/chemistry , Quercetin/chemistry , Gold/chemistry , Green Chemistry Technology/methods , Humans , MCF-7 Cells , Nanotechnology , Plant Extracts
15.
J Photochem Photobiol B ; 215: 112124, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33486396

ABSTRACT

Camptothecin (CPT), an alkaloid, was first discovered from plants and has potent anti-tumor activity. Since then, CPT analogs (namely Irinotecan and Topotecan) have been approved by the FDA for cancer treatments. Curcumin, on the other hand, is a widely used photosensitizer in photodynamic therapy (PDT) treatment. In our previous work, we have reported a straightforward strategy to construct a drug self-delivery system in which two-molecular species Irinotecan and Curcumin can self-assembly into a complex of ion pairs, namely ICN, through intermolecular non-covalent interactions. We found that ICN has slightly better chemotherapy efficacy than its individual components with much fewer side effects. In this paper, we aim to combine the chemotherapy and the PDT of ICN to further improve its anti-tumor performance. The efficient cellular uptake of ICNs was observed by confocal microscopy. Dichloro-dihydro-fluorescein diacetate (DCFH-DA) assay was used to detect the generation of singlet oxygen species. We found that the cell viability was 9% with both chemotherapy and PDT, and 31% with chemotherapy alone for the case with an ICN concentration of 10 µM, which demonstrated that the anti-tumor efficacy against the HT-29 cancer cell line was enhanced substantially with the combination therapy strategy. The study with an in vivo mouse model has further verified that the chemo-PDT dual therapy can inhibit tumor growth by 84% and 18.8% comparing with the control group and the chemotherapy group, respectively. Our results demonstrated that the new strategy using self-assembly and carrier-free nanoparticles with their chemo-PDT dual therapy may provide new opportunities to develop future combinatorial therapy methods in treating cancer.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Camptothecin/chemistry , Camptothecin/pharmacology , Diarylheptanoids/chemistry , Photochemotherapy/methods , Apoptosis/drug effects , Apoptosis/radiation effects , Combined Modality Therapy , HT29 Cells , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Intracellular Space/radiation effects
16.
Drug Deliv ; 28(1): 171-182, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33427515

ABSTRACT

10-HCPT is a topoisomerase I inhibitor effective in the treatment of liver cancer but its use is hampered by its resistance. The expression of hypoxia-inducible factor-1α (HIF-1α) is reportedly upregulated in liver cancer tissues, which is directly linked to the resistance of 10-HCPT. While BBR can significantly decrease the level of HIF-1α according to the literature report. Thus, the aim of this study was to prepare a novel intravenous 10-HCPT-BBR-loaded lipid microsphere (LM) and evaluate their synergistic effect on liver cancer treatment. The optimal preparation mainly included 10.0% oil phase (medium-chain triglyceride:long-chain triglyceride = 1:1), emulsifier (egg lecithin E80 and pluronic F68), antioxidant (0.02% NaHSO3), and pH regulator (0.1 mol/L Hcl). Then, the behaviors of BBR-10-HCPT loaded LM in vitro and in vivo were systematically investigated. In vitro, it showed an obvious sustained-release effect in different release mediums, good physicochemical stability at accelerated and long-term storage conditions, and great anti-proliferative capability toward human liver cancer Hep-3B cells. In vivo, the prepared LM exhibited a longer half-life and higher AUC compared to BBR injection and 10-HCPT injection. More importantly, it was found that The LM was distributed more in the liver, spleen, and tumors, but less in the lungs and heart, especially in the lung. And then, it showed significant inhibition of tumor growth against nude mouse with Hep-3B tumor, and the tumor inhibition rate reached 91.55%. Thus, the data obtained in our study suggested that BBR combined with 10-HCPT can raise curative effect and reduce the toxicity of 10-HCPT.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Berberine/pharmacology , Camptothecin/analogs & derivatives , Carcinoma, Hepatocellular , Liver Neoplasms , Microspheres , Topoisomerase I Inhibitors/pharmacology , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/chemistry , Berberine/administration & dosage , Berberine/chemistry , Camptothecin/administration & dosage , Camptothecin/chemistry , Camptothecin/pharmacology , Cell Line, Tumor , Drug Delivery Systems , Drug Liberation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Mice , Mice, Nude , Rats , Topoisomerase I Inhibitors/administration & dosage , Topoisomerase I Inhibitors/chemistry , Xenograft Model Antitumor Assays
17.
Biotechnol Prog ; 36(6): e3039, 2020 11.
Article in English | MEDLINE | ID: mdl-32558398

