Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 17 de 17
Filtrer
1.
Cureus ; 16(9): e68621, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39371844

RÉSUMÉ

Oxaliplatin is used in combination with fluorouracil and leucovorin as part of the FOLFOX (fluorouracil, leucovorin, and oxaliplatin) regimen for colorectal cancer (CRC). Oxaliplatin has been shown to cause thrombocytopenia in a majority of CRC patients receiving this drug. Although this thrombocytopenia mainly occurs through myelosuppression, in rare cases, it can be immune-mediated. However, unlike other chemotherapy-induced myelosuppression, oxaliplatin-induced thrombocytopenia presents with a sudden drop within hours to days of oxaliplatin administration. The majority of cases who present with oxaliplatin-induced thrombocytopenia typically present after actively being treated with oxaliplatin. Here, we present the case of a 59-year-old female with biopsy-proven CRC on FOLFOX therapy found to have oxaliplatin antibody-mediated thrombocytopenia. She was originally treated with FOLFOX; however, due to response and clinical symptoms, her regimen was changed to include FOLFIRI (leucovorin calcium, fluorouracil, and irinotecan hydrochloride) and bevacizumab before reinitiating FOLFOX due to disease progression. During this case, she presented with rectal bleeding and was found to have severe thrombocytopenia. She was treated with platelet transfusion, intravenous immunoglobulin, and steroids for concerns of immune thrombocytopenia; however, through the use of flow cytometry, oxaliplatin and leucovorin antibodies were discovered. Ultimately, oxaliplatin was permanently discontinued due to concerns about further events of thrombocytopenia.

2.
Explore (NY) ; 20(5): 103031, 2024.
Article de Anglais | MEDLINE | ID: mdl-38986288

RÉSUMÉ

Neutropenia, a common side effect of chemotherapy for ovarian cancer, was observed in a 47-year-old female patient undergoing a six-cycle chemotherapy regimen. She experienced recurrent neutropenia and leukopenia but refused granulocyte colony-stimulating factor (G-CSF) due to severe bone pain and high costs. Moxibustion combined with guasha therapy (MGT) was administered each time neutropenia occurred. The treatment involved guasha therapy on the bladder meridian (BL) and the governor vessel (GV), followed by moxibustion at Zhongwan (CV 12), Guanyuan (CV 4), and Shenzhu (GV 12) points over 2-3 days. This approach led to the recovery of neutrophil and leukocyte counts, enabling the patient to complete six chemotherapy cycles without G-CSF. These findings suggest that MGT may enhance neutrophil and leukocyte counts in patients with chemotherapy-induced myelosuppression, presenting a potential alternative for those intolerant to G-CSF. However, further high-quality research is needed to confirm its efficacy.


Sujet(s)
Moxibustion , Neutropénie , Tumeurs de l'ovaire , Humains , Femelle , Adulte d'âge moyen , Neutropénie/thérapie , Neutropénie/induit chimiquement , Tumeurs de l'ovaire/traitement médicamenteux , Moxibustion/méthodes , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Numération des leucocytes , Granulocytes neutrophiles , Association thérapeutique , Récidive
3.
Cancer Chemother Pharmacol ; 93(2): 151-159, 2024 02.
Article de Anglais | MEDLINE | ID: mdl-37926753

RÉSUMÉ

BACKGROUND: The association of chemotherapy-induced myelosuppression with tumor response and overall survival remained controversial. The study was conducted to investigate the association between them in small cell lung cancer (SCLC). METHODS: 204 eligible patients with SCLC were respectively included and categorized into three groups (no, mild, and severe myelosuppression) based on myelosuppression degree after the first chemotherapy. Curative efficacy of 2-cycle chemotherapy was evaluated by the objective response rate (ORR) and disease control rate (DCR). Univariate and multivariate logistic regression analyses were conducted to investigate their association. Receiver operator characteristic (ROC) curves, net reclassification index (NRI), and integrated discrimination improvement (IDI) were used to assess the predictive ability of myelosuppression. RESULTS: In the fully-adjusted model, mild (OR, 4.61; 95% CI, 1.35 to 18.27; P = 0.020) and severe (OR, 7.22; 95% CI, 1.30 to 72.44; P = 0.046) myelosuppression were positively associated with DCR. However, only mild myelosuppression was significantly associated with ORR (OR, 2.78; 95% CI, 1.30 to 6.14; P = 0.010). Although we observed evidence of increased ORR in severe myelosuppression, the difference was not statistically significant. Furthermore, based on the results of the ROC curve, NRI and IDI, chemotherapy-induced myelosuppression cannot be used as a accurate and independent predictor for curative efficacy, but it can improve overall prediction accuracy. CONCLUSION: Chemotherapy-induced myelosuppression was significantly associated with curative efficacy of 2-cycle chemotherapy in SCLC, which could help predict treatment efficacy and guide chemotherapy dosage.


Sujet(s)
Antinéoplasiques , Tumeurs du poumon , Carcinome pulmonaire à petites cellules , Humains , Carcinome pulmonaire à petites cellules/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Résultat thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Antinéoplasiques/usage thérapeutique
4.
Zhongguo Zhong Yao Za Zhi ; 48(13): 3612-3622, 2023 Jul.
Article de Chinois | MEDLINE | ID: mdl-37474994

RÉSUMÉ

This study aimed to analyze the effect of Bletilla striata polysaccharide(BSP) on endogenous metabolites in serum of tumor-bearing mice treated with 5-fluorouracil(5-FU) by untargeted metabolomics techniques and explore the mechanism of BSP in alleviating the toxic and side effects induced by 5-FU. Male BALB/C mice were randomly divided into a normal group, a model group, a 5-FU group, and a 5-FU + BSP group, with eight mice in each group. Mouse colon cancer cells(CT26) were transplanted into the mice except for those in the normal group to construct the tumor-bearing mouse model by subcutaneous injection, and 5-FU chemotherapy and BSP treatment were carried out from the second day of modeling. The changes in body weight, diarrhea, and white blood cell count in the peripheral blood were recorded. The mice were sacrificed and sampled when the tumor weight of mice in the model group reached approximately 1 g. TUNEL staining was used to detect the cell apoptosis in the small intestine of each group. The proportions of hematopoietic stem cells and myeloid progenitor cells in bone marrow were measured by flow cytometry. Five serum samples were selected randomly from each group for untargeted metabolomics analysis. The results showed that BSP was not effective in inhibiting colon cancer in mice, but diarrhea, leukopenia, and weight loss caused by 5-FU chemotherapy were significantly improved after BSP intervention. In addition, apoptotic cells decreased in the small intestinal tissues and the percentages of hematopoietic stem cells and myeloid progenitor cells in bone marrow were significantly higher after BSP treatment. Metabolomics results showed that the toxic and side effects of 5-FU resulted in significant decrease in 29 metabolites and significant increase in 22 metabolites in mouse serum. Among them, 19 disordered metabolites showed a return to normal levels in the 5-FU+BSP group. The results of pathway enrichment indicated that metabolic pathways mainly involved pyrimidine metabolism, arachidonic acid metabolism, and steroid hormone biosynthesis. Therefore, BSP may ameliorate the toxic and side effects of 5-FU in the intestinal tract and bone marrow presumably by regulating nucleotide synthesis, inflammatory damage, and hormone production.


Sujet(s)
Tumeurs du côlon , Fluorouracil , Animaux , Mâle , Souris , Tumeurs du côlon/traitement médicamenteux , Diarrhée , Fluorouracil/effets indésirables , Hormones , Métabolomique , Souris de lignée BALB C , Polyosides/pharmacologie
5.
Adv Ther ; 40(10): 4189-4215, 2023 10.
Article de Anglais | MEDLINE | ID: mdl-37490258

RÉSUMÉ

INTRODUCTION: Trilaciclib was recently approved in the USA for reducing chemotherapy-induced myelosuppression (CIM) among adults with extensive-stage small cell lung cancer (ES-SCLC) when administered prior to chemotherapy. There is limited understanding of real-world outcomes of trilaciclib. METHODS: A comprehensive literature review was conducted using a keyword search in the MEDLINE, Embase, and conference abstracts. Additional studies were identified through communications with the authors of relevant studies. Published and unpublished real-world studies of trilaciclib- and comparable non-trilaciclib-treated patients with ES-SCLC were included. Evidence on myelosuppressive hematologic adverse events (HAEs), cytopenia-related healthcare utilization, and other reported outcomes (e.g., hospitalizations, dose reduction, and treatment delay) were synthesized. If feasible, outcomes were compared qualitatively between the trilaciclib and historical reference groups, and between first-line trilaciclib initiators and the overall trilaciclib population. Weighted averages were estimated for selected outcomes using sample size as the weight. RESULTS: The literature search identified five unique studies based on eight records-two included trilaciclib only, two non-trilaciclib only, and one both. In trilaciclib cohorts, the weighted average prevalence of grade ≥ 3 myelosuppressive HAEs in ≥ 1 lineage, ≥ 2 lineages, and all three lineages was 40.5%, 14.5%, and 7.5%, respectively. All rates were numerically lower compared to the historical non-trilaciclib cohorts (58.8%, 28.0%, 13.0% respectively). Cytopenia-related healthcare utilization was also lower in the trilaciclib cohorts. In general, first-line trilaciclib initiators had numerically lower myelosuppressive HAEs and cytopenia-related healthcare utilization than the overall trilaciclib patients. CONCLUSIONS: The existing evidence suggests that trilaciclib may reduce single and multilineage grade ≥ 3 myelosuppressive HAEs and cytopenia-related healthcare utilization among patients with ES-SCLC in the real world. It is a promising new treatment for CIM prevention in ES-SCLC and may bring greater benefits to first-line trilaciclib initiators. Future studies are recommended to further evaluate the real-world effectiveness of trilaciclib.


