Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 792
Filter
1.
Int J Nanomedicine ; 19: 8417-8436, 2024.
Article in English | MEDLINE | ID: mdl-39176130

ABSTRACT

Purpose: Docetaxel (DTX) is a valuable anti-tumor chemotherapy drug with limited oral bioavailability. This study aims to develop an effective oral delivery system for DTX using natural nanoparticles (Nnps) derived from Coptidis Rhizoma extract. Methods: DTX-loaded self-assembled nanoparticles (Nnps-DTX) were created using an optimized heat-induction strategy. Nnps-DTX's shape, size, Zeta potential, and in vitro stability were all carefully examined. Additionally, the study investigated the encapsulation efficiency, loading capacity, crystal form, and intermolecular interactions of DTX in Nnps-DTX. Subsequently, the solubility, release, cellular uptake, metabolic stability, and preclinical pharmacokinetics of DTX in Nnps-DTX were systematically evaluated. Finally, the cytotoxicity of Nnps-DTX was assessed in three tumor cell lines. Results: Nnps-DTX was spherical in shape, 138.6 ± 8.2 nm in size, with a Zeta potential of -20.8 ± 0.6 mV, a DTX encapsulation efficiency of 77.6 ± 8.5%, and a DTX loading capacity of 6.8 ± 1.9%. Hydrogen bonds, hydrophobic interactions, and electrostatic interactions were involved in the formation of Nnps-DTX. DTX within Nnps-DTX was in an amorphous form, resulting in enhanced solubility (23.3 times) and release compared to free DTX. Following oral treatment, the mice in the Nnps-DTX group had DTX peak concentrations 8.8, 23.4, 44.6, and 5.7 times higher in their portal vein, systemic circulation, liver, and lungs than the mice in the DTX group. Experiments performed in Caco-2 cells demonstrated a significant increase in DTX uptake by Nnps-DTX compared to free DTX, which was significantly inhibited by indomethacin, an inhibitor of caveolae-mediated endocytosis. Furthermore, compared to DTX, DTX in Nnps-DTX demonstrated better metabolic stability in liver microsomes. Notably, Nnps-DTX significantly reduced the viability of MCF-7, HCT116, and HepG2 cells. Conclusion: The novel self-assembled nanoparticles considerably enhanced the cellular absorption, solubility, release, metabolic stability, and pharmacokinetics of oral DTX and demonstrated strong cytotoxicity against tumor cell lines.


Subject(s)
Docetaxel , Nanoparticles , Animals , Docetaxel/pharmacokinetics , Docetaxel/chemistry , Docetaxel/pharmacology , Docetaxel/administration & dosage , Humans , Administration, Oral , Nanoparticles/chemistry , Drugs, Chinese Herbal/pharmacokinetics , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/administration & dosage , Drugs, Chinese Herbal/pharmacology , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage , Mice , Cell Line, Tumor , Coptis chinensis , Particle Size , Male , Drug Liberation , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Cell Survival/drug effects , Biological Availability , Solubility , Rats, Sprague-Dawley , Mice, Inbred BALB C
2.
Sci Rep ; 14(1): 19075, 2024 08 17.
Article in English | MEDLINE | ID: mdl-39154055

ABSTRACT

Human induced pluripotent stem cell-derived sensory neuron (iPSC-dSN) models are a valuable resource for the study of neurotoxicity but are affected by poor replicability and reproducibility, often due to a lack of optimization. Here, we identify experimental factors related to culture conditions that substantially impact cellular drug response in vitro and determine optimal conditions for improved replicability and reproducibility. Treatment duration and cell seeding density were both found to be significant factors, while cell line differences also contributed to variation. A replicable dose-response in viability was demonstrated after 48-h exposure to docetaxel or paclitaxel. Additionally, a replicable dose-dependent reduction in neurite outgrowth was demonstrated, demonstrating the applicability of the model for the examination of additional phenotypes. Overall, we have established an optimized iPSC-dSN model for the study of taxane-induced neurotoxicity.


Subject(s)
Cell Survival , Induced Pluripotent Stem Cells , Sensory Receptor Cells , Taxoids , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/cytology , Taxoids/pharmacology , Sensory Receptor Cells/drug effects , Cell Survival/drug effects , Docetaxel/pharmacology , Neurotoxicity Syndromes/etiology , Bridged-Ring Compounds/pharmacology , Cell Differentiation/drug effects , Paclitaxel/pharmacology , Paclitaxel/toxicity , Cell Line , Cells, Cultured
3.
Int J Nanomedicine ; 19: 8603-8620, 2024.
Article in English | MEDLINE | ID: mdl-39188859

ABSTRACT

Background: Chemotherapeutic drugs have some drawbacks in antineoplastic therapy, mainly containing seriously toxic side effects caused by injection and multi-drug resistance (MDR). Co-delivery with two or more drugs via nanomicelles is a promising strategy to solve these problems. Oral chemotherapy is increasingly preferred owing to its potential to enhance the life quality of patients. Methods and Results: The study intended to develop mixed micelles using D-α-Tocopherol poly(ethylene glycol) 1000 succinate (TPGS) and soluplus for the co-encapsulation of docetaxel (DTX) and curcumin (CUR), marked as (DTX+CUR)-loaded mixed micelles, treating drug-resistant breast cancer by oral administration. The (DTX+CUR)-loaded mixed micelles had a uniform particle size (~64 nm), high drug loading and encapsulation efficiency, in vitro sustained-release properties and good pH-dependent stability. In vitro cell study, the (DTX+CUR)-loaded mixed micelles displayed the highest cellular uptake, cytotoxicity, cell apoptosis-inducing rates and cell ROS-inducing levels on MCF-7/Adr cells. Notably, in vivo pharmacokinetic studies, (DTX+CUR)-loaded mixed micelles enhanced markedly the oral absorption of DTX compared to pure DTX, with a relative oral bioavailability of 574%. The (DTX+CUR)-loaded mixed micelles by oral administration had the same anticancer efficacy as taxotere by injection in resistant breast cancer bearing mice. Conclusion: (DTX+CUR)-loaded mixed micelles could provide a potential formulation for treating drug-resistant breast cancers by oral administration.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Curcumin , Docetaxel , Drug Resistance, Neoplasm , Micelles , Polyethylene Glycols , Curcumin/pharmacokinetics , Curcumin/chemistry , Curcumin/administration & dosage , Curcumin/pharmacology , Docetaxel/pharmacokinetics , Docetaxel/administration & dosage , Docetaxel/chemistry , Docetaxel/pharmacology , Humans , Female , Animals , Breast Neoplasms/drug therapy , Administration, Oral , Drug Resistance, Neoplasm/drug effects , MCF-7 Cells , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacokinetics , Antineoplastic Agents/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Vitamin E/chemistry , Vitamin E/administration & dosage , Vitamin E/pharmacokinetics , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Polyvinyls/chemistry , Polyvinyls/pharmacokinetics , Polyvinyls/administration & dosage , Mice , Mice, Inbred BALB C , Particle Size , Taxoids/pharmacokinetics , Taxoids/administration & dosage , Taxoids/chemistry , Taxoids/pharmacology , Drug Liberation , Rats, Sprague-Dawley
4.
ACS Appl Mater Interfaces ; 16(34): 44528-44537, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39155662

ABSTRACT

The high level of accumulation of therapeutic agents in tumors is crucial for cancer treatment. Compared to the passive tumor-targeting effect, active tumor-targeting delivery systems, primarily mediated by peptides with high production costs and reduced circulation time, are highly desired. Platelet-driven technologies have opened new avenues for targeted drug delivery prevalently through a membrane coating strategy that involves intricate manufacturing procedures or the fucoidan-mediated hitchhiking method with limited platelet affinity. Here, a novel type of amphiphilic glycopolymer self-assembled micellar nanoparticle has been developed to adhere to naturally activated platelets in the blood. The simultaneous integration of fucose and sialic acid segments into glycopolymers enables closer mimicry of the structure of P-selectin glycoprotein ligand-1 (PSGL-1), thereby increasing the affinity for activated platelets. It results in the formation of glycopolymeric micelle-platelet hybrids, facilitating targeted drug delivery to tumors. The selective platelet-assisted cellular uptake of docetaxel (DTX)-loaded glycopolymeric micelles leads to lower IC50 values against 4T1 cells than that of free DTX. The directed tumor-targeting effect of activated platelets has significantly improved the tumor accumulation capacity of the glycopolymeric nanoparticles, with up to 21.0% found in tumors within the initial 0.2 h. Additionally, with acid-responsive drug release and inherent antimetastasis properties, the glycopolymeric nanoparticles ensured potent therapeutic efficacy, prolonged survival time, and reduced cardiotoxicity, presenting a new and unexplored strategy for platelet-directed drug delivery to tumors, showing promising prospects in treating localized tumors and preventing tumor metastasis.


Subject(s)
Blood Platelets , Docetaxel , Micelles , Nanoparticles , Docetaxel/chemistry , Docetaxel/pharmacology , Docetaxel/pharmacokinetics , Docetaxel/therapeutic use , Animals , Blood Platelets/metabolism , Blood Platelets/drug effects , Nanoparticles/chemistry , Mice , Cell Line, Tumor , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Mice, Inbred BALB C , Humans , Female , Drug Delivery Systems , Drug Carriers/chemistry , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/metabolism
5.
Cell Death Dis ; 15(8): 558, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39090086

ABSTRACT

Prostate cancer (PCa) is the second leading cause of cancer-related death in American men. PCa that relapses after hormonal therapies, referred to as castration resistant PCa (CRPC), often presents with metastases (mCRPC) that are the major cause of mortality. The few available therapies for mCRPC patients include taxanes docetaxel (DTX) and cabazitaxel (CBZ). However, development of resistance limits their clinical use. Mechanistically, resistance arises through upregulation of multidrug resistance (MDR) proteins such as MDR1/ABCB1, making ABCB1 an attractive therapeutic target. Yet, ABCB1 inhibitors failed to be clinically useful due to low specificity and toxicity issues. To study taxanes resistance, we produced CBZ resistant C4-2B cells (RC4-2B) and documented resistance to both CBZ and DTX in cell culture and in 3D prostaspheres settings. RNAseq identified increased expression of ABCB1 in RC4-2B, that was confirmed by immunoblotting and immunofluorescent analysis. ABCB1-specific inhibitor elacridar reversed CBZ and DTX resistance in RC4-2B cells, confirming ABCB1-mediated resistance mechanism. In a cell-based screen using a curated library of cytotoxic drugs, we found that DNA damaging compounds Camptothecin (CPT) and Cytarabine (Ara-C) overcame resistance as seen by similar cytotoxicity in parental C4-2B and resistant RC4-2B. Further, these compounds were cytotoxic to multiple PC cells resistant to taxanes with high ABCB1 expression and, therefore, can be used to conquer the acquired resistance to taxanes in PCa. Finally, inhibition of cyclin-dependent kinases 4/6 (CDK4/6) with small molecule inhibitors (CDK4/6i) potentiated cytotoxic effect of CPT or Ara-C in both parental and resistant cells. Overall, our findings indicate that DNA damaging agents CPT and Ara-C alone or in combination with CDK4/6i can be suggested as a new treatment regimen in CRPC patients, including those that are resistant to taxanes.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B , Docetaxel , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Prostatic Neoplasms, Castration-Resistant , Taxoids , Humans , Male , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/pathology , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/genetics , Drug Resistance, Neoplasm/drug effects , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily B/genetics , Cell Line, Tumor , Docetaxel/pharmacology , Drug Resistance, Multiple/drug effects , Taxoids/pharmacology , Taxoids/therapeutic use , Antineoplastic Agents/pharmacology
6.
J Med Chem ; 67(16): 14370-14393, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39102508

ABSTRACT

Myeloid cell leukemia 1 (Mcl-1) is a key regulator of the intrinsic apoptosis pathway. Overexpression of Mcl-1 is correlated with high tumor grade, poor survival, and both intrinsic and acquired resistance to cancer therapies. Herein, we disclose the structure-guided design of a small molecule Mcl-1 inhibitor, compound 26, that binds to Mcl-1 with subnanomolar affinity, inhibits growth in cell culture assays, and possesses low clearance in mouse and dog pharmacokinetic (PK) experiments. Evaluation of 26 as a single agent in Mcl-1 sensitive hematological and solid tumor xenograft models resulted in regressions. Co-treatment of Mcl-1-sensitive and Mcl-1 insensitive lung cancer derived xenografts with 26 and docetaxel or topotecan, respectively, resulted in an enhanced tumor response. These findings support the premise that pro-apoptotic priming of tumor cells by other therapies in combination with Mcl-1 inhibition may significantly expand the subset of cancers in which Mcl-1 inhibitors may prove beneficial.


Subject(s)
Antineoplastic Agents , Myeloid Cell Leukemia Sequence 1 Protein , Xenograft Model Antitumor Assays , Animals , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Humans , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Dogs , Structure-Activity Relationship , Female , Drug Discovery , Taxoids/pharmacology , Taxoids/pharmacokinetics , Taxoids/therapeutic use , Taxoids/chemistry , Docetaxel/pharmacology , Docetaxel/therapeutic use , Docetaxel/pharmacokinetics , Docetaxel/chemistry
7.
Cell Death Dis ; 15(8): 617, 2024 Aug 25.
Article in English | MEDLINE | ID: mdl-39183332

ABSTRACT

Resistance to the current Androgen Receptor Signaling Inhibitor (ARSI) therapies has led to higher incidences of therapy-induced neuroendocrine-like prostate cancer (t-NEPC). This highly aggressive subtype with predominant small-cell-like characteristics is resistant to taxane chemotherapies and has a dismal overall survival. t-NEPCs are mostly treated with platinum-based drugs with a combination of etoposide or taxane and have less selectivity and high systemic toxicity, which often limit their clinical potential. During t-NEPC transformation, adenocarcinomas lose their luminal features and adopt neuro-basal characteristics. Whether the adaptive neuronal characteristics of t-NEPC are responsible for such taxane resistance remains unknown. Pathway analysis from patient gene-expression databases indicates that t-NEPC upregulates various neuronal pathways associated with enhanced cellular networks. To identify transcription factor(s) (TF) that could be important for promoting the gene expression for neuronal characters in t-NEPC, we performed ATAC-Seq, acetylated-histone ChIP-seq, and RNA-seq in our NE-like cell line models and analyzed the promoters of transcriptionally active and significantly enriched neuroendocrine-like (NE-like) cancer-specific genes. Our results indicate that Pax5 could be an important transcription factor for neuronal gene expression and specific to t-NEPC. Pathway analysis revealed that Pax5 expression is involved in axonal guidance, neurotransmitter regulation, and neuronal adhesion, which are critical for strong cellular communications. Further results suggest that depletion of Pax5 disrupts neurite-mediated cellular communication in NE-like cells and reduces surface growth factor receptor activation, thereby, sensitizing them to docetaxel therapies. Moreover, t-NEPC-specific hydroxymethylation of Pax5 promoter CpG islands favors Pbx1 binding to induce Pax5 expression. Based on our study, we concluded that continuous exposure to ARSI therapies leads to epigenetic modifications and Pax5 activation in t-NEPC, which promotes the expression of genes necessary to adopt taxane-resistant NE-like cancer. Thus, targeting the Pax5 axis can be beneficial for reverting their taxane sensitivity.


Subject(s)
Docetaxel , Drug Resistance, Neoplasm , PAX5 Transcription Factor , Prostatic Neoplasms , Humans , Male , Docetaxel/pharmacology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Cell Line, Tumor , PAX5 Transcription Factor/metabolism , PAX5 Transcription Factor/genetics , Gene Expression Regulation, Neoplastic/drug effects , Antineoplastic Agents/pharmacology , Carcinoma, Neuroendocrine/metabolism , Carcinoma, Neuroendocrine/drug therapy , Carcinoma, Neuroendocrine/pathology , Carcinoma, Neuroendocrine/genetics , Promoter Regions, Genetic/genetics , Receptors, Androgen/metabolism , Receptors, Androgen/genetics
8.
Biomed Mater ; 19(6)2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39208838

ABSTRACT

The invasion and metastasis of tumors pose significant challenges in the treatment of ovarian cancer (OC), making it difficult to cure. One potential treatment approach that has gained attention is the use of matrix metalloproteinase reactive controlled release micelle preparations. In this study, we developed a novel PEG5000-PVGLIG-hyaluronic acid docetaxel/bakuchiol (PP-HA-DTX/BAK) micelles formulation with desirable characteristics such as particle size, narrow polydispersity index, and a ZETA potential of approximately -5 mV. The surface modification with HA facilitates tumor penetration into the tumor interior, while the incorporation of DSPE-PEG2000-PVGLIG-PEG5000helps conceal DSPE-PEG2000-HA, reducing off-target effects and prolonging drug circulation timein vivo. Bothin vitroandin vivoexperiments demonstrated that these micelles effectively inhibit proliferation, invasion, and metastasis of OC cells while promoting apoptosis. Therefore, our findings suggest that PP-HA-DTX/BAK micelles represent a safe and effective therapeutic strategy for treating OC.


Subject(s)
Docetaxel , Micelles , Neoplasm Invasiveness , Ovarian Neoplasms , Phenols , Polyethylene Glycols , Docetaxel/chemistry , Docetaxel/pharmacology , Docetaxel/administration & dosage , Female , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Humans , Animals , Cell Line, Tumor , Polyethylene Glycols/chemistry , Phenols/chemistry , Phenols/pharmacology , Mice , Apoptosis/drug effects , Hyaluronic Acid/chemistry , Taxoids/chemistry , Taxoids/pharmacology , Taxoids/administration & dosage , Cell Proliferation/drug effects , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage , Mice, Nude , Particle Size , Mice, Inbred BALB C , Neoplasm Metastasis , Drug Carriers/chemistry
9.
Biol Pharm Bull ; 47(8): 1437-1446, 2024.
Article in English | MEDLINE | ID: mdl-39143009

ABSTRACT

Bisdemethoxycurcumin (BDMC) is one of major forms of curcuminoids found in the rhizomes of turmeric. Docetaxel (DTX) is the standard of care for men diagnosed with androgen-independent prostate cancers. Here we report for the first time that BDMC could reinforce the effect of DTX against prostate cancer in vitro and in vivo. In vitro study, PC3 and LNCaP cells were cultured and treated with BDMC and DTX alone or in combination. The effects on cell viability were determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Apoptosis was assessed by annexin V/propidium iodide (PI) staining, while cell cycle was assessed by PI staining. Bax, Bcl-2, caspase, poly(ADP-ribose)polymerase (PARP), cyclin B1 and CDK1 expression were assayed by Western blot. We found that a combination treatment of BDMC (10 µM) with DTX (10 nM) was more effective in the inhibition of PC3 and LNCaP cell growth and induction of apoptosis as well as G2/M arrest, which is accompanied with the significant inhibition of Bcl-2, cyclin B1, CDK1 expression and significant increase of Bax, cleaved caspase-9, cleaved caspase-3 and cleaved PARP, than those by treatment of BDMC or DTX alone. Moreover, in vivo evaluation further demonstrated the superior anticancer efficacy of BDMC and DTX compared to DTX alone in a murine prostate cancer model. These results suggest that BDMC can be an attractive therapeutic candidate in enhancing the efficacy of DTX in prostate cancer treatment.


Subject(s)
Antineoplastic Agents , Apoptosis , Diarylheptanoids , Docetaxel , Prostatic Neoplasms , Male , Diarylheptanoids/pharmacology , Diarylheptanoids/therapeutic use , Humans , Animals , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Prostatic Neoplasms/metabolism , Docetaxel/pharmacology , Docetaxel/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Drug Synergism , Cyclin B1/metabolism , Mice, Nude , Xenograft Model Antitumor Assays , Mice , Curcumin/analogs & derivatives , Curcumin/pharmacology , Curcumin/therapeutic use , Cell Survival/drug effects , Taxoids/pharmacology , Taxoids/therapeutic use , Proto-Oncogene Proteins c-bcl-2/metabolism , Mice, Inbred BALB C , Poly(ADP-ribose) Polymerases/metabolism , CDC2 Protein Kinase/metabolism
10.
J Gene Med ; 26(8): e3725, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39134478

ABSTRACT

INTRODUCTION: Esophageal cancer is one of the major cancers in China. Most patients with esophageal cancer are diagnosed at an advanced stage, and the 5 year survival rate is discouraging. Combined chemotherapy is a common method for the treatment of esophageal cancer. METHODS: In this study, distearoyl phosphatidyl ethanolamine polyethylene glycol 2000 (DSPE-PEG2000) nanoliposomes (NLPs) encapsulating the anticancer drugs docetaxel (DOX) and oridonin (ORD) were prepared, and their ability to enhance the release of anticancer drugs was determined. The NLP system was characterized by transmission electron microscopy, particle size and encapsulation efficiency. In addition, the release characteristics and pharmacodynamics of these drugs were also studied in detail. RESULTS: When the DOX/ORD ratio was 2:1, the higher proportion of DOX led to a stronger synergy effect. DOX/ORD NLPs were prepared by the high-pressure homogenization method and had a uniform spherical morphology. The mean particle size and polydispersity index were determined to be 246.4 and 0.163, respectively. The stability results showed that no significant change was observed in particle size, zeta potential, Encapsulation efficiency and dynamic light scattering for DOX/ORD NLPs during the observation period. The results of in vitro release illustrated that the acidic environment of tumor might be beneficial to drug release. The three-dimensional tumorsphere showed that DOX/ORD NLPs can reach the interior of tumor spheres, which destroys the structure of cells, resulting in irregular spherical tumor spheres. The in vivo study results indicated that DOX/ORD NLPs had an obvious targeting effect on subcutaneous tumors and have the potential to actively deliver drugs to tumor tissues. Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining was used to detect apoptosis. The results showed that DOX/ORD NLP treatment could significantly induce apoptosis and inhibit tumor growth. CONCLUSION: The DOX/ORD NLPs prepared in this study can enhance the anti-tumor activity, and are expected to be a promising co-delivery platform for the treatment of esophageal cancer.


Subject(s)
Diterpenes, Kaurane , Docetaxel , Esophageal Neoplasms , Liposomes , Diterpenes, Kaurane/pharmacology , Diterpenes, Kaurane/chemistry , Diterpenes, Kaurane/administration & dosage , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/pathology , Docetaxel/pharmacology , Docetaxel/administration & dosage , Docetaxel/chemistry , Liposomes/chemistry , Animals , Humans , Cell Line, Tumor , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Nanoparticles/chemistry , Particle Size , Xenograft Model Antitumor Assays , Drug Liberation , Drug Delivery Systems/methods , Mice, Nude , Mice, Inbred BALB C , Nanoparticle Drug Delivery System/chemistry
11.
Eur J Pharm Biopharm ; 201: 114386, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38950717

ABSTRACT

The goal of the study was to fabricate folic acid functionalized docetaxel (DOC)/erlotinib (ERL)-loaded solid lipid nanoparticles (SLNs) to synergistically increase the anticancer activity against triple-negative breast cancer. DOC/ERL-SLNs were prepared by the high shear homogenization - ultrasound dispersion method (0.1 % w/v for DOC, and 0.3 %w/v for ERL) and optimized using Plackett Burman Design (PBD) followed by Box Behnken Design (BBD). The optimized SLNs demonstrated particle size < 200 nm, PDI < 0.35, and negative zeta potential with entrapment and loading efficiency of ∼80 and ∼4 %, respectively. The SLNs and folic acid functionalized SLNs (FA-SLNs) showed sustained release for both drugs, followed by Higuchi and Korsemeyer-Peppas drug release models, respectively. Further, the in vitro pH-stat lipolysis model demonstrated an approximately 3-fold increase in the bioaccessibility of drugs from SLNs compared to suspension. The TEM images revealed the spherical morphology of the SLNs. DOC/ERL loaded SLNs showed dose- and time-dependent cytotoxicity and exhibited a synergism at a molar ratio of 1:3 in TNBC with a combination index of 0.35 and 0.37, respectively. FA-DOC/ERL-SLNs showed enhanced anticancer activity as evidenced by MMP and ROS assay and further inhibited the colony-forming ability and the migration capacity of TNBC cells. Conclusively, the study has shown that SLNs are encouraging systems to improve the pharmaceutical attributes of poorly bioavailable drugs.


Subject(s)
Docetaxel , Drug Liberation , Drug Synergism , Erlotinib Hydrochloride , Lipids , Nanoparticles , Particle Size , Triple Negative Breast Neoplasms , Triple Negative Breast Neoplasms/drug therapy , Docetaxel/administration & dosage , Docetaxel/pharmacology , Docetaxel/pharmacokinetics , Humans , Nanoparticles/chemistry , Erlotinib Hydrochloride/administration & dosage , Erlotinib Hydrochloride/pharmacology , Erlotinib Hydrochloride/pharmacokinetics , Cell Line, Tumor , Female , Lipids/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Drug Carriers/chemistry , Cell Survival/drug effects , Folic Acid/chemistry , Liposomes
12.
PLoS One ; 19(7): e0306969, 2024.
Article in English | MEDLINE | ID: mdl-38990953

ABSTRACT

Docetaxel (Doc) plays a crucial role in clinical antineoplastic practice. However, it is continuously documented that tumors frequently develop chemoresistance and relapse, which may be related to polyploid giant cancer cells (PGCCs). The aim of this study was investigate the formation mechanism and biological behavior of PGCCs induced by Doc. Ovarian cancer cells were treated with Doc, and then the effect of Doc on cellular viability was evaluated by MTT assay and microscopic imaging analysis. The biological properties of PGCCs were further evaluated by Hoechst 33342 staining, cell cycle and DNA content assay, DNA damage response (DDR) signaling detection, ß-galactosidase staining, mitochondrial membrane potential detection, and reverse transcription-quantitative polymerase chain reaction. The results indicated that Doc reduced cellular viability; however, many cells were still alive, and were giant and polyploid. Doc increased the proportion of cells stayed in the G2/M phase and reduced the number of cells. In addition, the expression of γ-H2A.X was constantly increased after Doc treatment. PGCCs showed senescence-associated ß-galactosidase activity and an increase in the monomeric form of JC-1. The mRNA level of octamer-binding transcription factor 4 (OCT4) and krüppel-like factor 4 (KLF4) was significantly increased in PGCCs. Taken together, our results suggest that Doc induces G2/M cell cycle arrest, inhibits the proliferation and activates persistent DDR signaling to promote the formation of PGCCs. Importantly, PGCCs exhibit a senescence phenotype and express stem cell markers.


Subject(s)
Cellular Senescence , Docetaxel , Kruppel-Like Factor 4 , Neoplastic Stem Cells , Ovarian Neoplasms , Polyploidy , Humans , Docetaxel/pharmacology , Female , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/genetics , Cellular Senescence/drug effects , Cell Line, Tumor , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Octamer Transcription Factor-3/metabolism , Octamer Transcription Factor-3/genetics , Giant Cells/drug effects , Giant Cells/metabolism , Antineoplastic Agents/pharmacology , Phenotype , Cell Survival/drug effects , Membrane Potential, Mitochondrial/drug effects , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Taxoids/pharmacology , DNA Damage/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics
13.
Int J Mol Sci ; 25(13)2024 Jun 26.
Article in English | MEDLINE | ID: mdl-39000112

ABSTRACT

Androgen-receptor-negative, androgen-independent (ARneg-AI) prostate cancer aggressively proliferates and metastasizes, which makes treatment difficult. Hence, it is necessary to continue exploring cancer-associated markers, such as oncofetal Receptor Tyrosine Kinase like Orphan Receptor 1 (ROR1), which may serve as a form of targeted prostate cancer therapy. In this study, we identify that Penta-O-galloyl-ß-D-glucose (PGG), a plant-derived gallotannin small molecule inhibitor, modulates ROR1-mediated oncogenic signaling and mitigates prostate cancer phenotypes. Results indicate that ROR1 protein levels were elevated in the highly aggressive ARneg-AI PC3 cancer cell line. PGG was selectively cytotoxic to PC3 cells and induced apoptosis of PC3 (IC50 of 31.64 µM) in comparison to normal prostate epithelial RWPE-1 cells (IC50 of 74.55 µM). PGG was found to suppress ROR1 and downstream oncogenic pathways in PC3 cells. These molecular phenomena were corroborated by reduced migration, invasion, and cell cycle progression of PC3 cells. PGG minimally and moderately affected RWPE-1 and ARneg-AI DU145, respectively, which may be due to these cells having lower levels of ROR1 expression in comparison to PC3 cells. Additionally, PGG acted synergistically with the standard chemotherapeutic agent docetaxel to lower the IC50 of both compounds about five-fold (combination index = 0.402) in PC3 cells. These results suggest that ROR1 is a key oncogenic driver and a promising target in aggressive prostate cancers that lack a targetable androgen receptor. Furthermore, PGG may be a selective and potent anti-cancer agent capable of treating ROR1-expressing prostate cancers.


Subject(s)
Cell Proliferation , Glycogen Synthase Kinase 3 beta , Hydrolyzable Tannins , Prostatic Neoplasms , Proto-Oncogene Proteins c-akt , Receptor Tyrosine Kinase-like Orphan Receptors , Signal Transduction , Humans , Male , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Hydrolyzable Tannins/pharmacology , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Signal Transduction/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Apoptosis/drug effects , Antineoplastic Agents/pharmacology , Cell Movement/drug effects , PC-3 Cells , Gene Expression Regulation, Neoplastic/drug effects , Docetaxel/pharmacology
14.
Oncol Rep ; 52(2)2024 Aug.
Article in English | MEDLINE | ID: mdl-38963044

ABSTRACT

Lysine methyltransferase 5A (KMT5A) is the sole mammalian enzyme known to catalyse the mono­methylation of histone H4 lysine 20 and non­histone proteins such as p53, which are involved in the occurrence and progression of numerous cancers. The present study aimed to determine the function of KMT5A in inducing docetaxel (DTX) resistance in patients with breast carcinoma by evaluating glucose metabolism and the underlying mechanism involved. The upregulation or downregulation of KMT5A­related proteins was examined after KMT5A knockdown in breast cancer (BRCA) cells by Tandem Mass Tag proteomics. Through differential protein expression and pathway enrichment analysis, the upregulated key gluconeogenic enzyme fructose­1,6­bisphosphatase 1 (FBP1) was discovered. Loss of FBP1 expression is closely related to the development and prognosis of cancers. A dual­luciferase reporter gene assay confirmed that KMT5A inhibited the expression of FBP1 and that overexpression of FBP1 could enhance the chemotherapeutic sensitivity to DTX through the suppression of KMT5A expression. The KMT5A inhibitor UNC0379 was used to verify that DTX resistance induced by KMT5A through the inhibition of FBP1 depended on the methylase activity of KMT5A. According to previous literature and interaction network structure, it was revealed that KMT5A acts on the transcription factor twist family BHLH transcription factor 1 (TWIST1). Then, it was verified that TWSIT1 promoted the expression of FBP1 by using a dual­luciferase reporter gene experiment. KMT5A induces chemotherapy resistance in BRCA cells by promoting cell proliferation and glycolysis. After the knockdown of the KMT5A gene, the FBP1 related to glucose metabolism in BRCA was upregulated. KMT5A knockdown expression and FBP1 overexpression synergistically inhibit cell proliferation and block cells in the G2/M phase. KMT5A inhibits the expression of FBP1 by methylating TWIST1 and weakening its promotion of FBP1 transcription. In conclusion, KMT5A was shown to affect chemotherapy resistance by regulating the cell cycle and positively regulate glycolysis­mediated chemotherapy resistance by inhibiting the transcription of FBP1 in collaboration with TWIST1. KMT5A may be a potential therapeutic target for chemotherapy resistance in BRCA.


Subject(s)
Breast Neoplasms , Docetaxel , Drug Resistance, Neoplasm , Fructose-Bisphosphatase , Gene Expression Regulation, Neoplastic , Nuclear Proteins , Twist-Related Protein 1 , Humans , Breast Neoplasms/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm/genetics , Female , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Fructose-Bisphosphatase/genetics , Fructose-Bisphosphatase/metabolism , Docetaxel/pharmacology , Cell Line, Tumor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Cell Proliferation/drug effects , DNA Methylation
15.
Cell Rep ; 43(7): 114431, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38968071

ABSTRACT

Bromodomain-containing protein 4 (BRD4) has emerged as a promising therapeutic target in prostate cancer (PCa). Understanding the mechanisms of BRD4 stability could enhance the clinical response to BRD4-targeted therapy. In this study, we report that BRD4 protein levels are significantly decreased during mitosis in a PLK1-dependent manner. Mechanistically, we show that BRD4 is primarily phosphorylated at T1186 by the CDK1/cyclin B complex, recruiting PLK1 to phosphorylate BRD4 at S24/S1100, which are recognized by the APC/CCdh1 complex for proteasome pathway degradation. We find that PLK1 overexpression lowers SPOP mutation-stabilized BRD4, consequently rendering PCa cells re-sensitized to BRD4 inhibitors. Intriguingly, we report that sequential treatment of docetaxel and JQ1 resulted in significant inhibition of PCa. Collectively, the results support that PLK1-phosphorylated BRD4 triggers its degradation at M phase. Sequential treatment of docetaxel and JQ1 overcomes BRD4 accumulation-associated bromodomain and extra-terminal inhibitor (BETi) resistance, which may shed light on the development of strategies to treat PCa.


Subject(s)
Azepines , Cell Cycle Proteins , Docetaxel , Drug Resistance, Neoplasm , Mitosis , Polo-Like Kinase 1 , Prostatic Neoplasms , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins , Transcription Factors , Triazoles , Humans , Cell Cycle Proteins/metabolism , Male , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Phosphorylation , Proto-Oncogene Proteins/metabolism , Mitosis/drug effects , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Drug Resistance, Neoplasm/drug effects , Cell Line, Tumor , Azepines/pharmacology , Triazoles/pharmacology , Docetaxel/pharmacology , Proteolysis/drug effects , Nuclear Proteins/metabolism , Animals , CDC2 Protein Kinase/metabolism , Mice, Nude , Mice , Proteasome Endopeptidase Complex/metabolism , Bromodomain Containing Proteins , Repressor Proteins
16.
Int J Mol Sci ; 25(14)2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39063165

ABSTRACT

Human inflammatory breast cancer (IBC) and canine inflammatory mammary cancer (IMC) are highly aggressive neoplastic diseases that share numerous characteristics. In IBC and IMC, chemotherapy produces a limited pathological response and anti-androgen therapies have been of interest for breast cancer treatment. Therefore, the aim was to evaluate the effect of a therapy based on bicalutamide, a non-steroidal anti-androgen, with doxorubicin and docetaxel chemotherapy on cell proliferation, migration, tumor growth, and steroid-hormone secretion. An IMC-TN cell line, IPC-366, and an IBC-TN cell line, SUM149, were used. In vitro assays revealed that SUM149 exhibited greater sensitivity, reducing cell viability and migration with all tested drugs. In contrast, IPC-366 exhibited only significant in vitro reductions with docetaxel as a single agent or in different combinations. Decreased estrogen levels reduced in vitro tumor growth in both IMC and IBC. Curiously, doxorubicin resulted in low efficacy, especially in IMC. In addition, all drugs reduced the tumor volume in IBC and IMC by increasing intratumoral testosterone (T) levels, which have been related with reduced tumor progression. In conclusion, the addition of bicalutamide to doxorubicin and docetaxel combinations may represent a potential treatment for IMC and IBC.


Subject(s)
Anilides , Cell Proliferation , Docetaxel , Inflammatory Breast Neoplasms , Mammary Neoplasms, Animal , Nitriles , Tosyl Compounds , Tosyl Compounds/pharmacology , Humans , Animals , Female , Nitriles/pharmacology , Nitriles/therapeutic use , Cell Line, Tumor , Anilides/pharmacology , Dogs , Inflammatory Breast Neoplasms/drug therapy , Inflammatory Breast Neoplasms/pathology , Inflammatory Breast Neoplasms/metabolism , Cell Proliferation/drug effects , Docetaxel/pharmacology , Mammary Neoplasms, Animal/drug therapy , Mammary Neoplasms, Animal/pathology , Mammary Neoplasms, Animal/metabolism , Doxorubicin/pharmacology , Mice , Cell Survival/drug effects , Cell Movement/drug effects , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Xenograft Model Antitumor Assays , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Testosterone
17.
Anticancer Res ; 44(8): 3277-3285, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39060082

ABSTRACT

BACKGROUND/AIM: Lipids are essential for energy production, signaling, and membrane formation, hence increased lipid metabolism may lead to cancer growth. 4-cholesten-3-one (4Cone), a sterol metabolite, has various biological activities, including the inhibition of cancer growth. This study examined whether 4Cone could change the lipid profile of triple-negative breast cancer cells (MDA-MB-231) and whether in combination with the anti-cancer chemotherapy docetaxel (TXT) could further reduce cancer aggressiveness. MATERIALS AND METHODS: The effect of 4Cone, TXT, or their combination (4Cone/TXT) on migration and proliferation was examined utilizing the wound healing and MTT assays. The expression of the lipogenesis-related enzymes was assessed using RT-qPCR and lipid profile was examined using mass spectrometry. RESULTS: 4Cone and TXT individually reduced cell viability and migration of MDA-MB-231 cancer cells; however, their combination (4Cone/TXT) had a greater impact on both attributes. All treated cells showed markedly decreased levels of the multidrug resistance enzyme PGP as well as the lipogenic enzymes FASN, ACC1, SCD1, HMGCR, and DGAT. Furthermore, lipid fingerprints were markedly different in treated cells compared with the untreated group. 4Cone increased the percentage of sphingomyelin (SM) while it decreased the percentage of ceramide (Cer); 4Cone in conjunction with TXT had the reverse effect. Triglyceride levels were reduced in 4Cone- and 4Cone/TXT-treated cells, but interestingly, they increased in TXT-treated cells. Additionally, treated cancer cells exhibited changes in glycerophospholipid subclasses. CONCLUSION: 4Cone alone or in combination with TXT alters the lipid profile by reducing a key lipogenic enzyme, resulting in the inhibition of cell proliferation and migration.


Subject(s)
Cell Movement , Cell Proliferation , Docetaxel , Lipidomics , Humans , Docetaxel/pharmacology , Lipidomics/methods , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Cell Survival/drug effects , Drug Synergism , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Antineoplastic Agents/pharmacology , Lipid Metabolism/drug effects , MDA-MB-231 Cells
18.
Biomed Pharmacother ; 177: 117125, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39002444

ABSTRACT

Active targeting to cancer involves exploiting specific interactions between receptors on the surface of cancer cells and targeting moieties conjugated to the surface of vectors such that site-specific delivery is achieved. Prostate specific membrane antigen (PSMA) has proved to be an excellent target for active targeting to prostate cancer. We report the synthesis and use of a PSMA-specific ligand (Glu-NH-CO-NH-Lys) for the site-specific delivery of brusatol- and docetaxel-loaded poly(lactide-co-glycolide) (PLGA) nanoparticles to prostate cancer. The PSMA targeting ligand covalently linked to PLGA-PEG3400 was blended with methoxyPEG-PLGA to prepare brusatol- and docetaxel-loaded nanoparticles with different surface densities of the targeting ligand. Flow cytometry was used to evaluate the impact of different surface densities of the PSMA targeting ligand in LNCaP prostate cancer cells at 15 min and 2 h. Cytotoxicity evaluations of the targeted nanoparticles reveal differences based on PSMA expression in PC-3 and LNCaP cells. In addition, levels of reactive oxygen species (ROS) were measured using the fluorescent indicator, H2DCFDA, by flow cytometry. PSMA-targeted nanoparticles loaded with docetaxel and brusatol showed increased ROS generation in LNCaP cells compared to PC-3 at different time points. Furthermore, the targeted nanoparticles were evaluated in male athymic BALB/c mice implanted with PSMA-producing LNCaP cell tumors. Evaluation of the percent relative tumor volume show that brusatol-containing nanoparticles show great promise in inhibiting tumor growth. Our data also suggest that the dual drug-loaded targeted nanoparticle platform improves the efficacy of docetaxel in male athymic BALB/c mice implanted with PSMA-producing LNCaP cell tumors.


Subject(s)
Antigens, Surface , Docetaxel , Glutamate Carboxypeptidase II , Nanoparticles , Prostatic Neoplasms , Male , Docetaxel/pharmacology , Docetaxel/administration & dosage , Animals , Humans , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Glutamate Carboxypeptidase II/metabolism , Antigens, Surface/metabolism , Cell Line, Tumor , Nanoparticles/chemistry , Reactive Oxygen Species/metabolism , PC-3 Cells , Mice , Xenograft Model Antitumor Assays , Zebrafish , Mice, Nude , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage , Mice, Inbred BALB C , Nanoparticle Drug Delivery System/chemistry
19.
Niger J Clin Pract ; 27(7): 827-836, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-39082907

ABSTRACT

BACKGROUND: Docetaxel (DOC) is the main chemotherapeutic agent for the treatment of advanced metastatic prostate cancer. Docetaxel shows anticancer effects by preventing the depolymerization of microtubules in the cell, therefore preventing cell division. However, the low survival effect of docetaxel has prompted researchers to search for novel therapeutic agents. Fucoidan (FUC) is a sulfated polysaccharide derived from brown algae. It has many bioactivities which makes fucoidan a promising anticancer agent. In this study, the potential anti-tumorigenic and preventive effects of fucoidan with or without docetaxel in prostate cancer were investigated by analyzing different cell death modalities. METHODS: The in-vivo six groups (n = 8) were conducted; preventive (Pt), docetaxel treated after preventive (Pt-D), control, fucoidan (FUC), docetaxel (DOC), and FUC and DOC (FUC+DOC) combination. Apoptotic, necroptotic, and autophagic cell death-related protein expressions were assessed in tumor tissues by using immunohistochemical staining. Oxidative stress-related lipid peroxidation, glutathione peroxidase, and glutathione levels were also determined in tumor tissues. RESULTS: Although apoptotic, necroptotic, and autophagic cell deaths were significantly induced in agent-treated groups compared to the control. Apoptotic cell death was more significantly induced in FUC and FUC+DOC-treated groups. Necroptotic cell death was increased considerably by inducing MLKL protein expression in all treatment groups. In the FUC, Pt, and DOC groups, LC3A/B expressions were significantly increased. DOC, FUC+DOC, and Pt-D treatments caused a significant increase in Beclin-1 expression. Oxidative stress-related MDA, GPX, and GSH levels significantly decreased with FUC treatment. The anti-tumorigenic effects of FUC and DOC were also demonstrated through tumor size reduction. CONCLUSION: According to the findings of this study, FUC inhibited tumor growth temporally and dimensionally, especially in preventive applications. FUC and FUC+DOC combinations in both treatment groups showed anti-tumorigenic effects. The results of this study suggest that fucoidan is a promising anticancer agent against prostate cancer. FUC can be considered as a preventive or treatment agent in prostate cancer therapy with DOC. Further studies are needed to fully elucidate the mechanism of action of fucoidan in metastatic prostate cancer.


Subject(s)
Apoptosis , Docetaxel , Polysaccharides , Prostatic Neoplasms , Male , Polysaccharides/pharmacology , Prostatic Neoplasms/pathology , Prostatic Neoplasms/drug therapy , Docetaxel/pharmacology , Apoptosis/drug effects , Humans , Antineoplastic Agents/pharmacology , Animals , Oxidative Stress/drug effects , Cell Death/drug effects , Autophagy/drug effects , Mice
20.
Eur J Med Chem ; 276: 116670, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39018922

ABSTRACT

Development of resistance to therapy-induced cell death is a major hurdle in the effective treatment of advanced solid tumors. Erastin and RSL3 were originally found to induce synthetic lethality by induction of a novel form of cell death termed ferroptosis. Emerging evidence suggests that ferroptosis inducers enhance chemosensitivity of classic therapeutic agents by triggering ferroptotic cell death. In this study we evaluated the effects of erastin and RSL3 on the resistance of docetaxel, doxorubicin, and cisplatin, and revealed a mechanism whereby these ferroptosis inducers augment docetaxel efficacy in non-small cell lung cancer by regulating redox signaling to promote ferroptosis. Transcriptome analysis revealed that combination treatment modulated not only p53 signaling pathway but also immune responses and several signaling pathways including MAPK, NF-κB and PI3K/Akt. Considering that glutathione peroxidase 4 (GPX4) serves as the main effector to protect cells from ferroptosis, this study identified three novel non-covalent GPX4 inhibitors with the aid of pharmacophore-based virtual screening. The new ferroptosis-inducing compounds synergized with docetaxel to increase the cytotoxicity by promoting ferroptotic cell death in docetaxel-resistant A549/DTX cells. Collectively, the induction of ferroptosis contributed to docetaxel-induced cytotoxic effects and overcame drug resistance in A549/DTX cells. Ferroptosis has a great potential to become a new approach to attenuate resistance to some classic therapeutic drugs in cancer patients.


Subject(s)
Antineoplastic Agents , Carcinoma, Non-Small-Cell Lung , Docetaxel , Drug Resistance, Neoplasm , Ferroptosis , Lung Neoplasms , Ferroptosis/drug effects , Humans , Docetaxel/pharmacology , Docetaxel/chemistry , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , Drug Resistance, Neoplasm/drug effects , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Drug Screening Assays, Antitumor , Molecular Structure , Piperazines/pharmacology , Piperazines/chemistry , Drug Synergism , Dose-Response Relationship, Drug , Structure-Activity Relationship , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , Carbolines
SELECTION OF CITATIONS
SEARCH DETAIL