ABSTRACT

In the study, endophytic fungi isolated from Ophiorrhiza mungos were screened for camptothecin (CPT) biosynthetic potential by high performance liquid chromatography (HPLC). Among the 16 fungi screened, OmF3, OmF4, and OmF6 were identified to synthesize CPT. Further LC-MS analysis also showed the presence of CPT specific m/z of 349 for the extracts from OmF3, OmF4, and OmF6. However, the fragmentation masses with m/z of 320, 305, 277 and 220 specific to the CPT could be identified only for the OmF3 and OmF4. These CPT producing fungi were further identified as Meyerozyma sp. OmF3 and Talaromyces sp. OmF4. The cultures of these two fungi were then supplemented with nanoparticles and analyzed for the quantitative enhancement of CPT production by LC-MS/MS. From the result, Meyerozyma sp. OmF3 was found to produce 947.3 ± 12.66 µg/L CPT, when supplemented with 1 µg/mL zinc oxide nanoparticles and the same for uninduced parental strain OmF3 was only 1.77 ± 0.13 µg/L. At the same time, Talaromyces sp. OmF4 showed the highest production of 28.97 ± 0.37 µg/L of CPT when cultured with 10 µg/mL silver nanoparticles and the same for uninduced strain was 1.19 ± 0.24 µg/L. The observed quantitative enhancement of fungal CPT production is highly interesting as it is a rapid and cost effective method. The study is remarkable due to the identification of novel fungal sources for CPT production and its enhancement by nanoparticle supplementation.


Subject(s)
Antineoplastic Agents, Phytogenic/isolation & purification , Camptothecin/isolation & purification , Fungi/chemistry , Metal Nanoparticles/chemistry , Animals , Antineoplastic Agents, Phytogenic/biosynthesis , Antineoplastic Agents, Phytogenic/chemistry , Camptothecin/biosynthesis , Camptothecin/chemistry , Chromatography, High Pressure Liquid , Humans , Tandem Mass Spectrometry
18.
Mol Pharm ; 17(7): 2411-2425, 2020 07 06.
Article in English | MEDLINE | ID: mdl-32437163

ABSTRACT

Tumor-targeted drug delivery via chemotherapy is very effective on cancer treatment. For potential anticancer agent such as Camptothecin (CPT), high chemotherapeutic efficacy and accurate tumor targeting are equally crucial. Inspired by special CD44 binding capability from hyaluronic acid (HA), in this study, novel HA-coated CPT nanocrystals were successfully prepared by an antisolvent precipitation method for tumor-targeted delivery of hydrophobic drug CPT. These HA-coated CPT nanocrystals demonstrated high drug loading efficiency, improved aqueous dispersion, prolonged circulation, and enhanced stability resulting from their nanoscaled sizes and hydrophilic HA layer. Moreover, as compared to crude CPT and naked CPT nanocrystals, HA-coated CPT nanocrystals displayed dramatically enhanced in vitro anticancer activity, apoptosis-inducing potency against CD44 overexpressed cancer cells, and lower toxic effect toward normal cells due to pH-responsive drug release behavior and specific HA-CD44 mediated endocytosis. Additionally, HA-coated CPT nanocrystals performed fairly better antimigration activity and biocompatibility. The possible molecular mechanism regarding this novel drug formulation might be linked to intrinsic mitochondria-mediated apoptosis by an increase of Bax to Bcl-2 ratio and upregulation of P53. Consequently, HA-coated CPT nanocrystals are expected to be an effective nanoplatform in drug delivery for cancer therapy.


Subject(s)
Antineoplastic Agents/administration & dosage , Camptotheca/chemistry , Camptothecin/administration & dosage , Drug Delivery Systems/methods , Hyaluronic Acid/chemistry , Nanoparticles/chemistry , Neoplasms/metabolism , Phytotherapy/methods , Plant Extracts/administration & dosage , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Camptothecin/chemistry , Cell Movement/drug effects , Cell Survival/drug effects , Drug Liberation , Drug Stability , Hep G2 Cells , Humans , Hyaluronan Receptors/metabolism , Hyaluronic Acid/metabolism , Hydrogen-Ion Concentration , MCF-7 Cells , Neoplasms/pathology , Particle Size , Plant Extracts/chemistry , Protein Binding
19.
ACS Appl Mater Interfaces ; 11(43): 40704-40715, 2019 Oct 30.
Article in English | MEDLINE | ID: mdl-31577408

ABSTRACT

Anticancer nanomedicine-based multimodal imaging and synergistic therapy hold great promise in cancer diagnosis and therapy owing to their abilities to improve therapeutic efficiency and reduce unnecessary side effects, producing promising clinical prospects. Herein, we integrated chemotherapeutic drug camptothecin (CPT) and near-infrared-absorbing new indocyanine green (IR820) into a single system by charge interaction and obtained a tumor-microenvironment-activatable PCPTSS/IR820 nanoreactor to perform thermal/fluorescence/photoacoustic-imaging-guided chemotherapy and photothermal therapy simultaneously. Specifically, the generated PCPTSS/IR820 showed an excellent therapeutic agent loading content and size stability, and the trials in vitro and in vivo suggested that the smart PCPTSS/IR820 could deeply permeate into tumor tissues due to its suitable micellar size. Upon near-infrared laser irradiation, the nanoreactor further produced a terrific synergism of chemo-photo treatment for cancer therapy. Therefore, the PCPTSS/IR820 polyprodrug-based nanoreactor holds outstanding promise for multimodal imaging and combined dual therapy.


Subject(s)
Camptothecin , Drug Carriers , Hyperthermia, Induced , Indocyanine Green/analogs & derivatives , Nanostructures , Neoplasms , Photochemotherapy , Prodrugs , Tumor Microenvironment/drug effects , Animals , Camptothecin/chemistry , Camptothecin/pharmacokinetics , Camptothecin/pharmacology , Cell Line, Tumor , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Carriers/pharmacology , Female , Humans , Indocyanine Green/chemistry , Indocyanine Green/pharmacokinetics , Indocyanine Green/pharmacology , Mice , Mice, Inbred BALB C , Mice, Nude , Nanostructures/chemistry , Nanostructures/therapeutic use , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/therapy , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Prodrugs/pharmacology
20.
ACS Appl Mater Interfaces ; 11(17): 15262-15275, 2019 May 01.
Article in English | MEDLINE | ID: mdl-30964624

ABSTRACT

Nanoparticles camouflaged by red blood cell (RBC) membranes have attracted considerable attention owing to reservation of structure of membrane and surface proteins, endowing prominent cell-specific function including biocompatibility, prolonged circulation lifetime, and reduced reticular endothelial system (RES) uptake ability. Considering the drawbacks of carrier-free nanomedicine including the serious drug burst release, poor stability, and lack of immune escape function, herein we developed and fabricated a novel RBC membranes biomimetic combinational therapeutic system by enveloping the small molecular drug coassemblies of 10-hydroxycamptothecin (10-HCPT) and indocyanine green (ICG) in the RBC membranes for prolonged circulation, controlled drug release, and synergistic chemo-photothermal therapy (PTT). The self-reorganized RBCs@ICG-HCPT nanoparticles (NPs) exhibited a diameter of ∼150 nm with core-shell structure, high drug payload (∼92 wt %), and reduced RES uptake function. Taking advantage of the stealth functionality of RBC membranes, RBCs@ICG-HCPT NPs remarkably enhanced the accumulation at the tumor sites by passive targeting followed by cellular endocytosis. Upon the stimuli of near-infrared laser followed by acidic stimulation, RBCs@ICG-HCPT NPs showed exceptional instability by heat-mediated membrane disruption and pH change, thereby triggering the rapid disassembly and accelerated drug release. Consequently, compared with individual treatment, RBCs@ICG-HCPT NPs under dual-stimuli accomplished highly efficient apoptosis in cancer cells and remarkable ablation of tumors by chemo-PTT. This biomimetic nanoplatform based on carrier-free, small molecular drug coassemblies integrating imaging capacity as a promising theranostic system provides potential for cancer diagnosis and combinational therapy.


Subject(s)
Antineoplastic Agents, Phytogenic/chemistry , Biomimetics , Camptothecin/analogs & derivatives , Cell Membrane/chemistry , Infrared Rays , Nanoparticles/chemistry , Neoplasms/therapy , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis/drug effects , Camptothecin/chemistry , Camptothecin/pharmacology , Camptothecin/therapeutic use , Erythrocytes/cytology , Erythrocytes/metabolism , HeLa Cells , Humans , Hydrogen-Ion Concentration , Indocyanine Green/chemistry , Male , Mice , Mice, Nude , Nanoparticles/therapeutic use , Neoplasms/drug therapy , Neoplasms/pathology , Phototherapy , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL