Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 12.758
Filter
1.
Hinyokika Kiyo ; 70(6): 141-147, 2024 Jun.
Article in Japanese | MEDLINE | ID: mdl-38967025

ABSTRACT

The administration of cabazitaxel for patients with castration-resistant prostate cancer (CRPC) requires prior docetaxel therapy. Sequential chemotherapy may have to be discontinued due to docetaxelassociated side effects. This study investigated the relationship between treatment outcome of docetaxel and cabazitaxel and their associated side effects. We retrospectively analyzed 69 patients with CRPC who had been administered docetaxel withand without subsequent cabazitaxel at Toyonaka Municipal Hospital from October 2014 to June 2022. Twenty-eight patients (41%) discontinued docetaxel because of side effects, and the median number of docetaxel cycles at discontinuation was 2 (range : 1-11). Fourteen of these patients received no treatment following docetaxel. A comparison of the 28 patients who had discontinued docetaxel due to side effects with 41 patients who had not revealed a significant difference in the total numbers of chemotherapy cycles (2.5 vs 9 ; P<0.001) and time to treatment failure (56 days vs 301 days ; P= 0.001), with a trend toward shorter overall survival from the start of docetaxel treatment (259 days vs 512 days ; P=0.06). Multivariate analysis identified discontinuation of docetaxel due to side effects (OR=0.07 ; P<0.001) and lower hemoglobin (OR=0.01 ; P=0.001) as significant factors inhibiting the introduction of cabazitaxel. Reducing the side effects of docetaxel, including early drug switching, may allow more CRPC patients to be reached with cabazitaxel. Consequently, the resulting taxane-based chemotherapy may contribute to an additional survival advantage.


Subject(s)
Docetaxel , Prostatic Neoplasms, Castration-Resistant , Taxoids , Humans , Male , Taxoids/adverse effects , Taxoids/administration & dosage , Prostatic Neoplasms, Castration-Resistant/drug therapy , Docetaxel/administration & dosage , Docetaxel/adverse effects , Aged , Middle Aged , Retrospective Studies , Aged, 80 and over , Antineoplastic Agents/adverse effects , Antineoplastic Agents/administration & dosage , Treatment Outcome
2.
BMC Urol ; 24(1): 135, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38951868

ABSTRACT

The ARASENS trial recruited 1306 men with metastatic hormone sensitive prostate cancer. It investigated the effect of androgen deprivation therapy (ADT) and systemic therapy docetaxel in combination with a third novel drug - daralutamide, compared with placebo on overall survival. Triple therapy with ADT, docetaxel and darolutamide resulted in improved overall survival rates as compared with ADT, docetaxel and placebo (HR 0.68; 95% CI, 0.57-0.80; p < 0.001). The side effect profile for both treatments was similar. This randomised, double blinded, placebo controlled study, was assessed to have a low risk of bias using the Cochrane Risk of Bias 2 tool.


Subject(s)
Prostatic Neoplasms , Male , Humans , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Prostatic Neoplasms/mortality , Benzamides/therapeutic use , Randomized Controlled Trials as Topic , Survival Rate , Androgen Antagonists/therapeutic use , Docetaxel/therapeutic use , Pyrazoles
3.
Support Care Cancer ; 32(8): 513, 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39001936

ABSTRACT

PURPOSE: Anthracycline-cyclophosphamide followed by docetaxel-containing chemotherapy is effective for perioperative breast cancer treatment. However, these treatments frequently induce oral mucositis (OM), with an incidence ranging from 20 to 50%. The association of OM development between different chemotherapeutic treatments remains unclear. Consequently, this study aimed to compare OM development during docetaxel-containing chemotherapy between patients with and without OM experience during previous anthracycline-cyclophosphamide treatments to assess the association between OM development and treatment regimens. METHODS: Seventy-two patients with breast cancer receiving anthracycline-cyclophosphamide followed by docetaxel-containing chemotherapy as a perioperative treatment were categorized into the control (no prior OM experience with anthracycline-cyclophosphamide) and OM-experience (OM development during previous treatment) groups and retrospectively evaluated. The primary endpoint was the incidence of all-grade OM in the first docetaxel-containing chemotherapy cycle. Additionally, the incidences of OM and dysgeusia during all treatment cycles and factors associated with the incidence of OM were evaluated. RESULTS: The incidence of all-grade OM in the first cycle was significantly higher in the OM-experience group (54.2%) than in the control group (10.4%; P < 0.0001). Furthermore, its incidence in all treatment cycles was higher in the OM-experience group (66.7%) than in the control group (12.5%, P < 0.0001). However, the incidence of dysgeusia did not differ between the groups. Multivariate logistic regression analysis revealed OM experience during previous anthracycline-cyclophosphamide treatment and concomitant pertuzumab use as independent risk factors for OM development in subsequent docetaxel-containing chemotherapy. CONCLUSION: Our study suggests that patients experiencing OM with anthracycline-cyclophosphamide during perioperative breast cancer treatment exhibit symptoms following subsequent docetaxel-containing chemotherapy.


Subject(s)
Anthracyclines , Antineoplastic Combined Chemotherapy Protocols , Breast Neoplasms , Cyclophosphamide , Docetaxel , Stomatitis , Humans , Breast Neoplasms/drug therapy , Breast Neoplasms/surgery , Female , Docetaxel/administration & dosage , Docetaxel/adverse effects , Cyclophosphamide/administration & dosage , Cyclophosphamide/adverse effects , Middle Aged , Stomatitis/chemically induced , Stomatitis/epidemiology , Retrospective Studies , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Anthracyclines/adverse effects , Anthracyclines/administration & dosage , Adult , Aged , Incidence , Taxoids/adverse effects , Taxoids/administration & dosage , Risk Factors
4.
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
5.
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
6.
Sci Rep ; 14(1): 16164, 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39003369

ABSTRACT

The present study investigated the relationship between MSH3 and MSH6 genes in lung cancer patients. Genotyping of lung cancer patients and healthy controls was performed. Odds ratio values were calculated and survival analysis performed. Patients with mutant genotype (TT) for MSH6 polymorphism have 1.5-fold risk for the development of lung cancer (p = 0.03). For non-smokers, the mutant-type genotype had a threefold increased risk of lung cancer (p = 0.01). Patients administered with docetaxel and carbo/cisplatin and carrying GT genotype for MSH6 polymorphism, patients reported a decrease in median survival time (4.9 vs 9.13 months). MSH3 and MSH6 polymorphisms are involved in modulating the risk towards lung cancer. MSH6 polymorphism is associated with high mortality rate for patients undergoing cisplatin and docetaxel chemotherapy.


Subject(s)
Cisplatin , DNA-Binding Proteins , Genetic Predisposition to Disease , Lung Neoplasms , MutS Homolog 3 Protein , Humans , Lung Neoplasms/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/mortality , Male , Middle Aged , Female , Cisplatin/therapeutic use , MutS Homolog 3 Protein/genetics , DNA-Binding Proteins/genetics , Polymorphism, Single Nucleotide , Docetaxel/therapeutic use , India/epidemiology , Aged , Case-Control Studies , Genotype , Adult , Carboplatin/therapeutic use
7.
J Cancer Res Ther ; 20(3): 913-917, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-39023597

ABSTRACT

INTRODUCTION: Adjuvant chemoradiotherapy (CRT) is the optimal management strategy in resectable gastric cancer. There is a debate about the efficacy of more aggressive CRT plus chemotherapy regimens in adjuvant setting. This study aimed to compare the efficacy of adjuvant CRT plus docetaxel-cisplatin-fluorouracil (DCF) versus CRT plus fluorouracil-folinic acid (FUFA) in stage III gastric cancer. METHODS: Patients with a diagnosis of stage III gastric cancer treated with adjuvant therapy after curative resection were analyzed. Patients' disease characteristics and impacts of the regimens on median disease-free survival (DFS) and median overall survival (OS) were analyzed retrospectively. RESULTS: One hundred sixty-one patients (102 in FUFA arm and 59 in DCF arm) with a median age of 56.0 (29-79) were evaluated. In the DCF arm, there were more renal toxicities (31.6% vs 6.4% P < 0.001), emergency department admissions (64.9% vs 23.7%, P < 0.001), and dose reductions/treatment modifications in the DCF arm (51.6% vs 37.2, P < 0.001). The median follow-up was 23 months (1-124) in the FUFA arm and 26.0 months (1-77) in the DCF arm. The median DFS was 25.0 months (%95 CI, 12.7-37.2) in the DCF arm and 17.0 months (%95 CI, 2.6-31.3) in the FUFA arm, P = 0.66. The median OS was 28.0 months (%95 CI, 17.0-38.9) in the DCF arm and 25.0 months (%95 CI, 11.9-36.0) in the FUFA arm, P = 0.70. CONCLUSION: In conclusion, when compared with FUFA regimen, more aggressive therapy with DCF was more toxic and did not improve OS in adjuvant setting of stage III gastric cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Chemoradiotherapy, Adjuvant , Cisplatin , Docetaxel , Fluorouracil , Leucovorin , Neoplasm Staging , Stomach Neoplasms , Humans , Stomach Neoplasms/therapy , Stomach Neoplasms/pathology , Stomach Neoplasms/mortality , Male , Middle Aged , Female , Fluorouracil/administration & dosage , Fluorouracil/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Docetaxel/administration & dosage , Cisplatin/administration & dosage , Cisplatin/therapeutic use , Aged , Adult , Retrospective Studies , Chemoradiotherapy, Adjuvant/methods , Leucovorin/administration & dosage , Leucovorin/therapeutic use , Treatment Outcome
8.
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
9.
Zhonghua Yi Xue Za Zhi ; 104(26): 2401-2408, 2024 Jul 09.
Article in Chinese | MEDLINE | ID: mdl-38978363

ABSTRACT

Objective: To compare the efficacy and safety of carrelizumab combined with the modified TPF regimen (docetaxel, cisplatinand capecitabine) and TPF regimen alone in larynx preservation strategy for locally advanced resectable hypopharyngeal squamous cell carcinoma. Methods: A cohort study was conducted. Patients with locally advanced resectable hypopharyngeal carcinoma (cT3-4aN0-3bM0) who were treated at the Eye & ENT Hospital of Fudan University from January 2017 to April 2023 were enrolled in the study. One group was treated with a modified TPF regimen (TPF group) for 2-3 cycles (retrospective data), and the other group was a prospective phase Ⅱ trial with a modified TPF regimen combined with carrelizumab (TPFC group) for three cycles. The patients with complete or partial remission of the primary focus were treated with sequential radical radiotherapy and/or drug therapy. The patients in the TPFC group were treated with carrelizumab at the end of radiotherapy with a maximum of up to 18 doses. The patients with stable or progressive disease were given radical surgery, and those who refused the surgery were given radical chemoradiotherapy. Objective response rate (ORR), overall survival rate, progression-free survival (PFS) rate, larynx preservation rate (LPR), and adverse reactions were compared between the two groups. Results: There were 51 male patients in the TPFC group, with an median age of 57 (35, 69) years. Meanwhile, 44 patients were in the TPF group, among which 43 were male and one was female, with an median age of 62 (46, 70) years. The ORR of the TPFC group was higher than that of the TPF group [82.4% (42/51) vs 63.6% (28/44), P=0.039]. During a median follow-up of 24.4 (18.5, 31.4) months, the TPFC group showed a higher 2-year survival rate (84.8% vs 64.6%, P=0.013) and 2-year LPR (66.6% vs 48.6%, P=0.045) than those in the TPF group. In patients with poor effect of induction therapy for hypopharyngeal carcinoma, surgical combination therapy significantly prolonged the 2-year PFS rate (77.9% vs 18.2%, P<0.001) and 2-year survival rate (76.9% vs 45.5%, P=0.005)than those of non-surgical combination therapy. The incidences of nausea and/or vomiting, reactive cutaneous capillary endothelial proliferation, thyroid dysfunction, and rash were increased in the TPFC group (all P<0.05). There was no treatment-related death. Conclusion: Carrelizumab combined with a modified TPF regimen has good efficacy and safety and can improve the LPR of locally advanced hypopharyngeal carcinoma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Hypopharyngeal Neoplasms , Humans , Hypopharyngeal Neoplasms/drug therapy , Hypopharyngeal Neoplasms/therapy , Hypopharyngeal Neoplasms/pathology , Male , Female , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Middle Aged , Cisplatin/administration & dosage , Prospective Studies , Induction Chemotherapy , Cohort Studies , Retrospective Studies , Antibodies, Monoclonal, Humanized/therapeutic use , Docetaxel/therapeutic use , Docetaxel/administration & dosage , Treatment Outcome , Adult
10.
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
11.
Anticancer Res ; 44(7): 3155-3161, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38925833

ABSTRACT

BACKGROUND/AIM: The therapeutic landscape for metastatic castration-resistant prostate cancer (mCRPC) has changed dramatically with the introduction of several novel agents. However, limited data are available to determine whether the introduction of novel agents affected patient characteristics, treatment patterns, and outcomes compared with the period when these agents were not available. The objective of this study was to evaluate the impact of the introduction of novel mCRPC agents on patient characteristics, treatment patterns, and outcomes. PATIENTS AND METHODS: Two cohorts of Japanese patients diagnosed with mCRPC between 2009 and 2014 (Epoch 1) and 2015 and 2019 (Epoch 2) were retrospectively analyzed. RESULTS: A total of 125 treatment-naïve mCRPC patients, consisting of 42 patients in Epoch 1 and 83 patients in Epoch 2, were evaluated. We obtained the following results: (i) a dramatic shift in the first-line treatment from docetaxel to androgen receptor axis-targeted agents (ARATs), (ii) an age range expansion for first-line treatment, and (iii) an overall survival (OS) advantage in Eopch 2 compared to Epoch 1. Multivariate analysis in the overall population showed that Epoch 2 and low prostate-specific antigen (PSA) levels at the start of first-line treatment were independent prognostic factors for OS. CONCLUSION: In real-world mCRPC practice, the introduction of novel agents has improved the prognosis of mCRPC while allowing more patients to receive mCRPC treatment across a broad age range. In addition, low PSA levels at the start of first-line treatment were associated with improved OS, indicating the importance of starting mCRPC treatment while PSA levels are low.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Humans , Male , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/pathology , Prostatic Neoplasms, Castration-Resistant/mortality , Aged , Aged, 80 and over , Retrospective Studies , Middle Aged , Treatment Outcome , Docetaxel/therapeutic use , Prostate-Specific Antigen/blood , Neoplasm Metastasis , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Agents/therapeutic use
12.
Radiat Oncol ; 19(1): 76, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38890652

ABSTRACT

OBJECTIVE: This retrospective study aimed to investigate the factors influencing the occurrence of neutropenia in patients with endometrial cancer (EC) following adjuvant chemoradiotherapy (CRT). METHODS: Retrospective analysis of EC patients who underwent adjuvant CRT from January 2012 to June 2023 in the Department of Gynecology and Oncology of the First Affiliated Hospital of Shandong First Medical University. Neutropenia was defined as an Absolute Neutrophil Count (ANC) of peripheral blood neutrophils below 2 × 109/L. Factors affecting neutropenia in EC patients treated with CRT using Generalized Estimating Equation (GEE), and Logistic regression was used to further analyze the effect of adding radiotherapy to different chemotherapy cycles on neutropenia, so that patients receive optimal adjuvant CRT while the risk of neutropenia is appropriately controlled. RESULTS: A total of 144 patients met the inclusion criteria. They underwent 330 cycles of adjuvant chemotherapy, of whom 96 (66.7%) developed neutropenia, which occurred 140 times. The results of one-way GEE analysis showed that before CRT, White Blood Cell (WBC) (OR = 0.827; 95%CI, 0.701-0.976), ANC (OR = 0.749; 95%CI, 0.586-0.957), Absolute Monocyte Count (AMC) (OR = 0.047; 95%CI, 0.008-0.283), Blood Urea Nitrogen (BUN) (OR = 0.857; 95%CI, 0.741-0.991), platinum and docetaxel (platinum/docetaxel) dosing regimen (OR = 2.284; 95%CI, 1.130-4.618) were associated with neutropenia with adjuvant CRT for EC (p < 0.05), results of multifactorial GEE analysis showed that before adjuvant CRT ANC (OR = 0.552; 95%CI, 0.973-2.231), AMC (OR = 0.047; 95%CI, 0.004-0.052), platinum/docetaxel (OR = 2.437; 95%CI, 1.087-5.464) were an independent influence on neutropenia in adjuvant CRT for EC (p < 0.05). Multifactorial Logistic regression shows addition of radiotherapy to the first cycle of chemotherapy (OR = 4.413; 95%CI, 1.238-18.891) was an independent influence of neutropenia (p < 0.05). CONCLUSIONS: Patients with low pre-CRT ANC and AMC, platinum/docetaxel dosing regimens need to be closely monitored during each cycle of CRT. Also, the concurrent addition of radiotherapy should be avoided during the first cycle of chemotherapy.


Subject(s)
Chemoradiotherapy, Adjuvant , Endometrial Neoplasms , Neutropenia , Humans , Female , Retrospective Studies , Endometrial Neoplasms/therapy , Endometrial Neoplasms/drug therapy , Neutropenia/etiology , Middle Aged , Aged , Chemoradiotherapy, Adjuvant/adverse effects , Adult , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Prognosis , Docetaxel/administration & dosage , Docetaxel/adverse effects , Risk Factors
13.
Molecules ; 29(11)2024 May 25.
Article in English | MEDLINE | ID: mdl-38893380

ABSTRACT

Breast cancer is a major health concern and the leading cause of death among women worldwide. Standard treatment often involves surgery, radiotherapy, and chemotherapy, but these come with side effects and limitations. Researchers are exploring natural compounds like baicalin and baicalein, derived from the Scutellaria baicalensis plant, as potential complementary therapies. This study investigated the effects of baicalin and baicalein on the cytotoxic, proapoptotic, and genotoxic activity of doxorubicin and docetaxel, commonly used chemotherapeutic drugs for breast cancer. The analysis included breast cancer cells (MCF-7) and human endothelial cells (HUVEC-ST), to assess potential effects on healthy tissues. We have found that baicalin and baicalein demonstrated cytotoxicity towards both cell lines, with more potent effects observed in baicalein. Both flavonoids, baicalin (167 µmol/L) and baicalein (95 µmol/L), synergistically enhanced the cytotoxic, proapoptotic, and genotoxic activity of doxorubicin and docetaxel in breast cancer cells. In comparison, their effects on endothelial cells were mixed and depended on concentration and time. The results suggest that baicalin and baicalein might be promising complementary agents to improve the efficacy of doxorubicin and docetaxel anticancer activity. However, further research is needed to validate their safety and efficacy in clinical trials.


Subject(s)
Apoptosis , Breast Neoplasms , Docetaxel , Doxorubicin , Flavanones , Flavonoids , Humans , Flavonoids/pharmacology , Flavanones/pharmacology , Docetaxel/pharmacology , Doxorubicin/pharmacology , MCF-7 Cells , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , DNA Damage/drug effects , Drug Synergism , Antineoplastic Agents/pharmacology , Cell Survival/drug effects , Human Umbilical Vein Endothelial Cells/drug effects
14.
Sci Rep ; 14(1): 14083, 2024 06 18.
Article in English | MEDLINE | ID: mdl-38890326

ABSTRACT

Hand-foot syndrome (HFS) is a frequently occurring and treatment-requiring adverse effect of docetaxel. We previously reported that systemic dexamethasone (DEX) prevents the other docetaxel-induced adverse inflammatory effects in a dose-dependent manner. This study aimed to evaluate the dose-dependent efficacy of systemic DEX in attenuating HFS in patients with breast cancer receiving docetaxel. Patients with breast cancer receiving docetaxel (75 mg/m2)-containing regimens (n = 111) were divided into 4 and 8 mg/day DEX groups, with each DEX dose administered on days 2-4, and analyzed retrospectively. Development of all-grade HFS in all treatment cycles was significantly lower in the 8 mg group (50.0%) than in the 4 mg group (73.0%, P = 0.03), with primary endpoint accomplishment. Moreover, its development in the first cycle was also lower in the 8 mg group than in the 4 mg group. These results were confirmed in a propensity score-matched population. Logistic regression analysis suggested higher DEX dosage as an independent preventive factor (adjusted odds ratio 0.35; 95% confidence interval 0.14-0.86, P = 0.02 for all cycles; 0.26, 0.11-0.63, P = 0.003 for the first cycle). Our study suggests that systemic DEX prevents the occurrence of docetaxel-induced HFS in patients with breast cancer in a dose-dependent manner in a real-world setting.


Subject(s)
Breast Neoplasms , Dexamethasone , Docetaxel , Hand-Foot Syndrome , Humans , Docetaxel/adverse effects , Docetaxel/administration & dosage , Dexamethasone/administration & dosage , Dexamethasone/adverse effects , Dexamethasone/therapeutic use , Female , Breast Neoplasms/drug therapy , Hand-Foot Syndrome/etiology , Middle Aged , Retrospective Studies , Aged , Adult , Dose-Response Relationship, Drug , Antineoplastic Agents/adverse effects , Antineoplastic Agents/administration & dosage
16.
Int J Nanomedicine ; 19: 5071-5094, 2024.
Article in English | MEDLINE | ID: mdl-38846644

ABSTRACT

Background: The commercial docetaxel (DTX) formulation causes severe side effects due to polysorbate 80 and ethanol. Novel surfactant-free nanoparticle (NP) systems are needed to improve bioavailability and reduce side effects. However, controlling the particle size and stability of NPs and improving the batch-to-batch variation are the major challenges. Methods: DTX-loaded bovine serum albumin nanoparticles (DTX-BSA-NPs) were prepared by a novel thermal-driven self-assembly/microfluidic technology. Single-factor analysis and orthogonal test were conducted to obtain the optimal formulation of DTX-BSA-NPs in terms of particle size, encapsulation efficiency (EE), and drug loading (DL). The effects of oil/water flow rate and pump pressure on the particle size, EE, and DL were investigated to optimize the preparation process of DTX-BSA-NPs. The drug release, physicochemical properties, stability, and pharmacokinetics of NPs were evaluated. Results: The optimized DTX-BSA-NPs were uniform, with a particle size of 118.30 nm, EE of 89.04%, and DL of 8.27%. They showed a sustained release of 70% over 96 hours and an increased stability. There were some interactions between the drug and excipients in DTX-BSA-NPs. The half-life, mean residence time, and area under the curve (AUC) of DTX-BSA-NPs increased, but plasma clearance decreased when compared with DTX. Conclusion: The thermal-driven self-assembly/microfluidic combination method effectively produces BSA-based NPs that improve the bioavailability and stability of DTX, offering a promising alternative to traditional formulations.


Subject(s)
Biological Availability , Docetaxel , Drug Stability , Nanoparticles , Particle Size , Serum Albumin, Bovine , Docetaxel/pharmacokinetics , Docetaxel/chemistry , Docetaxel/administration & dosage , Animals , Serum Albumin, Bovine/chemistry , Serum Albumin, Bovine/pharmacokinetics , Serum Albumin, Bovine/administration & dosage , Nanoparticles/chemistry , Taxoids/pharmacokinetics , Taxoids/chemistry , Taxoids/administration & dosage , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/chemistry , Antineoplastic Agents/administration & dosage , Drug Liberation , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Rats, Sprague-Dawley , Male , Drug Compounding/methods , Rats
17.
J Int Med Res ; 52(6): 3000605241254778, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38835107

ABSTRACT

Pulmonary blastoma (PB) is a rare, highly malignant tumor prone to distant metastasis and recurrence, and the prognosis of these patients is often poor. We report a case of metastatic PB with a good prognosis with the aim of providing data to support a clinical diagnosis and treatment. In December 2015, a 43-year-old male patient was admitted to our hospital because of a cough and blood-stained sputum. Positron emission-computed tomography showed massive high-density imaging in the lower lobe of the right lung, with a maximum cross-section of 76 × 58 mm. Thoracoscopic-assisted right lower lobectomy with lymph node dissection was performed. After 1 month, computed tomography showed a high possibility of metastasis. The patient then received docetaxel and cisplatin chemotherapy for a total of six courses. After chemotherapy, enhanced computed tomography showed considerable absorption of pleural effusion, and a left lobe pulmonary nodule was not detected. The postoperative pathological diagnosis was PB, and epithelial and mesenchymal differentiation components were observed. The patient continued to visit the hospital regularly for re-examination and imaging examinations. Currently, no signs of recurrence or distant metastasis have been detected.


Subject(s)
Lung Neoplasms , Pulmonary Blastoma , Humans , Male , Adult , Pulmonary Blastoma/diagnosis , Pulmonary Blastoma/pathology , Pulmonary Blastoma/surgery , Lung Neoplasms/pathology , Lung Neoplasms/diagnosis , Lung Neoplasms/secondary , Lung Neoplasms/surgery , Lung Neoplasms/diagnostic imaging , Prognosis , Tomography, X-Ray Computed , Positron Emission Tomography Computed Tomography , Cisplatin/therapeutic use , Cisplatin/administration & dosage , Pneumonectomy , Docetaxel/therapeutic use , Docetaxel/administration & dosage
18.
Curr Med Res Opin ; 40(7): 1137-1143, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38857167

ABSTRACT

OBJECTIVE: This study aimed to assess the efficacy and safety of FOLFIRI and paclitaxel in patients with advanced gastric cancer (AGC) who were previously treated with first-line modified docetaxel, cisplatin, 5-fluorouracil (mDCF), or 5-fluorouracil, oxaliplatin, docetaxel (FLOT). METHODS: Patients who received a triplet regimen in the first line setting and were treated with FOLFIRI or paclitaxel in the second-line treatment were included. RESULTS: The study included 198 patients, with 115 receiving FOLFIRI and 83 receiving paclitaxel. The median age was 58 (range = 24-69). The median progression-free survival (mPFS) was 5.2 [95% confidence interval (CI) = 4.4-5.5] months in the FOLFIRI arm, and 4.1 (95% CI = 3.3-4.6) months in the paclitaxel arm (p = .007). The median overall survival (mOS) was 9.4 (95% CI = 7.4-10.5) months in the FOLFIRI arm and 7.2 (95% CI = 5.6-8.3) months in the paclitaxel arm (p = .008). Grade 3-4 neuropathy was higher in patients receiving paclitaxel compared to those receiving FOLFIRI (p = .04). Grade 3-4 diarrhea was 8% in the FOLFIRI arm and 2.4% in the paclitaxel arm (p = .02). CONCLUSION: Beyond progression with docetaxel-based triplet chemotherapy, FOLFIRI may be preferred as a second-line treatment over paclitaxel due to its longer mPFS and mOS.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Fluorouracil , Stomach Neoplasms , Taxoids , Humans , Middle Aged , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Stomach Neoplasms/mortality , Female , Male , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Taxoids/administration & dosage , Taxoids/therapeutic use , Taxoids/adverse effects , Fluorouracil/administration & dosage , Fluorouracil/therapeutic use , Fluorouracil/adverse effects , Turkey , Young Adult , Paclitaxel/administration & dosage , Paclitaxel/adverse effects , Docetaxel/administration & dosage , Docetaxel/therapeutic use , Leucovorin/administration & dosage , Leucovorin/therapeutic use , Leucovorin/adverse effects , Treatment Outcome , Oxaliplatin/administration & dosage , Oxaliplatin/adverse effects , Oxaliplatin/therapeutic use , Bridged-Ring Compounds/administration & dosage , Bridged-Ring Compounds/therapeutic use , Bridged-Ring Compounds/adverse effects , Camptothecin/analogs & derivatives , Camptothecin/administration & dosage , Camptothecin/therapeutic use , Camptothecin/adverse effects
19.
Chem Biol Drug Des ; 103(6): e14562, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38898371

ABSTRACT

Docetaxel (DTX) resistance poses a significant challenge in the treatment of prostate cancer (PCa), often leading to chemotherapy failure. This study investigates the ability of piperine, a compound derived from black pepper, to enhance the sensitivity of PCa cells to DTX and elucidates its underlying mechanism. We established a DTX-resistant PCa cell line, DU145/DTX, to conduct our experiments. Through a series of assays, including MTT for cell viability, flow cytometry for apoptosis, Transwell for cell migration and invasion, and western blot for protein expression analysis, we assessed the effects of piperine on these cellular functions and on the Notch signaling pathway components. Our results demonstrated that we successfully established the DTX-resistant PCa cell line DU145/DTX. Piperine effectively decreased the viability of both DU145 and its DTX-resistant counterpart, DU145/DTX, in a concentration and time-dependent manner when used alone and in combination with DTX. Notably, piperine also induced apoptosis and reduced the migration and invasion capabilities of these cells. At the molecular level, piperine down-regulated the Notch pathway by inhibiting Notch1 and Jagged1 signaling, as well as reducing the expression of downstream effectors Hey1 and hes family bHLH transcription factor 1. The study concludes that piperine's ability to modulate the Notch signaling pathway and induce apoptosis highlights its potential as a complementary treatment for DTX-resistant PCa, paving the way for the use of traditional Chinese medicinal compounds in modern oncology treatment strategies.


Subject(s)
Alkaloids , Apoptosis , Benzodioxoles , Cell Movement , Docetaxel , Drug Resistance, Neoplasm , Piperidines , Polyunsaturated Alkamides , Prostatic Neoplasms , Signal Transduction , Polyunsaturated Alkamides/pharmacology , Polyunsaturated Alkamides/chemistry , Humans , Benzodioxoles/pharmacology , Benzodioxoles/chemistry , Alkaloids/pharmacology , Alkaloids/chemistry , Piperidines/pharmacology , Piperidines/chemistry , Docetaxel/pharmacology , Male , Cell Line, Tumor , Signal Transduction/drug effects , Drug Resistance, Neoplasm/drug effects , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Apoptosis/drug effects , Cell Movement/drug effects , Receptors, Notch/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Cell Survival/drug effects , Receptor, Notch1/metabolism
20.
Clin Drug Investig ; 44(7): 513-525, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38937403

ABSTRACT

BACKGROUND AND OBJECTIVE: Trastuzumab targets human epidermal growth factor receptor 2 (HER2) receptors and is indicated for treating HER2-positive metastatic breast cancer. BP02, a recombinant IgG1 kappa humanized monoclonal antibody, is being developed as a trastuzumab biosimilar. The objective of this study was to evaluate the equivalence of BP02 with reference trastuzumab (RT: Herceptin®-EU) in patients with HER2-positive metastatic breast cancer. METHODS: This double-blinded, 1:1 randomized, parallel-group, active-controlled, phase III equivalence trial recruited women aged 18-75 years with histologically/cytologically confirmed HER2- positive, locally recurrent or metastatic breast cancer with systemic metastasis, from 59 sites in India. Patients were randomly allocated 1:1 stratified by estrogen receptor/progesterone receptor status to receive BP02/RT (8-mg/kg loading dose on day 1-cycle 1, 6 mg/kg on day 1-cycles 2-8, of each 3-week cycle) combined with docetaxel (75 mg/m2 on day 1-cycles 1-8) [induction phase]. Participants with complete or partial response, or stable disease at the end of the induction phase continued the study drug until disease progression/treatment discontinuation [maintenance phase]. The primary efficacy endpoint was the objective response rate per Response Evaluation Criteria in Solid Tumors (RECIST) 1.1. RESULTS: Between 23 September, 2020 and 16 September, 2022, 690 patients were recruited (n = 345 each to BP02/RT). At the end of the induction phase (intent-to-treat population), a similar proportion of patients achieved an objective response rate with BP02 (n = 231 [67.0%], 95% confidence interval [CI] 62.0, 71.9) and RT (n = 238 [69.0%], 95% CI 64.1, 73.9). The 95% CI of risk difference (-2.03, 95% CI -9.15, 5.09) and 90% CI of risk ratio (0.97, 90% CI 0.89, 1.06) were within equivalence margins of ± 13% and (0.80, 1.25), respectively. Treatment-emergent adverse events leading to treatment withdrawal were reported in 2.9% and 3.2% patients with BP02 and RT, respectively. CONCLUSIONS: BP02 showed an equivalent efficacy and similar safety profile to RT at the end of 24 weeks. CLINICAL TRIAL REGISTRATION: CTRI Number: CTRI/2020/04/024456.


Subject(s)
Biosimilar Pharmaceuticals , Breast Neoplasms , Receptor, ErbB-2 , Trastuzumab , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Middle Aged , Biosimilar Pharmaceuticals/administration & dosage , Biosimilar Pharmaceuticals/therapeutic use , Biosimilar Pharmaceuticals/adverse effects , Biosimilar Pharmaceuticals/pharmacology , Receptor, ErbB-2/metabolism , Adult , Double-Blind Method , Trastuzumab/administration & dosage , Trastuzumab/therapeutic use , Trastuzumab/adverse effects , Aged , Young Adult , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/adverse effects , Adolescent , Docetaxel/therapeutic use , Docetaxel/administration & dosage , Docetaxel/adverse effects , Neoplasm Metastasis , India , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...