Sujet(s)
Tumeurs du poumon , Carcinome pulmonaire à petites cellules , Adulte , Humains , Carcinome pulmonaire à petites cellules/traitement médicamenteux , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Tumeurs du poumon/traitement médicamenteux
6.
Int J Mol Sci ; 24(3)2023 Feb 03.
Article de Anglais | MEDLINE | ID: mdl-36769312

RÉSUMÉ

STS1 and STS2, as the protein phosphatases that dephosphorylate FLT3 and cKIT, negatively regulate the self-renewal and differentiation of hematopoietic stem and progenitor cells (HSPCs). To obtain the small molecule inhibitors of STS1/STS2 phosphatase activity used to expand HSPCs both in vitro and in vivo, we establish an in vitro phosphatase assay using the recombinant proteins of the STS1/STS2 histidine phosphatase (HP) domain, by which we screened out baicalein (BC) as one of the effective inhibitors targeting STS1 and STS2. Then, we further demonstrate the direct binding of BC with STS1/STS2 using molecular docking and capillary electrophoresis and verify that BC can restore the phosphorylation of FLT3 and cKIT from STS1/STS2 inhibition. In a short-term in vitro culture, BC promotes profound expansion and enhances the colony-forming capacity of both human and mouse HSPCs along with the elevation of phospho-FLT3 and phospho-cKIT levels. Likewise, in vivo administration with BC significantly increases the proportions of short-term hematopoietic stem cells (ST-HSCs), multipotent progenitors (MPPs) and especially long-term HSCs (LT-HSCs) in healthy mouse bone marrow and increases the numbers of colony-forming units (CFU) formed by HSPCs as well. More importantly, pre-administration of BC significantly enhances the survival of mice with lethal 5-fluorouracil (5-FU) injection due to the alleviation of 5-FU-induced myelosuppression, as evidenced by the recovery of bone marrow histologic injury, the increased proportions of LT-HSCs, ST-HSCs and MPPs, and enhanced colony-forming capacity. Collectively, our study not only suggests BC as one of the small molecule candidates to stimulate HSPC expansion both in vitro and in vivo when needed in either physiologic or pathologic conditions, but also supports STS1/STS2 as potential therapeutic drug targets for HSPC expansion and hematopoietic injury recovery.


Sujet(s)
Fluorouracil , Cellules souches hématopoïétiques , Animaux , Humains , Souris , Différenciation cellulaire , Fluorouracil/pharmacologie , Fluorouracil/métabolisme , Cellules souches hématopoïétiques/métabolisme , Simulation de docking moléculaire , Phosphoric monoester hydrolases/métabolisme , Cellules souches
7.
Cancers (Basel) ; 15(4)2023 Feb 08.
Article de Anglais | MEDLINE | ID: mdl-36831423

RÉSUMÉ

Purpose: Develop and validate an accessible prediction model using machine learning (ML) to predict the risk of chemotherapy-induced myelosuppression (CIM) in children with Wilms' tumor (WT) before chemotherapy is administered, enabling early preventive management. Methods: A total of 1433 chemotherapy cycles in 437 children with WT who received chemotherapy in our hospital from January 2009 to March 2022 were retrospectively analyzed. Demographic data, clinicopathological characteristics, hematology and blood biochemistry baseline results, and medication information were collected. Six ML algorithms were used to construct prediction models, and the predictive efficacy of these models was evaluated to select the best model to predict the risk of grade ≥ 2 CIM in children with WT. A series of methods, such as the area under the receiver operating characteristic curve (AUROC), the calibration curve, and the decision curve analysis (DCA) were used to test the model's accuracy, discrimination, and clinical practicability. Results: Grade ≥ 2 CIM occurred in 58.5% (839/1433) of chemotherapy cycles. Based on the results of the training and validation cohorts, we finally identified that the extreme gradient boosting (XGB) model has the best predictive efficiency and stability, with an AUROC of up to 0.981 in the training set and up to 0.896 in the test set. In addition, the calibration curve and the DCA showed that the XGB model had the best discrimination and clinical practicability. The variables were ranked according to the feature importance, and the five variables contributing the most to the model were hemoglobin (Hgb), white blood cell count (WBC), alkaline phosphatase, coadministration of highly toxic chemotherapy drugs, and albumin. Conclusions: The incidence of grade ≥ 2 CIM was not low in children with WT, which needs attention. The XGB model was developed to predict the risk of grade ≥ 2 CIM in children with WT for the first time. The model has good predictive performance and stability and has the potential to be translated into clinical applications. Based on this modeling and application approach, the extension of CIM prediction models to other pediatric malignancies could be expected.

8.
Article de Chinois | WPRIM (Pacifique Occidental) | ID: wpr-981492

RÉSUMÉ

This study aimed to analyze the effect of Bletilla striata polysaccharide(BSP) on endogenous metabolites in serum of tumor-bearing mice treated with 5-fluorouracil(5-FU) by untargeted metabolomics techniques and explore the mechanism of BSP in alleviating the toxic and side effects induced by 5-FU. Male BALB/C mice were randomly divided into a normal group, a model group, a 5-FU group, and a 5-FU + BSP group, with eight mice in each group. Mouse colon cancer cells(CT26) were transplanted into the mice except for those in the normal group to construct the tumor-bearing mouse model by subcutaneous injection, and 5-FU chemotherapy and BSP treatment were carried out from the second day of modeling. The changes in body weight, diarrhea, and white blood cell count in the peripheral blood were recorded. The mice were sacrificed and sampled when the tumor weight of mice in the model group reached approximately 1 g. TUNEL staining was used to detect the cell apoptosis in the small intestine of each group. The proportions of hematopoietic stem cells and myeloid progenitor cells in bone marrow were measured by flow cytometry. Five serum samples were selected randomly from each group for untargeted metabolomics analysis. The results showed that BSP was not effective in inhibiting colon cancer in mice, but diarrhea, leukopenia, and weight loss caused by 5-FU chemotherapy were significantly improved after BSP intervention. In addition, apoptotic cells decreased in the small intestinal tissues and the percentages of hematopoietic stem cells and myeloid progenitor cells in bone marrow were significantly higher after BSP treatment. Metabolomics results showed that the toxic and side effects of 5-FU resulted in significant decrease in 29 metabolites and significant increase in 22 metabolites in mouse serum. Among them, 19 disordered metabolites showed a return to normal levels in the 5-FU+BSP group. The results of pathway enrichment indicated that metabolic pathways mainly involved pyrimidine metabolism, arachidonic acid metabolism, and steroid hormone biosynthesis. Therefore, BSP may ameliorate the toxic and side effects of 5-FU in the intestinal tract and bone marrow presumably by regulating nucleotide synthesis, inflammatory damage, and hormone production.


Sujet(s)
Animaux , Mâle , Souris , Tumeurs du côlon/traitement médicamenteux , Diarrhée , Fluorouracil/effets indésirables , Hormones , Métabolomique , Souris de lignée BALB C , Polyosides/pharmacologie
9.
Quant Imaging Med Surg ; 12(3): 1871-1881, 2022 Mar.
Article de Anglais | MEDLINE | ID: mdl-35284263

RÉSUMÉ

Background: Systemic lung cancer treatment-induced changes in bone marrow attenuation assessed via dual-energy CT-based virtual non-calcium (VNCa) imaging of the axial skeleton and their relationship to hematological laboratory have not yet been investigated. Methods: VNCa bone marrow images of the axial skeleton derived from 93 unenhanced reduced dose dual-energy CTs of the thorax and abdomen of 31 patients were retrospectively analyzed. Each patient had received one pre-therapy baseline exam and two consecutive follow-up exams (FU1 and FU2) at a mean of 7.7 and 11.7 weeks after start of therapy. Concurrent hematologic laboratory data were available for every exam. Seven regions of interest were placed in the spine and pelvis and mean VNCa bone marrow attenuation was measured. Twenty-two Patients receiving highly myelotoxic treatment (Group A) were compared to 9 patients receiving less toxic substances (Group B). Results: Median bone marrow attenuation in Group A/Group B was -31.8 HU (IQR 12.7)/40.6 HU (IQR, 12.2) at baseline, -46.5 HU (IQR, 12.5)/-43.8 HU (15.7) at FU1 and -46.9 HU (IQR, 22.0)/-38.5 HU (IQR, 18.5) at FU2. In both subgroups the reduction of the mean attenuation between baseline and FU1 was statistically significant although in Group A it was more pronounced; no significant difference was found between FU1 and FU2. The differences between Groups were not statistically significant. Leukopenia rates at FU1 and FU 2 were 50% and 54.5% in Group A and 0% and 22% in Group B. Anemia rates rose from 31.8% at baseline to 90% at FU1 and 86.4% at FU2 in Group A and fell from 77.8% at baseline to 66.7% at FU1 and further to 55.6% at FU2 in Group B. Conclusions: Both highly myelotoxic as well as-to a smaller degree-less myelotoxic systemic therapy led to a significant drop in bone marrow attenuation with no significant tendency towards subsequent elevation irrespective of the treatment's degree of toxicity or the presence of myelosuppression and not even under hematological supportive therapy. The results suggest that in this clinical setting an increase in bone marrow attenuation should be regarded as suspicious for tumor infiltration.

10.
J Med Econ ; 24(sup1): 71-83, 2021 Nov.
Article de Anglais | MEDLINE | ID: mdl-34873975

RÉSUMÉ

AIMS: Proliferating hematopoietic stem and progenitor cells (HSPCs) are susceptible to chemotherapy-induced damage, resulting in myelosuppressive adverse events (AEs) such as neutropenia, anemia, and thrombocytopenia that are associated with high health care costs and decreased quality of life (QoL). In this study, a trial-based cost-effectiveness analysis was performed to help assess the economic impact of administering trilaciclib, a myeloprotective therapy that protects multilineage HSPCs from chemotherapy-induced damage, prior to standard first-line chemotherapy, using data from a pivotal Phase II study of trilaciclib in the setting of extensive-stage small cell lung cancer (ES-SCLC, NCT03041311). METHOD: The aim of this study was to assess the cost-effectiveness of administering trilaciclib prior to chemotherapy versus chemotherapy alone among patients with ES-SCLC from a United States payer perspective. Data on the rate and frequency of myelosuppressive AEs and health utility were derived from the pivotal study of trilaciclib. Costs of managing myelosuppressive AEs and costs of chemotherapy treatment were sourced from published literature. Outcomes included the number of myelosuppressive AEs, costs (in 2021 US dollars), quality-adjusted life-years (QALYs), incremental cost, incremental QALY, and an incremental cost-effectiveness ratio. RESULTS: Administering trilaciclib prior to chemotherapy was associated with a reduction in neutropenia (82%), febrile neutropenia (75%), anemia (43%), and thrombocytopenia (96%) compared with chemotherapy alone. Additionally, trilaciclib prior to chemotherapy was cost-saving compared with chemotherapy alone ($99,919 vs $118,759, respectively) and associated with QALY improvement (0.150 vs 0.145, respectively). Probabilistic sensitivity analyses showed 58% of iterations projecting cost savings and QALY improvement with trilaciclib. CONCLUSIONS: The findings suggest that the use of trilaciclib prior to first-line chemotherapy in patients with ES-SCLC can be cost-beneficial owing to fewer myelosuppressive AEs and lower costs, together with a favorable QoL profile.


Sujet(s)
Antinéoplasiques , Tumeurs du poumon , Carcinome pulmonaire à petites cellules , Antinéoplasiques/effets indésirables , Humains , Tumeurs du poumon/traitement médicamenteux , Pyrimidines , Pyrroles , Qualité de vie
11.
Cancer Chemother Pharmacol ; 87(5): 689-700, 2021 05.
Article de Anglais | MEDLINE | ID: mdl-33595690

RÉSUMÉ

PURPOSE: Trilaciclib is a first-in-class CDK4/6 inhibitor that transiently arrests hematopoietic stem and progenitor cells (HSPCs) in the G1 phase of the cell cycle to preserve them from chemotherapy-induced damage (myelopreservation). We report integrated analyses of preclinical and clinical data that informed selection of the recommended Phase II dose (RP2D) used in trilaciclib trials in extensive-stage small cell lung cancer (ES-SCLC). METHODS: A semi-mechanistic pharmacokinetic/pharmacodynamic (PK/PD) model developed from preclinical data guided selection of an optimal dose for G1 bone marrow arrest in a first-in-human Phase I study (G1T28-1-01). PK, PD, safety, and efficacy data from G1T28-1-01 and two Phase Ib/IIa studies (G1T28-02/-03) in ES-SCLC were analyzed to support RP2D selection. RESULTS: Model simulation of bone marrow arrest based on preclinical data predicted that a ≥ 192 mg/m2 dose would induce a 40-50% decrease in total bone marrow proliferation in humans and almost 100% cell cycle arrest of cycling HSPCs. Consistent with this model, analysis of bone marrow aspirates in healthy volunteers after trilaciclib 192 mg/m2 administration demonstrated almost 100% G1 arrest in HSPCs and 40% decrease in total bone marrow proliferation, with minimal toxicity. G1T28-02/-03 reported similar PK parameters with trilaciclib 200 mg/m2 but slightly lower exposures than expected compared with healthy volunteers; consequently, 240 and 280 mg/m2 doses were also tested to match healthy volunteer exposures. Based on PK and relevant safety data, 240 mg/m2 was selected as the RP2D, which was also favored by myelopreservation endpoints in G1T28-02/-03. CONCLUSION: Integrated PK/PD, safety, and efficacy data support 240 mg/m2 as the RP2D for trilaciclib. CLINICALTRIALS. GOV IDENTIFIERS: NCT02243150; NCT02499770; NCT02514447.


Sujet(s)
Tumeurs du poumon/traitement médicamenteux , Pyrimidines/administration et posologie , Pyrroles/administration et posologie , Carcinome pulmonaire à petites cellules/traitement médicamenteux , Adolescent , Adulte , Essais cliniques comme sujet , Femelle , Humains , Tumeurs du poumon/anatomopathologie , Mâle , Adulte d'âge moyen , Stadification tumorale , Pyrimidines/pharmacocinétique , Pyrimidines/pharmacologie , Pyrroles/pharmacocinétique , Pyrroles/pharmacologie , Carcinome pulmonaire à petites cellules/anatomopathologie , Jeune adulte
12.
Trials ; 21(1): 844, 2020 Oct 12.
Article de Anglais | MEDLINE | ID: mdl-33046128

RÉSUMÉ

BACKGROUND: Traditional Chinese medicine (TCM) has a long history of use in breast cancer, but lacking systematic evidence to support its clinical benefits. In this study, we evaluated the prophylactic and therapeutic effects of moxibustion combined with decoctions for treating chemotherapy-induced myelosuppression (CIM) in early-stage breast cancer patients. METHODS: This is a randomized controlled clinical trial single-blinded for TCM decoction but not moxibustion. Patients are equally divided into the control group without decoction and moxibustion treatment (control), the decoction+moxibustion group (MD), and the placebo+moxibustion group (MP), according to the following stratification factors: age (below 40s, 40s, 50s, and 60s or above), chemotherapy regimen (anthracyclines, taxanes, anthracyclines+taxane, and others), and chemotherapy strategy (adjuvant and neoadjuvant). The TCM decoction is Wenshen Shengbai Decoction. The anticipated sample size is 462 cases (154 cases in each group). All participants are expected to treat with chemotherapy and recombinant human granulocyte colony-stimulating factor (rhG-CSF). The primary outcomes include the proportion of patients with relief of leukopenia and/or neutropenia, the myelosuppression-associated serious adverse event including grade 3-4 leukopenia and/or neutropenia, and febrile neutropenia, and the dose of rhG-CSF. The secondary outcomes include chemotherapy adherence, stratified analysis, adverse reactions, quality of life by EORTC Breast-Cancer-Specific Quality of Life Questionnaire including EORTC QLQ-C30 (V3.0) and QLQ-BR23, TCM Constitution, and 3-year disease-free survival and overall survival. Baseline information including age, surgical approach, chemotherapy regimen and strategy, pathological stage, and molecular subtype will be recorded. DISCUSSION: This will be the first randomized controlled trial to evaluate the efficacy of moxibustion combined with TCM decoction in treating CIM in early-stage breast cancer patients, aiming to standardize the TCM decoction and moxibustion method, thus providing evidence for its clinical benefit. TRIAL REGISTRATION: chictr.org.cn ChiCTR-INR-16009557 . Registered on 23 October 2016.


Sujet(s)
Tumeurs du sein , Moxibustion , Antibiotiques antinéoplasiques/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Femelle , Humains , Médecine traditionnelle chinoise , Qualité de vie , Essais contrôlés randomisés comme sujet
13.
J Pharmacokinet Pharmacodyn ; 46(1): 53-63, 2019 Feb.
Article de Anglais | MEDLINE | ID: mdl-30617672

RÉSUMÉ

In many models of pharmacodynamic systems with delays, a delay of an input is introduced by means of the convolution with the gamma distribution. An approximation of the convolution integral of bound functions based on a system of ordinary differential equations that utilizes properties of the binomial series has been introduced. The approximation converges uniformly on every compact time interval and an estimate of the approximation error has been found [Formula: see text] where [Formula: see text] is the number of differential equations and [Formula: see text] is the shape parameter of the gamma distribution. The accuracy of approximation has been tested on a set of input functions for which the convolution is known explicitly. For tested functions, [Formula: see text] has resulted in an accurate approximation, if [Formula: see text]. However, if [Formula: see text] the error of approximation decreases slowly with increasing [Formula: see text], and [Formula: see text] might be necessary to achieve acceptable accuracy. Finally, the approximation was applied to estimate parameters for the distributed delay model of chemotherapy-induced myelosuppression from previously published WBC count data in rats treated with 5-fluorouracil.


Sujet(s)
Modèles biologiques , Algorithmes , Animaux , Fluorouracil/pharmacologie , Précurseurs des granulocytes/effets des médicaments et des substances chimiques , Humains , Rats
14.
Article de Anglais | WPRIM (Pacifique Occidental) | ID: wpr-691381

RÉSUMÉ

<p><b>OBJECTIVE</b>To investigate the potential efficacy of panaxadiol saponins component (PDS-C), a biologically active fraction isolated from total ginsenosides, to reverse chemotherapy-induced myelosuppression and pancytopenia caused by cyclophamide (CTX).</p><p><b>METHODS</b>Mice with myelosuppression induced by CTX were treated with PDS-C at a low- (20 mg/kg), moderate- (40 mg/kg), or high-dose (80 mg/kg) for 7 consecutive days. The level of peripheral white blood cell (WBC), neutrophil (NEU) and platelet (PLT) were measured, the histopathology and colony formation were observed, the protein kinase and transcription factors in hematopoietic cells were determined by immunohistochemical staining and Western blot.</p><p><b>RESULTS</b>In response to PDS-C therapy, the peripheral WBC, NEU and PLT counts of CTX-induced myelosuppressed mice were significantly increased in a dose-dependent manner. Similarly, bone marrow histopathology examination showed reversal of CTX-induced myelosuppression with increase in overall bone marrow cellularity and the number of hematopoietic cells (P<0.01). PDS-C also promoted proliferation of granulocytic and megakaryocyte progenitor cells in CTX-treated mice, as evidenced by significantly increase in colony formation units-granulocytes/monocytes and -megakaryocytes (P<0.01). The enhancement of hematopoiesis by PDS-C appears to be mediated by an intracellular signaling pathway, this was evidenced by the up-regulation of phosphorylated mitogen-activated protein kinase (p-MEK) and extracellular signal-regulated kinases (p-ERK), and receptor tyrosine kinase (C-kit) and globin transcription factor 1 (GATA-1) in hematopoietic cells of CTX-treated mice (P<0.05).</p><p><b>CONCLUSIONS</b>PDS-C possesses hematopoietic growth factor-like activities that promote proliferation and also possibly differentiation of hematopoietic progenitor cells in myelosuppressed mice, probably mediated by a mechanism involving MEK and ERK protein kinases, and C-kit and GATA-1 transcription factors. PDS-C may potentially be a novel treatment of myelosuppression and pancytopenia caused by chemotherapy.</p>


Sujet(s)
Animaux , Souris , Antinéoplasiques , Prolifération cellulaire , Cyclophosphamide , Extracellular Signal-Regulated MAP Kinases , Métabolisme , Facteur de transcription GATA-1 , Métabolisme , Ginsénosides , Pharmacologie , Utilisations thérapeutiques , Hématopoïèse , Mitogen-Activated Protein Kinase Kinases , Métabolisme , Cellules myéloïdes , Anatomopathologie , Panax , Chimie , Pancytopénie , Traitement médicamenteux , Anatomopathologie , Phosphorylation , Protéines proto-oncogènes c-kit , Métabolisme , Saponines , Pharmacologie , Régulation positive
15.
Chin J Integr Med ; 24(3): 200-206, 2018 Mar.
Article de Anglais | MEDLINE | ID: mdl-28432529

RÉSUMÉ

OBJECTIVE: To investigate the potential efficacy of panaxadiol saponins component (PDS-C), a biologically active fraction isolated from total ginsenosides, to reverse chemotherapy-induced myelosuppression and pancytopenia caused by cyclophamide (CTX). METHODS: Mice with myelosuppression induced by CTX were treated with PDS-C at a low- (20 mg/kg), moderate- (40 mg/kg), or high-dose (80 mg/kg) for 7 consecutive days. The level of peripheral white blood cell (WBC), neutrophil (NEU) and platelet (PLT) were measured, the histopathology and colony formation were observed, the protein kinase and transcription factors in hematopoietic cells were determined by immunohistochemical staining and Western blot. RESULTS: In response to PDS-C therapy, the peripheral WBC, NEU and PLT counts of CTX-induced myelosuppressed mice were significantly increased in a dose-dependent manner. Similarly, bone marrow histopathology examination showed reversal of CTX-induced myelosuppression with increase in overall bone marrow cellularity and the number of hematopoietic cells (P<0.01). PDS-C also promoted proliferation of granulocytic and megakaryocyte progenitor cells in CTX-treated mice, as evidenced by significantly increase in colony formation units-granulocytes/monocytes and -megakaryocytes (P<0.01). The enhancement of hematopoiesis by PDS-C appears to be mediated by an intracellular signaling pathway, this was evidenced by the up-regulation of phosphorylated mitogen-activated protein kinase (p-MEK) and extracellular signal-regulated kinases (p-ERK), and receptor tyrosine kinase (C-kit) and globin transcription factor 1 (GATA-1) in hematopoietic cells of CTX-treated mice (P<0.05). CONCLUSIONS: PDS-C possesses hematopoietic growth factor-like activities that promote proliferation and also possibly differentiation of hematopoietic progenitor cells in myelosuppressed mice, probably mediated by a mechanism involving MEK and ERK protein kinases, and C-kit and GATA-1 transcription factors. PDS-C may potentially be a novel treatment of myelosuppression and pancytopenia caused by chemotherapy.


Sujet(s)
Antinéoplasiques/effets indésirables , Cyclophosphamide/effets indésirables , Ginsénosides/usage thérapeutique , Hématopoïèse/effets des médicaments et des substances chimiques , Cellules myéloïdes/anatomopathologie , Panax/composition chimique , Pancytopénie/traitement médicamenteux , Saponines/pharmacologie , Animaux , Prolifération cellulaire/effets des médicaments et des substances chimiques , Extracellular Signal-Regulated MAP Kinases/métabolisme , Facteur de transcription GATA-1/métabolisme , Ginsénosides/pharmacologie , Souris , Mitogen-Activated Protein Kinase Kinases/métabolisme , Cellules myéloïdes/effets des médicaments et des substances chimiques , Pancytopénie/induit chimiquement , Pancytopénie/anatomopathologie , Phosphorylation/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-kit/métabolisme , Régulation positive/effets des médicaments et des substances chimiques
16.
Cancer Chemother Pharmacol ; 80(2): 343-353, 2017 Aug.
Article de Anglais | MEDLINE | ID: mdl-28656382

RÉSUMÉ

PURPOSE: To investigate whether a more frequent monitoring of the absolute neutrophil counts (ANC) during myelosuppressive chemotherapy, together with model-based predictions, can improve therapy management, compared to the limited clinical monitoring typically applied today. METHODS: Daily ANC in chemotherapy-treated cancer patients were simulated from a previously published population model describing docetaxel-induced myelosuppression. The simulated values were used to generate predictions of the individual ANC time-courses, given the myelosuppression model. The accuracy of the predicted ANC was evaluated under a range of conditions with reduced amount of ANC measurements. RESULTS: The predictions were most accurate when more data were available for generating the predictions and when making short forecasts. The inaccuracy of ANC predictions was highest around nadir, although a high sensitivity (≥90%) was demonstrated to forecast Grade 4 neutropenia before it occurred. The time for a patient to recover to baseline could be well forecasted 6 days (±1 day) before the typical value occurred on day 17. CONCLUSIONS: Daily monitoring of the ANC, together with model-based predictions, could improve anticancer drug treatment by identifying patients at risk for severe neutropenia and predicting when the next cycle could be initiated.


Sujet(s)
Antinéoplasiques/effets indésirables , Modèles biologiques , Tumeurs/traitement médicamenteux , Neutropénie/induit chimiquement , Taxoïdes/effets indésirables , Antinéoplasiques/administration et posologie , Simulation numérique , Docetaxel , Surveillance des médicaments/méthodes , Humains , Numération des leucocytes/méthodes , Neutropénie/prévention et contrôle , Granulocytes neutrophiles/effets des médicaments et des substances chimiques , Granulocytes neutrophiles/métabolisme , Taxoïdes/administration et posologie , Facteurs temps
17.
J Ethnopharmacol ; 152(3): 575-84, 2014 Mar 28.
Article de Anglais | MEDLINE | ID: mdl-24534527

RÉSUMÉ

ETHNOPHARMACOLOGICAL RELEVANCE: Fufang e׳jiao jiang (FEJ), which has been widely used in clinic to replenish qi (vital energy) and nourish blood, is a famous traditional Chinese medicine formula made up of Colla corii asini (donkey-hide gelatin prepared by stewing and concentrating from the hide of Equus asinus Linnaeus.), Radix codonopsis pilosulae (the root of Codonopsis pilosula (Franch.) Nannf.), Radix ginseng rubra (the steamed and dried root of Panax ginseng C.A. Mey.), Fructus crataegi (the fruit of Crataegus pinnatifida Bunge) and Radix rehmanniae preparata (the steamed and sun dried tuber of Rehmannia glutinosa (Gaertn.) Libosch. ex Fisch. & C.A. Mey.). The present study aimed to investigate the hematopoietic effects of FEJ on myelosuppressed mice induced by radiotherapy and chemotherapy systematically and to explore the underlying hematopoietic regulation mechanisms. METHODS: The myelosuppressed mouse model was induced by (60)Co radiation, cyclophosphamide and chloramphenicol. FEJ was then administered by i.g. at the dosages of 5, 10, or 20 mL/kg·d for 10d. The numbers of blood cells from peripheral blood and bone marrow nucleated cells (BMNC) were counted. Body weight and the thymus and spleen indices were also measured. The numbers of hemopoietic progenitor cells and colony-forming unit-fibroblast (CFU-F) were measured in vitro. The ratio of hematopoietic stem cells (HSC) in BMNC, cell cycle and apoptosis of BMNC were determined by flow cytometry. The histology of femoral bone was examined by H&E staining. The levels of transforming growth factor-ß (TGF-ß), tumor necrosis factor-α (TNF-α), erythropoietin (EPO), granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin-3 (IL-3) and interleukin-6 (IL-6) in serum were measured by ELISA. IL-1ß, IL-3, IL-6 mRNA levels in spleen were detected by real-time quantitative PCR (RT-qPCR). In addition, bone marrow stromal cells (BMSC) were cultured in vitro followed by treatment with different doses of FEJ (2.5, 5, 10 µL/mL) for 48 h. Then the levels of cytokines (IL-6, SCF, GM-CSF) in the conditioned media and their mRNA levels in BMSC were determined by ELISA and RT-qPCR, respectively. RESULTS: FEJ could significantly increase the numbers of peripheral blood cells and BMNC, and reverse the loss of body weight and the atrophy of thymus and spleen in a dose-dependent manner. The quantities of hemopoietic progenitor cells and CFU-F in bone marrow were also significantly increased in a dose-dependent manner after FEJ administration. A high-dose FEJ of 20 mL/kg·d could significantly increase the ratio of HSC in BMNC, promote bone marrow cells entering the proliferative cycle phase (S+G2/M) and prevent cells from proceeding to the apoptotic phase. FEJ could also improve the femoral bone marrow morphology. Furthermore, FEJ could increase the levels of GM-CSF and IL-3 and reduce the level of TGF-ß in serum, and enhance the expressions of IL-1ß and IL-3 mRNA in spleen. Lastly, the levels of cytokines (IL-6, SCF, GM-CSF) in the conditioned media and their mRNA levels in BMSC were elevated after treatment with FEJ. CONCLUSIONS: FEJ was clearly confirmed to promote the recovery of bone marrow hemopoietic function in a myelosuppressed mouse model, which may be attributed to (i) improving bone marrow hematopoietic microenvironment; (ii) facilitating the cell proliferation and preventing BMNC from apoptosis; (iii) stimulating the expressions of IL-1ß, IL-3, IL-6, SCF and GM-CSF and inhibiting the expression of TGF-ß.


Sujet(s)
Antinéoplasiques/toxicité , Médicaments issus de plantes chinoises/pharmacologie , Hémopathies/traitement médicamenteux , Lésions radiques expérimentales/traitement médicamenteux , Animaux , Cellules de la moelle osseuse/effets des médicaments et des substances chimiques , Cellules de la moelle osseuse/effets des radiations , Chloramphénicol/toxicité , Cyclophosphamide/toxicité , Cytokines/métabolisme , Relation dose-effet des médicaments , Médicaments issus de plantes chinoises/administration et posologie , Test ELISA , Femelle , Hémopathies/induit chimiquement , Hémopathies/étiologie , Cellules souches hématopoïétiques/effets des médicaments et des substances chimiques , Cellules souches hématopoïétiques/effets des radiations , Mâle , Souris , Souris de lignée BALB C , ARN messager/métabolisme , Réaction de polymérisation en chaine en temps réel
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE