Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 7.603
1.
Pharm Biol ; 62(1): 367-393, 2024 Dec.
Article En | MEDLINE | ID: mdl-38745507

CONTEXT: Rice bran arabinoxylan compound (RBAC) is a natural immunomodulator with anticancer properties. OBJECTIVE: This study critically evaluates the available evidence on the biological pathways of RBAC and its effects on cancer treatment. METHODS: This secondary analysis of a scoping review includes studies evaluating the mechanisms of RBAC on healthy or malignant cells, animal models, or humans for cancer prevention or treatment. Data from randomized controlled trials on survival and quality of life outcomes were subjectd to meta analysis. RESULTS: The evidence synthesis was based on 38 articles. RBAC exhibited antitumor properties by promoting apoptosis and restoring immune function in cancer patients to enhance inflammatory and cytotoxic responses to block tumorigenesis. RBAC works synergistically with chemotherapeutic agents by upregulating drug transport. In a clinical trial, combining RBAC with chemoembolization in treating liver cancer showed improved response, reduced recurrence rates, and prolonged survival. RBAC also augments the endogenous antioxidant system to prevent oxidative stress and protect against radiation side effects. In addition, RBAC has chemoprotective effects. Animals and humans have exhibited reduced toxicity and side effects from chemotherapy. Meta analysis indicates that RBAC treatment increases the survival odds by 4.02-times (95% CI: 1.67, 9.69) in the first year and 2.89-times (95% CI: 1.56, 5.35) in the second year. CONCLUSION: RBAC is a natural product with immense potential in cancer treatment. Additional research is needed to characterize, quantify, and standardize the active ingredients in RBAC responsible for the anticancer effects. More well-designed, large-scale clinical trials are required to substantiate the treatment efficacies further.


Neoplasms , Oryza , Xylans , Xylans/pharmacology , Humans , Animals , Neoplasms/drug therapy , Biological Products/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/administration & dosage , Randomized Controlled Trials as Topic , Antineoplastic Agents/pharmacology
2.
Medicine (Baltimore) ; 103(20): e38207, 2024 May 17.
Article En | MEDLINE | ID: mdl-38758870

This study aimed to investigate changes in hand sensation (finger tactile threshold and two-point discrimination) and function in patients with malignant lymphoma, particularly during the early stages of chemotherapy with vincristine. Eighteen patients with malignant lymphoma were enrolled in this study. Data on the Common Terminology Criteria for Adverse Events Version 4.0, the visual analog scale for hand numbness, the Semmes Weinstein monofilament test, static and moving two-point discrimination (2PD), grip strength, pinch strength, and the Purdue Pegboard test were collected at 3 time points: before the start of chemotherapy (T0), after the first cycle of chemotherapy (T1), and after the second cycle of chemotherapy (T2). No significant changes were observed in Semmes Weinstein monofilament test at T0, T1, or T2 in either hand. However, the static 2PD was significantly worse for the right ring, little, and left middle fingers, whereas the moving 2PD was significantly worse for the right ring, left index, middle, and ring fingers. Furthermore, the visual analog scale scores for hand numbness and left-hand grip strength worsened significantly. Right-hand grip strength, pinch strength of both hands, and Purdue Pegboard test showed no significant deterioration. Chemotherapy with vincristine may affect hand sensation and function in patients with malignant lymphoma by exacerbating finger 2PD and hand numbness. Additionally, during the early stages of vincristine chemotherapy, it is important to monitor for a decrease in grip strength specifically in the left hand.


Hand Strength , Hand , Lymphoma , Vincristine , Humans , Vincristine/adverse effects , Vincristine/administration & dosage , Male , Female , Middle Aged , Lymphoma/drug therapy , Aged , Adult , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Agents, Phytogenic/administration & dosage , Hypesthesia/chemically induced
3.
Zhongguo Zhong Yao Za Zhi ; 49(8): 2117-2127, 2024 Apr.
Article Zh | MEDLINE | ID: mdl-38812227

Piperlongumine(PL), a natural alkaloid extracted from Piperis Longi Fructus, has attracted much attention in recent years because of its strong anti-tumor activity, little toxicity to normal cells, and excellent sensitizing effect combined with chemotherapy and radiotherapy, which endow PL with unique advantages as an anti-tumor drug. However, similar to other alkaloids, PL has low water solubility and poor bioavailability. To improve the application of PL in the clinical treatment of tumors, researchers have constructed various nano-drug delivery systems to increase the efficiency of PL delivery. This paper reviewed the physicochemical properties, anti-tumor mechanism, combined therapies, and nano-drug delivery systems of PL in recent years. The review aimed to provide a reference for further research on the anti-tumor effect and nano-drug delivery system of PL. Moreover, this review is expected to provide a reference for the development and application of PL in the anti-tumor therapies.


Dioxolanes , Neoplasms , Dioxolanes/chemistry , Humans , Animals , Neoplasms/drug therapy , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Delivery Systems , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/administration & dosage , Nanoparticle Drug Delivery System/chemistry , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacology , Piperidones
4.
Curr Pharm Biotechnol ; 25(5): 599-622, 2024.
Article En | MEDLINE | ID: mdl-38807329

Colorectal cancer (CRC) is a complex and multifactorial disorder in middle-aged people. Several modern medicines are available for treating and preventing it. However, their therapeutic uses are limited due to drawbacks, such as gastric perforation, diarrhea, intestinal bleeding, abdominal cramps, hair loss, nausea, vomiting, weight loss, and adverse reactions. Hence, there is a continuous quest for safe and effective medicines to manage human health problems, like CRC. In this context, herbal medicines are considered an alternative disease control system. It has become popular in countries, like American, European, and Asian, due to its safety and effectiveness, which has been practiced for 1000 years. During the last few decades, herbal medicines have been widely explored through multidisciplinary fields for getting active compounds against human diseases. Several herbal bioactives, like curcumin, glycyrrhizin, paclitaxel, chlorogenic acid, gallic acid, catechin, berberine, ursolic acid, betulinic acid, chrysin, resveratrol, quercetin, etc., have been found to be effective against CRC. However, their pharmacological applications are limited due to low bioavailability and therapeutic efficacy apart from their several health benefits. An effective delivery system is required to increase their bioavailability and efficacy. Therefore, targeted novel drug delivery approaches are promising for improving these substances' solubility, bioavailability, and therapeutic effects. Novel carrier systems, such as liposomes, nanoparticles, micelles, microspheres, dendrimers, microbeads, and hydrogels, are promising for delivering poorly soluble drugs to the target site, i.e., the colon. Thus, the present review is focused on the pathophysiology, molecular pathways, and diagnostic and treatment approaches for CRC. Moreover, an emphasis has been laid especially on herbal bioactive-based novel delivery systems and their clinical updates.


Colorectal Neoplasms , Humans , Colorectal Neoplasms/drug therapy , Drug Carriers/chemistry , Animals , Nanoparticles/chemistry , Drug Delivery Systems/methods , Phytochemicals/therapeutic use , Phytochemicals/chemistry , Phytochemicals/administration & dosage , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/therapeutic use , Antineoplastic Agents, Phytogenic/chemistry
5.
Sci Rep ; 14(1): 10075, 2024 05 02.
Article En | MEDLINE | ID: mdl-38698201

Intraperitoneal (IP) chemotherapy with paclitaxel (PTX) for gastric cancer (GC) with peritoneal metastasis (PM) is considered a promising treatment approach, however, there are no useful biomarkers to predict the efficacy of IP therapy. We examined the association between intra-peritoneal exosomes, particularly exosomal micro-RNAs (exo-miRNAs), and IP-chemo sensitivity. MKN45 cells that were cultured with intra-peritoneal exosomes from patients who did not respond to IP therapy with PTX (IPnon-respond group) exhibited resistance to PTX compared with exosomes from responding patients (IPrespond group) (p = 0.002). A comprehensive search for exo-miRNAs indicated that miR-493 was significantly up-regulated in exosomes from the IPnon-respond group compared with those collected from the IPrespond group. The expression of miR-493 in PTX-resistant MKN45 cells (MKN45PTX-res) was higher compared with that in MKN45. In addition, MKN45PTX-res cells exhibited lower MAD2L1 gene and protein expression compared with MKN45. Finally, miR-493 enhancement by transfection of miR-493 mimics significantly down-regulated MAD2L1 expression in MKN45 cells and reduced PTX sensitivity. Our results suggest that intra-peritoneal exo-miR-493 is involved in chemoresistance to PTX by downregulating MAD2L1 in GC with PM. Exo-miR-493 may be a biomarker for chemoresistance and prognosis of GC patients with PM and may also be a promising therapeutic target.


Drug Resistance, Neoplasm , Exosomes , Gene Expression Regulation, Neoplastic , Mad2 Proteins , MicroRNAs , Paclitaxel , Peritoneal Neoplasms , Stomach Neoplasms , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Paclitaxel/administration & dosage , Drug Resistance, Neoplasm/genetics , Exosomes/metabolism , Exosomes/genetics , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/drug therapy , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/metabolism , Cell Line, Tumor , Male , Female , Mad2 Proteins/metabolism , Mad2 Proteins/genetics , Middle Aged , Gene Expression Regulation, Neoplastic/drug effects , Aged , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Antineoplastic Agents, Phytogenic/administration & dosage
6.
Mol Pharm ; 21(5): 2327-2339, 2024 May 06.
Article En | MEDLINE | ID: mdl-38576375

In the present study, we investigated the role of lipid composition of camptothecin (CPT)-loaded liposomes (CPT-Lips) to adjust their residence time, drug distribution, and therefore the toxicities and antitumor activity. The CPT was loaded into liposomes using a click drug loading method, which utilized liposomes preloaded with GSH and then exposed to CPT-maleimide. The method produced CPT-Lips with a high encapsulation efficiency (>95%) and sustained drug release. It is shown that the residence times of CPT-Lips in the body were highly dependent on lipid compositions with an order of non-PEGylated liposomes of unsaturated lipids < non-PEGylated liposomes of saturated lipids < PEGylated liposomes of saturated lipids. Interestingly, the fast clearance of CPT-Lips resulted in significantly decreased toxicities but did not cause a significant decrease in their in vivo antitumor activity. These results suggested that the lipid composition could effectively adjust the residence time of CPT-Lips in the body and further optimize their therapeutic index, which would guide the development of a liposomal formulation of CPT.


Camptothecin , Lipids , Liposomes , Camptothecin/chemistry , Camptothecin/administration & dosage , Camptothecin/pharmacokinetics , Camptothecin/pharmacology , Liposomes/chemistry , Animals , Mice , Lipids/chemistry , Humans , Drug Liberation , Drug Delivery Systems/methods , Polyethylene Glycols/chemistry , Cell Line, Tumor , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/pharmacology , Female , Click Chemistry/methods , Mice, Inbred BALB C
7.
J Control Release ; 369: 765-774, 2024 May.
Article En | MEDLINE | ID: mdl-38593976

The combination of chemotherapy and gene therapy holds great promise for the treatment and eradication of tumors. However, due to significant differences in physicochemical properties between chemotherapeutic agents and functional nucleic acid drugs, direct integration into a single nano-agent is hindered, impeding the design and construction of an effective co-delivery nano-platform for synergistic anti-tumor treatments. In this study, we have developed an mRNA-responsive two-in-one nano-drug for effective anti-tumor therapy by the direct self-assembly of 2'-fluoro-substituted antisense DNA against P-glycoprotein (2'F-DNA) and chemo drug paclitaxel (PTX). The 2'-fluoro modification of DNA could significantly increase the interaction between the therapeutic nucleic acid and the chemotherapeutic drug, promoting the successful formation of 2'F-DNA/PTX nanospheres (2'F-DNA/PTX NSs). Due to the one-step self-assembly process without additional carrier materials, the prepared 2'F-DNA/PTX NSs exhibited considerable loading efficiency and bioavailability of PTX. In the presence of endogenous P-glycoprotein mRNA, the 2'F-DNA/PTX NSs were disassembled. The released 2'F-DNA could down-regulate the expression of P-glycoprotein, which decreased the multidrug resistance of tumor cells and enhanced the chemotherapy effect caused by PTX. In this way, the 2'F-DNA/PTX NSs could synergistically induce the apoptosis of tumor cells and realize the combined anti-tumor therapy. This strategy might provide a new tool to explore functional intracellular co-delivery nano-systems with high bioavailability and exhibit potential promising in the applications of accurate diagnosis and treatment of tumors.


Genetic Therapy , Paclitaxel , RNA, Messenger , RNA, Messenger/administration & dosage , Paclitaxel/administration & dosage , Paclitaxel/pharmacology , Paclitaxel/chemistry , Humans , Animals , Genetic Therapy/methods , Cell Line, Tumor , Mice, Nude , Neoplasms/therapy , Neoplasms/drug therapy , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacology , Mice, Inbred BALB C , DNA/administration & dosage , Nanoparticles/chemistry , Female
8.
J Control Release ; 369: 622-629, 2024 May.
Article En | MEDLINE | ID: mdl-38604383

Enhancing the delivery and release efficiency of hydroxyl agents, constrained by high pKa values and issues of release rate or unstable linkage, is a critical challenge. To address this, a self-immolative linker, composed of a modifiable p-hydroxybenzyl ether and a fast cyclization adapter (N-(ortho-hydroxyphenyl)-N-methylcarbamate) was strategically designed, for the synthesis of prodrugs. The innovative linker not only provides a side chain modification but also facilitates the rapid release of the active payloads, thereby enabling precise drug delivery. Particularly, five prodrug model compounds (J1, J2, J3, J5 and J6) were synthesized to evaluate the release rates by using ß-glucuronic acid as trigger and five hydroxyl compounds as model payloads. Significantly, all prodrug model compounds could efficiently release the hydroxyl payloads under the action of ß-glucuronidase, validating the robustness of the linker. And then, to assess the drug delivery and release efficiency using endogenous albumin as a transport vehicle, J1148, a SN38 prodrug modified with maleimide side chain was synthesized. Results demonstrated that J1148 covalently bound to plasma albumin through in situ Michael addition, effectively targeting the tumor microenvironment. Activated by ß-glucuronidase, J1148 underwent a classical 1, 6-elimination, followed by rapid cyclization of the adapter, thereby releasing SN38. Impressively, J1148 showed excellent therapeutic efficacy against human colonic cancer xenograft model, leading to a significant reduction or even disappearance of tumors (3/6 of mice cured). These findings underscore the potential of the designed linker in the delivery system of hydroxyl agents, positioning it at the forefront of advancements in drug delivery technology.


Drug Delivery Systems , Irinotecan , Prodrugs , Prodrugs/administration & dosage , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Animals , Humans , Irinotecan/administration & dosage , Irinotecan/pharmacokinetics , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Camptothecin/pharmacokinetics , Camptothecin/chemistry , Drug Liberation , Mice, Nude , Cell Line, Tumor , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacokinetics , Female , Mice , Albumins/administration & dosage , Albumins/chemistry , Glucuronidase/metabolism , Mice, Inbred BALB C
9.
Int J Pharm ; 656: 124078, 2024 May 10.
Article En | MEDLINE | ID: mdl-38569978

The role of tumor stroma in solid tumors has been widely recognized in cancer progression, metastasis and chemoresistance. Cancer-associated fibroblasts (CAFs) play a crucial role in matrix remodeling and promoting cancer cell stemness and resistance via reciprocal crosstalk. Residual tumor tissue after surgical removal as well as unresectable tumors face therapeutic challenges to achieve curable outcome. In this study, we propose to develop a dual delivery approach by combining p21-activated kinase 1 (PAK1) inhibitor (FRAX597) to inhibit tumor stroma and chemotherapeutic agent paclitaxel (PTX) to kill cancer cells using electrospun nanofibers. First, the role of the PAK1 pathway was established in CAF differentiation, migration and contraction using relevant in vitro models. Second, polycaprolactone polymer-based nanofibers were fabricated using a uniaxial electrospinning technique to incorporate FRAX597 and/or PTX, which showed a uniform texture and a prolonged release of both drugs for 16 days. To test nanofibers, stroma-rich 3D heterospheroid models were set up which showed high resistance to PTX nanofibers compared to stroma-free homospheroids. Interestingly, nanofibers containing PTX and FRAX597 showed strong anti-tumor effects on heterospheroids by reducing the growth and viability by > 90 % compared to either of single drug-loaded nanofibers. These effects were reflected by reduced intra-spheroidal expression levels of collagen 1 and α-smooth muscle actin (α-SMA). Overall, this study provides a new therapeutic strategy to inhibit the tumor stroma using PAK1 inhibitor and thereby enhance the efficacy of chemotherapy using nanofibers as a local delivery system for unresectable or residual tumor. Use of 3D models to evaluate nanofibers highlights these models as advanced in vitro tools to study the effect of controlled release local drug delivery systems before animal studies.


Nanofibers , Paclitaxel , p21-Activated Kinases , Paclitaxel/administration & dosage , Paclitaxel/pharmacology , Nanofibers/administration & dosage , p21-Activated Kinases/antagonists & inhibitors , p21-Activated Kinases/metabolism , Humans , Cell Line, Tumor , Spheroids, Cellular/drug effects , Polyesters/chemistry , Polyesters/administration & dosage , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/metabolism , Drug Delivery Systems/methods , Cell Movement/drug effects , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacology , Drug Liberation , Cell Differentiation/drug effects
10.
Expert Rev Clin Pharmacol ; 17(5-6): 525-532, 2024.
Article En | MEDLINE | ID: mdl-38652518

BACKGROUND: Paclitaxel hypersensitivity reactions (HSRs) are prevalent, especially in females. The common paclitaxel pretreatment, dexamethasone, may inhibit chemotherapy efficacy and accelerate tumor progression. We aimed to balance paclitaxel HSRs and the lowest dexamethasone dose for gynecologic malignancies. METHODS: We retrospectively examined 1,074 cycles of 3-weekly paclitaxel-containing treatment for 231 gynecologic malignancies at Xiangya Hospital. HSR incidence with different dexamethasone regimens was the primary outcome. Risk factors were examined in all cycles using univariate and multivariate models with generalized estimating equations. A subgroup analysis of initial exposure to paclitaxel was also conducted. RESULTS: HSR occurred in 33 patients (14.29%) and 49 cycles (4.56%), including 69.39% in cycles 1-2. There were no severe HSRs (grade ≥3). Different premedication regimens, including dexamethasone dosage and route, ranitidine presence or absence, didn't affect HSR incidence in univariate and multivariate analyzes (p > 0.05). Premenopausal women exerted fewer HSRs (ORadj 0.22, 95%CI 0.08-0.58; p = 0.002). At the first exposure to paclitaxel, more than 10 mg of dexamethasone didn't diminish HSRs (OR 0.83, 95%CI 0.27-2.59; p = 0.753). CONCLUSIONS: In gynecologic malignancies, 10 mg dexamethasone along with 20 mg diphenhydramine may be adequate to prevent paclitaxel HSRs without ranitidine. It is necessary to reevaluate paclitaxel premedication regimens.


Antineoplastic Agents, Phytogenic , Dexamethasone , Dose-Response Relationship, Drug , Drug Hypersensitivity , Genital Neoplasms, Female , Paclitaxel , Humans , Female , Dexamethasone/administration & dosage , Paclitaxel/administration & dosage , Paclitaxel/adverse effects , Retrospective Studies , Genital Neoplasms, Female/drug therapy , Middle Aged , Drug Hypersensitivity/prevention & control , Drug Hypersensitivity/etiology , Adult , Aged , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/adverse effects , Risk Factors , Incidence
11.
Biomed Pharmacother ; 174: 116482, 2024 May.
Article En | MEDLINE | ID: mdl-38520866

Chemoimmunotherapy has emerged as a promising strategy for improving the efficacy of cancer treatment. Herein, we present PD-1 receptor-presenting membrane-coated paclitaxel dimers nanoparticles (PD-1@PTX2 NPs) for enhanced treatment efficacy. PD-1 cell membrane-cloaked PTX dimer exhibited effective cellular uptake and increased cytotoxicity against cancer cells. PD-1@PTX2 NPs could selectively bind with PD-L1 ligands expressed on breast cancer cells. Our nanoparticles exhibit a remarkable tumor growth inhibition rate of 71.3% in mice bearing 4T1 xenografts and significantly prolong survival in mouse models of breast cancer. Additionally, our nanoparticles promoted a significant 3.2-fold increase in CD8+ T cell infiltration and 73.7% regulatory T cell (Treg) depletion within tumors, boosting a robust antitumor immune response. These findings underscore the potential of utilizing immune checkpoint receptor-presented PTX nanoparticles to enhance the efficacy of chemoimmunotherapy, providing an alternative approach for improving cancer treatment.


Immunotherapy , Mice, Inbred BALB C , Nanoparticles , Paclitaxel , Programmed Cell Death 1 Receptor , Paclitaxel/pharmacology , Paclitaxel/administration & dosage , Animals , Nanoparticles/chemistry , Programmed Cell Death 1 Receptor/metabolism , Female , Humans , Immunotherapy/methods , Mice , Cell Line, Tumor , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/immunology , Cell Membrane/metabolism , Cell Membrane/drug effects , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/administration & dosage , Xenograft Model Antitumor Assays , Dimerization , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , B7-H1 Antigen/metabolism
12.
Phytomedicine ; 128: 155521, 2024 Jun.
Article En | MEDLINE | ID: mdl-38489891

BACKGROUND: The ancient Chinese herb Salvia miltiorrhiza Bunge (Danshen), plays the important role in cardiovascular and cerebrovascular disease. Furthermore, Danshen could also be used for curing carcinogenesis. Up to now, the anti-tumor effects of the main active constituents of Danshen have made great progress. However, the bioavailability of the active constituents of Danshen were restricted by their unique physical characteristics, like low oral bioavailability, rapid degradation in vivo and so on. PURPOSE: With the leap development of nano-delivery systems, the shortcomings of the active constituents of Danshen have been greatly ameliorated. This review tried to summarize the recent progress of the active constituents of Danshen based delivery systems used for anti-tumor therapeutics. METHODS: A systematic literature search was conducted using 5 databases (Embase, Google scholar, PubMed, Scopus and Web of Science databases) for the identification of relevant data published before September 2023. The words "Danshen", "Salvia miltiorrhiza", "Tanshinone", "Salvianolic acid", "Rosmarinic acid", "tumor", "delivery", "nanomedicine" and other active ingredients contained in Danshen were searched in the above databases to gather information about pharmaceutical decoration for the active constituents of Danshen used for anti-tumor therapeutics. RESULTS: The main extracts of Danshen could inhibit the proliferation of tumor cells effectively and a great deal of studies were conducted to design drug delivery systems to ameliorate the anti-tumor effect of the active contents of Danshen through different ways, like improving bioavailability, increasing tumor targeting ability, enhancing biological barrier permeability and co-delivering with other active agents. CONCLUSION: This review systematically represented recent progress of pharmaceutical decorations for the active constituents of Danshen used for anti-tumor therapeutics, revealing the diversity of nano-decoration skills and trying to inspire more designs of Danshen based nanodelivery systems, with the hope that bringing the nanomedicine of the active constituents of Danshen for anti-tumor therapeutics from bench to bedside in the near future.


Antineoplastic Agents, Phytogenic , Drugs, Chinese Herbal , Salvia miltiorrhiza , Salvia miltiorrhiza/chemistry , Drugs, Chinese Herbal/administration & dosage , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/chemistry , Humans , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacology , Drug Delivery Systems , Animals , Neoplasms/drug therapy , Nanoparticle Drug Delivery System/chemistry , Nanoparticles/chemistry
13.
J Pharm Pharmacol ; 76(6): 631-645, 2024 Jun 06.
Article En | MEDLINE | ID: mdl-38507715

PURPOSE: Although resveratrol (RES) is an efficacious molecule, its therapeutic activity is impeded by significant limitations, such as rapid oral absorption, poor oral bioavailability, and low water solubility. Therefore, the preparation of RES in different pharmaceutical carriers represents an important tool to enhance its therapeutic applications. This study aims to potentiate the anti-cancer activity of RES by formulating it into a novel nanocarrier called Smart Lipid. METHODS: RES-loaded Smart Lipids were prepared by high-shear hot homogenization method utilizing a 21 × 32 factorial design with three factors at different levels: the total lipid concentration, the concentration of surfactant, and the type of surfactant. The responses were evaluated based on entrapment efficiency percentages and particle size. RESULTS: Our novel optimized RES-loaded Smart Lipid formula showed small particle size (288.63 ± 5.55 nm), good zeta potential (-16.44 ± 0.99 mV), and an entrapment efficiency of 86.346 ± 3.61% with spherical, clearly distinct, and no signs of fusion by transmission electron microscopy. Further characterization was done using differential scanning calorimetry, which showed no interaction between the drug and other components as the optimum lyophilized formula showed a peak at 54.75°C, which represents the lipid mixture, with an undetectable characteristic peak of the drug, which indicates entrapment of the drug, and the structure of the compounds was confirmed by Fourier transform-infrared spectroscopy, in which the majority of the drug's characteristic peaks disappeared when loaded into Smart Lipid, which may indicate Smart Lipid's ability to reduce the stretching and bending between bonds in RES. In addition, the optimized formula showed a sustained release pattern compared to RES suspension. Finally, the cytotoxic activity of the optimized RES-loaded Smart Lipid on different cell lines (human breast adenocarcinoma (MCF7), human hepatocellular carcinoma (HepG2), and human colon cancer cells (HT29)) was assessed through MTT assay (7-fold reduction in the IC50, from 3.7 ± 0.5 µM for free RES to 0.5 ± 0.033 µM for Smart Lipid loaded formula against MCF7, 3-fold reduction in the IC50 against HepG2 cells, from 10.01 ± 0.35 to 3.16 ± 0.21 µMm, and a more than 10-fold reduction in the IC50 from more than 100 to 10 ± 0.57 µM against HT-29 cells) and its effect on cell cycle progression and apoptosis induction were assessed using flow cytometry and annexin V kit, respectively. Our results showed that RES-loaded Smart Lipid significantly reduced cell viability, induced cell cycle arrest at G0/G1 phase, and apoptosis compared to free formula and free RES suspension. CONCLUSION: Loading RES into this novel kind of nanocarrier enhanced RES absorption, cellular accumulation, and improved its anticancer properties.


Drug Carriers , Lipids , Particle Size , Resveratrol , Resveratrol/pharmacology , Resveratrol/administration & dosage , Resveratrol/chemistry , Humans , Lipids/chemistry , Drug Carriers/chemistry , Hep G2 Cells , Nanoparticles/chemistry , Drug Compounding/methods , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/administration & dosage , Solubility , Calorimetry, Differential Scanning , Surface-Active Agents/chemistry , Surface-Active Agents/pharmacology , Cell Line, Tumor , Chemistry, Pharmaceutical/methods , Drug Liberation , Drug Design/methods , MCF-7 Cells , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/chemistry , Stilbenes/pharmacology , Stilbenes/chemistry , Stilbenes/administration & dosage
14.
J Control Release ; 369: 179-198, 2024 May.
Article En | MEDLINE | ID: mdl-38368947

Engineering human enzymes for therapeutic applications is attractive but introducing new amino acids may adversely affect enzyme stability and immunogenicity. Here we used a mammalian membrane-tethered screening system (ECSTASY) to evolve human lysosomal beta-glucuronidase (hBG) to hydrolyze a glucuronide metabolite (SN-38G) of the anticancer drug irinotecan (CPT-11). Three human beta-glucuronidase variants (hBG3, hBG10 and hBG19) with 3, 10 and 19 amino acid substitutions were identified that display up to 40-fold enhanced enzymatic activity, higher stability than E. coli beta-glucuronidase in human serum, and similar pharmacokinetics in mice as wild-type hBG. The hBG variants were two to three orders of magnitude less immunogenic than E. coli beta-glucuronidase in hBG transgenic mice. Intravenous administration of an immunoenzyme (hcc49-hBG10) targeting a sialyl-Tn tumor-associated antigen to mice bearing human colon xenografts significantly enhanced the anticancer activity of CPT-11 as measured by tumor suppression and mouse survival. Our results suggest that genetically-modified human enzymes represent a good alternative to microbially-derived enzymes for therapeutic applications.


Camptothecin , Glucuronidase , Irinotecan , Mice, Transgenic , Prodrugs , Animals , Prodrugs/administration & dosage , Humans , Irinotecan/administration & dosage , Irinotecan/pharmacokinetics , Glucuronidase/genetics , Glucuronidase/metabolism , Camptothecin/analogs & derivatives , Camptothecin/pharmacokinetics , Camptothecin/administration & dosage , Camptothecin/therapeutic use , Protein Engineering , Mice , Cell Line, Tumor , Female , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/therapeutic use , Neoplasms/drug therapy , Neoplasms/immunology , Xenograft Model Antitumor Assays , Enzyme Stability , Mice, Nude
15.
Curr Drug Targets ; 25(3): 201-219, 2024.
Article En | MEDLINE | ID: mdl-38231060

Breast cancer is a widespread condition that kills more women from cancer-related causes than any other type of cancer globally. Women who have estrogen-dependent, initial metastatic breast cancer frequently receive treatment with surgery, radiation therapy, and chemotherapy. They may also get more specialized treatments like tamoxifen or aromatase inhibitors (anastrozole or letrozole). The World Health Organisation reported in 2012 that by 2030, breast cancer will be more common worldwide. There are several phytochemicals, such as isoflavones, coumestans, lignans, and prenylflavonoides. Isoflavones have been shown in studies to prevent the spread of breast cancer and to trigger apoptosis. Targeting BCs in metastatic breast cancer may be made possible by combining well-formulated phytochemicals in nanoparticles or other novel drug delivery agents with currently accepted endocrine and/or conventional chemotherapies. Cell signaling, regulation of cell cycles, oxidative stress action, and inflammation could be positively impacted by phytoconstituents. They have the ability to alter non-coding RNAs, to prevent the proliferation and regeneration of cancer cells. The availability of novel approaches helps in disease targeting, safety, effectiveness and efficacy. The current literature helps to know the available drugs i.e. phytoconstituents or novel drug delivery like nanoparticle, microsphere, micelles, liposomes and neosomes. The literature has been taken from PubMed, Google Scholar, SciFinder, or other internet sites.


Breast Neoplasms , Phytochemicals , Humans , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Female , Phytochemicals/pharmacology , Phytochemicals/therapeutic use , Phytochemicals/chemistry , Drug Delivery Systems , Antineoplastic Agents, Phytogenic/therapeutic use , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/administration & dosage , Nanoparticles/chemistry
16.
Future Cardiol ; 19(3): 127-135, 2023 03.
Article En | MEDLINE | ID: mdl-37334764

Paclitaxel drug-coated balloons (DCBs) have been shown to improve patency and lower revascularization rates compared with plain old balloon angioplasty. DCBs continue to evolve by improving balloon-coating techniques that minimize the quantity of particles washed off into the bloodstream while maximizing drug retention and vascular-healing profile. Against this backdrop, it is clear that the future of antiproliferatives for the superficial femoral artery will focus on enhancements in device coating materials that will improve the efficiency of drug delivery. The Ranger DCB system recently gained US FDA approval for use. This review discusses the background of DCBs and how the Ranger DCB builds on these previous platforms based on experimental and clinical data.


Drug-coated balloons are medical devices used to open blocked arteries (a procedure called angioplasty) in patients who have atherosclerotic disease. The drug coating is provided to help keep the arteries open after treatment with the balloon. This is thought to occur through several mechanisms. In this review, we discuss recent advances in technology related to drug-coated balloons focusing on the recently introduced Ranger drug-coated Balloon.


Angioplasty, Balloon , Antineoplastic Agents, Phytogenic , Paclitaxel , Peripheral Arterial Disease , Drug Delivery Systems , Humans , Femoral Artery , Coated Materials, Biocompatible , Paclitaxel/administration & dosage , Paclitaxel/therapeutic use , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/therapeutic use , Peripheral Arterial Disease/drug therapy
17.
Life Sci ; 296: 120435, 2022 May 01.
Article En | MEDLINE | ID: mdl-35247437

AIMS: Glioblastoma (GBM) is the most common and deadliest type of brain cancer, and the current therapeutic options are not curative, imposing the need for novel strategies. Asiatic acid (AA) is a natural compound and has been explored due to its anti-glioma activity and lower toxicity to healthy tissues compared with conventional chemotherapeutic agents. However, its poor water-solubility is an obstacle for clinical application. Poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) were proposed in this work for Asiatic acid (AA) delivery. MAIN METHODS: A central composite design was implemented to optimize the NPs, and their surface was further modified with transferrin (Tf), for targeted delivery to GBM cells. The anti-glioma activity of the NPs was studied in vitro using human GBM cells and immortalized human astrocytes. KEY FINDINGS: The NPs exhibited a mean size smaller than 200 nm, with low polydispersity and negative zeta potential, indicating their suitability for brain tumor delivery. The NPs also exhibited high encapsulation efficiency and maintained a slow and controlled release of AA for 20 days. In vitro cell studies showed that NPs were able to maintain the anti-glioma activity of the natural compound and that the surface modification with Tf molecules was able to increase the cellular uptake in GBM cells, enhancing their selectivity and decreasing toxicity in healthy cells. SIGNIFICANCE: Overall, this work provided guidance for designing brain-targeting delivery systems of natural compounds.


Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Nanoparticles/administration & dosage , Pentacyclic Triterpenes/administration & dosage , Transferrin/chemistry , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/pharmacology , Astrocytes/drug effects , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Drug Delivery Systems/methods , Drug Liberation , Glioblastoma/pathology , Humans , Nanoparticles/chemistry , Pentacyclic Triterpenes/pharmacokinetics , Pentacyclic Triterpenes/pharmacology , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Receptors, Transferrin/metabolism
18.
Sci Rep ; 12(1): 1658, 2022 02 01.
Article En | MEDLINE | ID: mdl-35105904

More than 10% of women diagnosed with breast cancer during reproductive age carry hereditary germline pathogenic variants in high-penetrance BRCA genes or in others genes involved in DNA repair mechanisms such as PALB2, BRIP or ATM. Anticancer treatments may have an additional negative impact on the ovarian reserve and subsequently on the fertility of young patients carrying such mutations. Recently, the combination of carboplatin and paclitaxel is being recommended to these BRCA-mutated patients as neoadjuvant therapy. However, the impact on the ovary is unknown. Here, we investigated their effect of on the ovarian reserve using mice carriers of BRCA1-interacting protein C-terminal helicase-1 (BRIP1) mutation that plays an important role in BRCA1-dependent DNA repair. Results revealed that the administration of carboplatin or paclitaxel did not affect the ovarian reserve although increased DNA double-strand breaks were observed with carboplatin alone. Co-administration of carboplatin and paclitaxel resulted in a significant reduction of the ovarian reserve leading to a lower IVF performance, and an activation of the PI3K-Pten pathway, irrespective of the genetic background. This study suggests that co-administration of carboplatin and paclitaxel induces cumulative ovarian damage and infertility but a heterozygote genetic predisposition for DNA damage related to BRCA1 gene function does not increase this risk.


Antineoplastic Agents, Phytogenic/adverse effects , Carboplatin/adverse effects , Fanconi Anemia Complementation Group Proteins/genetics , Genes, Tumor Suppressor , Germ-Line Mutation , Ovarian Reserve/drug effects , Paclitaxel/adverse effects , RNA Helicases/genetics , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Apoptosis/drug effects , Apoptosis/genetics , BRCA1 Protein/genetics , Breast Neoplasms/genetics , Carboplatin/administration & dosage , DNA Breaks, Double-Stranded/drug effects , DNA Repair/drug effects , DNA Repair/genetics , Embryonic Development/drug effects , Embryonic Development/genetics , Female , Fertilization in Vitro/methods , Heterozygote , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Ovarian Follicle/drug effects , Ovarian Follicle/metabolism , Paclitaxel/administration & dosage
19.
Pharm Biol ; 60(1): 417-426, 2022 Dec.
Article En | MEDLINE | ID: mdl-35175172

CONTEXT: Osthole shows antitumor effects in various tumours. Studies describing the effect of osthole on retinoblastoma (RB) are rare. OBJECTIVE: This study investigates the antitumor activity of osthole on RB. MATERIALS AND METHODS: RB cells were treated with different concentrations of osthole and then subjected to cell viability, colony formation, apoptosis, and western blot assays. The expression of hsa_circ_0007534 in RB tissues was determined by qRT-PCR. Hsa_circ_0007534 overexpression plasmid (oe-circ_0007534), miR-214-3p mimics and negative controls were transfected into RB cells to investigate cell viability. Athymic nude mice were injected with Y-79 cells to establish subcutaneous RB models. These mice were treated with osthole (0.5 mmol/kg) or corn oil for 36 days. Tumour tissues were collected for further analysis. RESULTS: Osthole inhibited cell viability of RB cells with an IC50 of 200 µM for 24 h treatment and 120 µM for 48 h treatment, respectively. Hsa_circ_0007534 was increased significantly in RB tissues as compared to the matched nontumor tissues (p < 0.001). Oe-circ_0007534 counteracted the inhibitory effect of osthole on cell viability and colony numbers of Y-79 cells (p < 0.01). In vivo experiments indicated osthole significantly decreased the expression of hsa_circ_0007534 (p < 0.01) and increased the level of miR-214-3p in vivo. Furthermore, as compared to the control, osthole decreased the ratios of p-PI3K/PI3K, p-AKT/AKT and p-mTOR/mTOR (p < 0.01). However, hsa_circ_0007534 overexpression reversed the effect of osthole on the PI3K/AKT/mTOR pathway. DISCUSSION AND CONCLUSIONS: Osthole exhibited an antitumour effect in RB, providing a scientific basis for further research and clinical applications of osthole in RB treatment.


Antineoplastic Agents, Phytogenic/pharmacology , Coumarins/pharmacology , MicroRNAs/genetics , Retinoblastoma/drug therapy , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Apoptosis/drug effects , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Coumarins/administration & dosage , Humans , Male , Mice , Mice, Nude , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Retinoblastoma/genetics , Retinoblastoma/pathology , TOR Serine-Threonine Kinases/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
20.
Gene ; 823: 146275, 2022 May 20.
Article En | MEDLINE | ID: mdl-35189245

BACKGROUND: Silibinin, the principal flavonoid derived from milk thistle seeds, has been demonstrated to have strong inhibitory effects against human malignancies. The inhibitory function of silibinin on ovarian cancer, however, is not fully identified. In this essay, both in vivo and in vitro investigations were conducted to survey the silibinin's blocking effects on ovarian cancer. METHODS: The impacts of silibinin on two ovarian cancer cell lines, SKOV-3 and A2870, were determined by evaluating cell viability, migration, invasion, and apoptosis. Q-RT-PCR and western blotting techniques were carried out to explore the protein levels of signaling pathway markers. A mouse xenograft model was utilized to determine the silibinin efficacy in inhibiting tumor growth. RESULTS: After cell treatment with silibinin, cell viability, migration, and invasion were appreciably inhibited in cancer cell lines, but cell apoptosis was promoted. Also, silibinin reversed the epithelial-mesenchymal transition (EMT) mechanism by inducing E-cadherin expression and reducing N-cadherin and vimentin expression, suppressing the levels of regulators related to EMT such as Snail, Slug, and ZEB1 transcription factors, and also decreasing PI3K/AKT, Smad2/3, and ß-catenin intermediate molecules in vitro. Silibinin effectively ameliorated tumor growth in vivo. CONCLUSION: silibinin could be considered a potent agent against ovarian cancer based on the results.


Antineoplastic Agents, Phytogenic/administration & dosage , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Ovarian Neoplasms/drug therapy , Silybin/administration & dosage , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Epithelial-Mesenchymal Transition/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Neoplasm Invasiveness , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Silybin/pharmacology , Xenograft Model Antitumor Assays
...