Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.041
Filter
1.
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
2.
Med Oncol ; 41(9): 219, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39105986

ABSTRACT

Bladder cancer is a common malignancy worldwide, posing a substantial healthcare challenge. Current standard treatment regimens are primarily based on cisplatin, but their success is often limited by cisplatin resistance and associated toxicities. Therefore, there is an urgent need to develop effective and less toxic therapies as alternatives to cisplatin. We screened the activity of FDA-approved anti-cancer drugs on a panel of cisplatin-resistant bladder cancer cell lines. Based on initial responses, cabazitaxel was selected for further evaluation of its inhibitory effects on the phenotypic properties of these cells. Cabazitaxel, primarily used for metastatic castration-resistant prostate cancer, demonstrated remarkable efficacy in inhibiting colony formation, proliferation, and migration of cisplatin-resistant bladder cancer cells. This study highlights the potential of drug repurposing as a cost-effective and efficient strategy to overcome drug resistance in bladder cancer.


Subject(s)
Antineoplastic Agents , Cisplatin , Drug Resistance, Neoplasm , Taxoids , Urinary Bladder Neoplasms , Humans , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Cisplatin/pharmacology , Cisplatin/therapeutic use , Taxoids/pharmacology , Taxoids/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Movement/drug effects
3.
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
4.
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
5.
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
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.
Eur J Pharm Biopharm ; 203: 114435, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39103002

ABSTRACT

The clinical usage of docetaxel (DTX) is severely hindered by the dose-limiting neutropenia and peripheral neurotoxicity of polysorbate 80-solubilized DTX injection, and there are no alternative formulations until now. In this study, we developed a new liposomal formulation of DTX to reduce its toxicities, accompanying with the greatly improved antitumor activity. The DTX was encapsulated into liposomes in the form of hydrophilic glutathione (GSH)-conjugated prodrugs using a click drug loading method, which achieved a high encapsulation efficiency (∼95 %) and loading capacity (∼30 % wt). The resulting liposomal DTX-GSH provided a sustained and efficient DTX release (∼50 % within 48 h) in plasma, resulting in a greatly improved antitumor activities as compared with that of polysorbate 80-solubilized DTX injection in the subcutaneous and orthotopic 4T1 breast tumor bearing mice. Even large tumors > 500 mm3 could be effectively inhibited and shrunk after the administration of liposomal DTX-GSH. More importantly, the liposomal DTX-GSH significantly decreased the neutropenia and peripheral neurotoxicity as compared with that of polysorbate 80-solubilized DTX injection at the equivalent dose. These data suggested that the liposomal DTX-GSH might become a superior alternative formulation to the commercial DTX injection.


Subject(s)
Antineoplastic Agents , Docetaxel , Glutathione , Liposomes , Mice, Inbred BALB C , Docetaxel/administration & dosage , Docetaxel/pharmacokinetics , Docetaxel/pharmacology , Docetaxel/chemistry , Animals , Mice , Glutathione/chemistry , Female , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Cell Line, Tumor , Prodrugs/administration & dosage , Prodrugs/chemistry , Prodrugs/pharmacology , Taxoids/administration & dosage , Taxoids/pharmacology , Taxoids/pharmacokinetics , Taxoids/chemistry , Polysorbates/chemistry , Neutropenia/chemically induced , Neutropenia/drug therapy
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.
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
10.
Toxicol In Vitro ; 100: 105894, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38996827

ABSTRACT

Here, we revealed the reversibility of cabazitaxel (CBZ)-induced apoptosis in PC-3 castration resistant metastatic prostate cancer cells (mCRPC) through the hallmarks of apoptosis. The recovery of PC-3 cells from apoptosis upon removal of CBZ at different recovery periods was evaluated by Annexin V, DNA damage, oxidative damage, mitochondrial membrane depolarization, and caspase activation. Our results showed that the administration of CBZ caused apoptosis for 72 h in PC-3 cells. However, recovered cells exhibited decreased nuclear damage, plasma membrane disruption, ROS level, release cytochrome c level and caspase-3 activation with upregulation of Bcl-2 expression upon removal of especially 1 nM CBZ for 24 h recovery period in PC-3 cells. Our study indicates that CBZ treated PC-3 cells can recover after apoptotic cell death. However, advanced molecular analysis should elucidate the relationship between the molecular mechanisms of recovery and taxane response or resistance in PC-3 mCRPC cells.


Subject(s)
Antineoplastic Agents , Apoptosis , Prostatic Neoplasms, Castration-Resistant , Reactive Oxygen Species , Taxoids , Male , Humans , Apoptosis/drug effects , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/pathology , Taxoids/pharmacology , Antineoplastic Agents/pharmacology , Reactive Oxygen Species/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Caspase 3/metabolism , PC-3 Cells , Cytochromes c/metabolism , Membrane Potential, Mitochondrial/drug effects , DNA Damage/drug effects , Cell Line, Tumor
11.
Int J Pharm ; 661: 124439, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38972520

ABSTRACT

Liposomes functionalized with monoclonal antibodies offer targeted therapy for cancer, boasting advantages like sustained drug release, enhanced stability, passive accumulation in tumors, and interaction with overexpressed receptors on cancer cells. This study aimed to develop and characterize anti-EGFR immunoliposomes loaded with cabazitaxel and assess their properties against prostate cancer in vitro and in vivo. Using a Box-Behnken design, a formulation with soy phosphatidylcholine, 10% cholesterol, and a 1:20 drug-lipid ratio yielded nanometric particle size, low polydispersity and high drug encapsulation. Immunoliposomes were conjugated with cetuximab through DSPE-PEG-Maleimide lipid anchor. Characterization confirmed intact antibody structure and interaction with EGFR receptor following conjugation. Cabazitaxel was dispersed within the liposomes in the amorphous state, confirmed by solid-state analyses. In vitro release studies showed slower cabazitaxel release from immunoliposomes. Immunoliposomes had enhanced cabazitaxel cytotoxicity in EGFR-overexpressing DU145 cells without affecting non-tumor L929 cells. Cetuximab played an important role to improve cellular uptake in a time-dependent fashion in EGFR-overexpressing prostate cancer cells. In vivo, immunoliposomes led to significant tumor regression, improved survival, and reduced weight loss in xenograft mice. While cabazitaxel induced leukopenia, consistent with clinical findings, histological analysis revealed no evident toxicity. In conclusion, the immunoliposomes displayed suitable physicochemical properties for cabazitaxel delivery, exhibited cytotoxicity against EGFR-expressing prostate cancer cells, with high cell uptake, and induced significant tumor regression in vivo, with manageable systemic toxicity.


Subject(s)
Cetuximab , Drug Liberation , ErbB Receptors , Liposomes , Prostatic Neoplasms , Taxoids , Xenograft Model Antitumor Assays , Male , Animals , ErbB Receptors/immunology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Humans , Cell Line, Tumor , Taxoids/administration & dosage , Taxoids/pharmacokinetics , Taxoids/pharmacology , Taxoids/chemistry , Cetuximab/administration & dosage , Mice , Mice, Nude , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/chemistry , Polyethylene Glycols/chemistry , Polyethylene Glycols/administration & dosage , Particle Size , Drug Delivery Systems
12.
Toxicol Appl Pharmacol ; 489: 116993, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38870637

ABSTRACT

We tested the effect of substituents at the (1) C3´, C3´N, (2) C10, and (3) C2-meta-benzoate positions of taxane derivatives on their activity against sensitive versus counterpart paclitaxel-resistant breast (MCF-7) and ovarian (SK-OV-3) cancer cells. We found that (1) non-aromatic groups at both C3´ and C3´N positions, when compared with phenyl groups at the same positions of a taxane derivative, significantly reduced the resistance of ABCB1 expressing MCF-7/PacR and SK-OV-3/PacR cancer cells. This is, at least in the case of the SB-T-1216 series, accompanied by an ineffective decrease of intracellular levels in MCF-7/PacR cells. The low binding affinity of SB-T-1216 in the ABCB1 binding cavity can elucidate these effects. (2) Cyclopropanecarbonyl group at the C10 position, when compared with the H atom, seems to increase the potency and capability of the derivative in overcoming paclitaxel resistance in both models. (3) Derivatives with fluorine and methyl substituents at the C2-meta-benzoate position were variously potent against sensitive and resistant cancer cells. All C2 derivatives were less capable of overcoming acquired resistance to paclitaxel in vitro than non-substituted analogs. Notably, fluorine derivatives SB-T-121205 and 121,206 were more potent against sensitive and resistant SK-OV-3 cells, and derivatives SB-T-121405 and 121,406 were more potent against sensitive and resistant MCF-7 cells. (4) The various structure-activity relationships of SB-T derivatives observed in two cell line models known to express ABCB1 favor their complex interaction not based solely on ABCB1.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B , Drug Resistance, Neoplasm , Humans , Drug Resistance, Neoplasm/drug effects , MCF-7 Cells , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily B/genetics , Structure-Activity Relationship , Taxoids/pharmacology , Taxoids/chemistry , Cell Line, Tumor , Paclitaxel/pharmacology , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Benzoates/pharmacology , Benzoates/chemistry , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology
13.
ESMO Open ; 9(6): 103486, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38914452

ABSTRACT

BACKGROUND: Paclitaxel resistance limits durability of response in patients with initial clinical benefit. Overexpression of spleen tyrosine kinase (SYK) has been proposed as a possible resistance mechanism. This phase I trial evaluated the safety and preliminary activity of the SYK inhibitor TAK-659 combined with paclitaxel in patients with advanced taxane-refractory solid tumors. PATIENTS AND METHODS: Patients with advanced solid tumors and prior progression on taxane-based therapy received intravenous infusion of paclitaxel on days 1, 8, and 15 plus oral TAK-659 daily in 28-day cycles. The dose-escalation phase included six cohorts treated at different dose levels; the dose-expansion phase included patients with ovarian cancer treated at the highest dose level. Toxicity was graded using the National Cancer Institute Common Terminology Criteria for Adverse Events version 5.0. Efficacy was evaluated using Response Evaluation Criteria in Solid Tumors version 1.1. RESULTS: Our study included 49 patients. Maximum tolerated dose was not reached, but higher rates of adverse events were observed at higher dose levels. There were no treatment-related deaths. The most common treatment-related adverse events of any grade were increased aspartate aminotransferase (n = 31; 63%), increased alanine aminotransferase (n = 26; 53%), decreased neutrophil count (n = 26; 53%), and decreased white blood cell count (n = 26; 53%). Most adverse events were either grade 1 or 2. In the 44 patients with evaluable disease, 12 (27%) had stable disease as the best overall response, including three patients with prolonged stable disease, and 4 patients (9%) achieved a partial response. CONCLUSIONS: The combination of paclitaxel and TAK-659 showed preliminary activity possibly overcoming resistance to taxane-based therapy as well as a tolerable safety profile in patients with advanced solid tumors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Neoplasms , Paclitaxel , Humans , Paclitaxel/therapeutic use , Paclitaxel/pharmacology , Paclitaxel/administration & dosage , Female , Middle Aged , Aged , Neoplasms/drug therapy , Male , Adult , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Resistance, Neoplasm , Taxoids/therapeutic use , Taxoids/pharmacology , Maximum Tolerated Dose , Syk Kinase/metabolism
14.
Int J Mol Sci ; 25(12)2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38928449

ABSTRACT

Triple-negative breast cancer (TNBC) patients are treated with traditional chemotherapy, such as the taxane class of drugs. One such drug, paclitaxel (PTX), can be effective in treating TNBC; however, many tumors will develop drug resistance, which can lead to recurrence. In order to improve patient outcomes and survival, there lies a critical need to understand the mechanism behind drug resistance. Our lab made the novel observation that decreased expression of the Adenomatous Polyposis Coli (APC) tumor suppressor using shRNA caused PTX resistance in the human TNBC cell line MDA-MB-157. In cells lacking APC, induction of apoptosis by PTX was decreased, which was measured through cleaved caspase 3 and annexin/PI staining. The current study demonstrates that CRISPR-mediated APC knockout in two other TNBC lines, MDA-MB-231 and SUM159, leads to PTX resistance. In addition, the cellular consequences and molecular mechanisms behind APC-mediated PTX response have been investigated through analysis of the BCL-2 family of proteins. We found a significant increase in the tumor-initiating cell population and increased expression of the pro-survival family member Bcl-2, which is widely known for its oncogenic behavior. ABT-199 (Venetoclax), is a BH3 mimetic that specifically targets Bcl-2. ABT-199 has been used as a single or combination therapy in multiple hematologic malignancies and has shown promise in multiple subtypes of breast cancer. To address the hypothesis that APC-induced Bcl-2 increase is responsible for PTX resistance, we combined treatment of PTX and ABT-199. This combination treatment of CRISPR-mediated APC knockout MDA-MB-231 cells resulted in alterations in apoptosis, suggesting that Bcl-2 inhibition restores PTX sensitivity in APC knockout breast cancer cells. Our studies are the first to show that Bcl-2 functional inhibition restores PTX sensitivity in APC mutant breast cancer cells. These studies are critical to advance better treatment regimens in patients with TNBC.


Subject(s)
Apoptosis , Drug Resistance, Neoplasm , Proto-Oncogene Proteins c-bcl-2 , Triple Negative Breast Neoplasms , Humans , Drug Resistance, Neoplasm/genetics , Cell Line, Tumor , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Apoptosis/drug effects , Female , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Sulfonamides/pharmacology , Paclitaxel/pharmacology , Up-Regulation/drug effects , Taxoids/pharmacology , Bridged-Ring Compounds , Bridged Bicyclo Compounds, Heterocyclic
15.
Anticancer Res ; 44(7): 2815-2821, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38925843

ABSTRACT

BACKGROUND/AIM: The cytoprotective heat shock protein 27 (HSP27) acts as a protein chaperone, antioxidant, and apoptosis regulator and is involved in cytoskeletal remodeling in prostate cancer. This study was designed to assess the effect of prostate cancer therapeutics on HSP27 to identify drugs that may benefit from an HSP27 inhibitor combination therapy. MATERIALS AND METHODS: Cell counting was utilized to assess drug treatment efficiency. Changes in protein levels after drug treatment were assessed using western blot analysis. RESULTS: Abiraterone, cabazitaxel, docetaxel and enzalutamide significantly reduced cell proliferation in LNCaP and PC3 cells. Treatment with abiraterone and enzalutamide led to a significant reduction in HSP27 protein levels. In contrast, treatment with cabazitaxel and docetaxel did not change the HSP27 protein levels. CONCLUSION: Treatment with abiraterone and enzalutamide reduces HSP27 protein in an AR-independent manner and thus suppresses HSP27-correlated resistance mechanisms. However, docetaxel and cabazitaxel do not alter HSP27 protein levels, so that taxanes' efficacy may be enhanced by combining them with HSP27-inhibiting drugs.


Subject(s)
Androstenes , Antineoplastic Agents , Benzamides , Cell Proliferation , Docetaxel , Drug Resistance, Neoplasm , HSP27 Heat-Shock Proteins , Nitriles , Phenylthiohydantoin , Prostatic Neoplasms , Taxoids , Humans , Male , Taxoids/pharmacology , Taxoids/therapeutic use , Docetaxel/pharmacology , Phenylthiohydantoin/pharmacology , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/therapeutic use , HSP27 Heat-Shock Proteins/metabolism , Drug Resistance, Neoplasm/drug effects , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Androstenes/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Molecular Chaperones/metabolism , Heat-Shock Proteins/metabolism
16.
Bioorg Chem ; 150: 107569, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38905886

ABSTRACT

Erianin, a natural compound derived from Dendrobium, has shown significant anticancer properties against a wide range of cancer cells. Despite the identification of multiple mechanisms of action for erianin, none of these mechanisms fully account for its broad-spectrum effect. In this study, we aimed to identify the cellular target and underlying mechanism responsible for the broad-spectrum antitumor effects of erianin. We found that erianin effectively inhibited tubulin polymerization in cancer cells and purified tubulin. Through competition binding assays and X-ray crystallography, it was revealed that erianin bound to the colchicine site of ß-tubulin. Importantly, the X-ray crystal structure of the tubulin-erianin complex was solved, providing clear insight into the orientation and position of erianin in the colchicine-binding site. Erianin showed activity against paclitaxel-resistant cells, evidenced by G2/M cell cycle arrest, apoptosis-related PARP and Caspase-3 cleavage, and in vivo xenograft studies. The study concluded that erianin bound reversibly to the colchicine site of ß-tubulin, inhibited tubulin polymerization, and displayed anticancer activity against paclitaxel-resistant cells, offering valuable insights for further exploration as potential anticancer agents.


Subject(s)
Antineoplastic Agents , Cell Proliferation , Colchicine , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Tubulin , Humans , Tubulin/metabolism , Tubulin/chemistry , Colchicine/pharmacology , Colchicine/chemistry , Colchicine/metabolism , Binding Sites , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Animals , Drug Resistance, Neoplasm/drug effects , Cell Proliferation/drug effects , Structure-Activity Relationship , Molecular Structure , Dose-Response Relationship, Drug , Mice , Apoptosis/drug effects , Taxoids/pharmacology , Taxoids/chemistry , Tubulin Modulators/pharmacology , Tubulin Modulators/chemistry , Crystallography, X-Ray , Bridged-Ring Compounds/chemistry , Bridged-Ring Compounds/pharmacology , Mice, Nude , Cell Line, Tumor , Biological Products/chemistry , Biological Products/pharmacology , Bibenzyls/chemistry , Bibenzyls/pharmacology , Phenol
17.
Biomed Pharmacother ; 177: 116822, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38906029

ABSTRACT

Aberration of Notch signaling is one of the key events involved in the development and progression of head and neck squamous cell carcinoma (HNSCC). The Notch pathway controls the tissue-specific differentiation of normal squamous epithelial cells and is frequently altered in squamous carcinomas, thus affecting their proliferation, growth, survival, and chemosensitivity or resistance against anti-cancer agents. In this study, we show that the use of novel, small-molecule inhibitors of Notch signaling, such as FLI-06, can have a beneficial effect on increasing the chemosensitivity of HNSCC to taxane-based chemotherapy. Inhibition of Notch signaling by FLI-06 alone virtually blocks the proliferation and growth of HNSCC cells in both 2D and 3D cultures and the zebrafish model, which is accompanied by down-regulation of key Notch target genes and proteins. Mechanistically, FLI-06 treatment causes cell cycle arrest in the G1-phase and induction of apoptosis in HNSCC, which is accompanied by increased c-JunS63 phosphorylation. Combining FLI-06 with Docetaxel shows a synergistic effect and partially blocks the cell growth of aggressive HNSCC cells via enhanced apoptosis and modification of c-JunS243 phosphorylation via GSK-3ß inhibition. In conclusion, inhibition of Notch signaling in HNSCC cells that retain active Notch signaling significantly supports taxane-based anticancer activities via modulation of both the GSK-3ß and the c-Jun.


Subject(s)
Apoptosis , Cell Proliferation , Head and Neck Neoplasms , Receptors, Notch , Squamous Cell Carcinoma of Head and Neck , Taxoids , Zebrafish , Humans , Animals , Receptors, Notch/metabolism , Receptors, Notch/antagonists & inhibitors , Cell Line, Tumor , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/metabolism , Taxoids/pharmacology , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/metabolism , Cell Proliferation/drug effects , Apoptosis/drug effects , Docetaxel/pharmacology , Signal Transduction/drug effects , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/metabolism , Drug Resistance, Neoplasm/drug effects , Drug Synergism
18.
ESMO Open ; 9(6): 103446, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38838502

ABSTRACT

BACKGROUND: This phase II nonrandomized study evaluated the efficacy and safety of AZD4635 in combination with durvalumab (Arm A) or durvalumab plus cabazitaxel (Arm B) in patients with metastatic castration-resistant prostate cancer (mCRPC) previously treated with docetaxel and ≥1 novel hormonal agent. PATIENTS AND METHODS: The primary endpoint was radiographic progression-free survival (rPFS) per RECIST v1.1 (soft tissue) or the Prostate Cancer Clinical Trials Working Group 3 (bone). Secondary endpoints included safety, tolerability, overall survival, confirmed prostate-specific antigen (PSA50) response, pharmacokinetics, and objective response rate. Enrollment in Arm A was stopped following a sponsor decision unrelated to safety. The study was stopped based on the planned futility analysis due to low PSA50 response in Arm B. RESULTS: In the final analysis (1 November 2021), 30 patients were treated (Arm A, n = 2; Arm B, n = 28). The median rPFS in Arm B was 5.8 months (95% confidence interval 4.2-not calculable). Median rPFS was 5.8 months versus 4.2 months for patients with high versus low blood-based adenosine signature. The most common treatment-related adverse events in Arm B were nausea (50.0%), diarrhea (46.4%), anemia and neutropenia (both 35.7%), asthenia (32.1%), and vomiting (28.6%). Overall, AZD4635 in combination with durvalumab or AZD4635 in combination with cabazitaxel and durvalumab showed limited efficacy in patients with mCRPC. CONCLUSIONS: Although the safety profile of both combinations was consistent with known safety data of the individual agents, the results of this trial do not support further development of the combinations.


Subject(s)
Antibodies, Monoclonal , Antineoplastic Combined Chemotherapy Protocols , Prostatic Neoplasms, Castration-Resistant , Humans , Male , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/pathology , Aged , Middle Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/administration & dosage , Taxoids/therapeutic use , Taxoids/pharmacology , Taxoids/administration & dosage , Aged, 80 and over , Progression-Free Survival , Neoplasm Metastasis
19.
Adv Sci (Weinh) ; 11(33): e2404800, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38934894

ABSTRACT

The treatment of colorectal cancer is always a major challenge in the field of cancer research. The number of estimated new cases of colorectal cancer worldwide in 2020 is 1 148 515, and the estimated number of deaths is 576 858, revealing that mortality accounted for approximately half of the disease incidence. The development of new drugs and strategies for colorectal cancer treatment is urgently needed. Thermosensitive injectable hydrogel PDLLA-PEG-PDLLA (PLEL) loaded with cabazitaxel (CTX) is used to explore its anti-tumor effect on mice with orthotopic colorectal cancer. CTX/PLEL is characterized by a solution state at room temperature and a hydrogel state at physiologic temperature. The excipients MPEG-PCL and PDLLA-PEG-PDLLA have good biocompatibility and biodegradability. The simple material synthesis and preparation process renders this system cost-effective and more conducive to clinical transformation. An orthotopic colorectal cancer model is established by transplantation subcutaneous tumors onto the cecum of mice. According to the results of experiments in vivo, CTX/PLEL significantly inhibits orthotopic colorectal cancer and liver metastasis in mice. The results indicate that CTX/PLEL nanoparticle preparations have high security and excellent anti-tumor effects, and have great application potential in colorectal cancer therapy.


Subject(s)
Colorectal Neoplasms , Disease Models, Animal , Hydrogels , Liver Neoplasms , Animals , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Mice , Hydrogels/chemistry , Liver Neoplasms/drug therapy , Liver Neoplasms/secondary , Taxoids/pharmacology , Taxoids/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Mice, Inbred BALB C , Cell Line, Tumor
20.
J Biomater Sci Polym Ed ; 35(14): 2220-2249, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38944817

ABSTRACT

Innovation chemotherapeutic nano drug delivery systems (NDDSs) with various pharmacological achievement have become one of the hopeful therapeutic strategies in cancer therapy. This study focused on low pH, and high levels of glutathione (GSH) as two prominent characteristics of the tumor microenvironment (TME) to design a novel TME-targeted pH/redox dual-responsive P (AMA-co-DMAEMA)-b-PCL-SS-PCL-b-P (AMA-co-DMAEMA) nanoparticles (NPs) for deep tumor penetration and targeted anti-tumor therapy. The positively charged NPs exhibit strong electrostatic interactions with negatively charged cell membranes, significantly enhancing cellular uptake. Moreover, these NPs possess the unique size-shrinkable property, transitioning from 98.24 ± 27.78 to 45.56 ± 20.62 nm within the TME. This remarkable size change fosters an impressive uptake of approximately 100% by MDA-MB-231 cells within just 30 min, thereby greatly improving drug delivery efficiency. This size switchability enables passive targeting through the enhanced permeability and retention (EPR) effect, facilitating deep penetration into tumors. The NPs also demonstrate improved pH/redox-triggered drug release (∼70% at 24 h) within the TME and exhibit no toxicity in cell viability test. The cell cycle results of treated cells with docetaxel (DTX)-loaded NPs revealed G2/M (84.6 ± 1.16%) arrest. The DTX-loaded NPs showed more apoptosis (62.6 ± 3.7%) than the free DTX (51.8 ± 3.2%) in treated cells. The western blot and RT-PCR assays revealed that apoptotic genes and proteins expression of treated cells were significantly upregulated with the DTX-loaded NPs vs. the free DTX (Pvalue<.001). In conclusion, these findings suggest that this novel-engineered NPs holds promise as a TME-targeted NDDS.


Subject(s)
Docetaxel , Drug Liberation , Nanoparticles , Oxidation-Reduction , Tumor Microenvironment , Hydrogen-Ion Concentration , Tumor Microenvironment/drug effects , Humans , Docetaxel/chemistry , Docetaxel/pharmacology , Cell Line, Tumor , Nanoparticles/chemistry , Drug Carriers/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/administration & dosage , Taxoids/chemistry , Taxoids/pharmacology , Taxoids/administration & dosage , Taxoids/pharmacokinetics , Particle Size , Apoptosis/drug effects , Cell Survival/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL