Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Front Nutr ; 10: 1089487, 2023.
Article in English | MEDLINE | ID: mdl-36761228

ABSTRACT

Plant-based functional foods have attracted increasing research interest to validate their use in preventing metabolic disease. Since it is increasingly recognized that inflammation, oxidative stress, and circadian rhythm play vital roles in various metabolic diseases, including diabetes, obesity and non-alcoholic liver disease, plant proteins, protein hydrolysates, and food extracts that intervene in these biological processes are promising dietary supplements to prevent metabolic diseases. Here, we reviewed the recent research on plant-based foods used for metabolic disease prevention and provided new perspectives regarding the current study gaps and future directions in this field.

3.
Food Res Int ; 162(Pt A): 111887, 2022 12.
Article in English | MEDLINE | ID: mdl-36461269

ABSTRACT

Incidence of anxiety and depression has been surging in recent years, causing unignorable mental health crisis across the globe. Mounting studies demonstrated that overgrowth of detrimental gut microbes is driving the development of anxiety and depression. Our previous studies suggested that ferulic acid (FA) and feruloylated oligosaccharides (FOs) were potent in regulating gut microbiome and microbial metabolism in a variety of disease settings, including neuroinflammation. Given the increasing evidence solidifying the role of gut-brain axis in neurological disorders, we here investigated the therapeutic potential of FA and FOs in anxiety and depression. In present study we found that FA and FOs effectively alleviated anxiety and depression-like behavior in mice, while increasing the abundance of Firmicutes, Solibacillus, Acinetobacter and Arthrobacter, and decreasing the abundance of Parabacteroides, Oscollospira and Rummeliibacillus. In addition, FA and FOs were efficacious in enhancing phenylalanine, tyrosine and tryptophan biosynthesis, phenylalanine and caffeine metabolism in mice having depression. Our results validated FA and FOs as effective nutrition to prevent anxiety and depression, as well as provided mechanistic insight into their anti-anxiety and anti-depression function. We suggested that FOs mitigated the symptom of depression in mice potentially via changing gut microbiome structure and microbial metabolism.


Subject(s)
Gastrointestinal Microbiome , Mice , Animals , Anxiety , Oligosaccharides/pharmacology , Oligosaccharides/therapeutic use , Phenylalanine
4.
Food Chem Toxicol ; 166: 113246, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35728726

ABSTRACT

Irinotecan is a first-line treatment for colorectal cancer and the prodrug of 7-ethyl-10-hydroxy-camptothecin (SN-38). However, its fatal gastrointestinal (GI) toxicity raises serious concern. In liver, irinotecan generates its inactive metabolite, SN-38G via UDP-glucuronosyltransferase (UGT)1A1. Subsequently, SN-38G is excreted into GI tract where it is reactivated by microbiome to yield the toxic metabolite, SN-38. Activation of toll-like receptor (TLR)/myeloid differentiation primary response 88 (MyD88) by bacterial endotoxin decreases drug-metabolizing enzymes. In this study, we treated C57BL6/J mice with 50 mg/kg irinotecan once daily until observing grade 4 diarrhea. Mice were sacrificed on day0, day2 and day8. Based on the finding in C57BL6/J mice, we repeated the treatment in Tlr2-/-, Tlr4-/- and Myd88-/- mice to determine the impact of inflammation on UGT metabolism. Our toxicity study in C57BL6/J mice showed that mice started bloody diarrhea after 6 days' injection of irinotecan. Ugt1a1 expression in GI tract started decreasing after 24h since first dose, before the onset of diarrhea. In Tlr4-/- and Myd88-/- mice, no Ugt1a1 reduction was observed in distal GI tract after irinotecan injection. In Tlr2-/- mice, intestinal Ugt1a1 expression was down-regulated. Our results indicate that after two doses of irinotecan, mice started losing capability of detoxifying SN-38. TLR4 plays more important role in Ugt1a1 reduction than TLR2, despite that TLR2 and TLR4 share MyD88 as common adaptor protein. We concluded that irinotecan reduced intestinal Ugt1a1 via TLR4/MyD88 pathway, which eventually triggers the onset of diarrhea. Our finding unveils a novel mechanism underlying irinotecan-induced diarrhea and provides a new direction to prevent chemotherapy side effect.


Subject(s)
Antineoplastic Agents, Phytogenic , Glucuronosyltransferase , Irinotecan , Myeloid Differentiation Factor 88 , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/toxicity , Diarrhea/chemically induced , Diarrhea/metabolism , Glucuronosyltransferase/metabolism , Irinotecan/adverse effects , Mice , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Uridine Diphosphate
5.
Chem Biol Interact ; 360: 109946, 2022 Jun 01.
Article in English | MEDLINE | ID: mdl-35430260

ABSTRACT

Irinotecan, a first-line chemotherapy for gastrointestinal (GI) cancers has been causing fatal toxicities like bloody diarrhea and steatohepatitis for years. Irinotecan goes through multiple-step drug metabolism after injection and one of its intermediates 7-ethyl-10-hydroxy-camptothecin (SN-38) is responsible for irinotecan side effect. However, it is unclear what is the disposition kinetics of SN-38 in the organs subjected to toxicity. No studies ever quantified the effect of each enzyme or transporter on SN-38 distribution. In current study, we established a new physiologically based pharmacokinetic (PBPK) model to predict the disposition kinetics of irinotecan. The PBPK model was calibrated with in-house mouse pharmacokinetic data and evaluated with external datasets from the literature. We separated the contribution of each parameters in irinotecan pharmacokinetics by calculating the normalized sensitivity coefficient (NSC). The model gave robust prediction of SN-38 distribution in GI tract, the site of injury. We identified that bile excretion and UDP-glucuronosyltransferases (UGT) played more important roles than fecal excretion and renal clearance in SN-38 pharmacokinetics. Our NSC showed that the impact of enzyme and transporter on irinotecan and SN-38 pharmacokinetics evolved when time continued. Additionally, we mapped out the effect of inflammation on irinotecan metabolic pathways with PBPK modelling. We discovered that inflammation significantly increased the blood and liver exposure of irinotecan and SN-38 in the mice receiving bacterial endotoxin. Inflammation suppressed UGT, microbial metabolism but increased fecal excretion. The present PBPK model can serve as an efficacious and versatile tool to quantitively assess the risk of irinotecan toxicity.


Subject(s)
Antineoplastic Agents, Phytogenic , Camptothecin , Animals , Antineoplastic Agents, Phytogenic/toxicity , Camptothecin/adverse effects , Diarrhea/chemically induced , Glucuronosyltransferase/metabolism , Inflammation/chemically induced , Inflammation/drug therapy , Irinotecan , Mice
6.
Toxicol Res (Camb) ; 11(1): 184-194, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35237423

ABSTRACT

Neratinib is a pan-HER tyrosine kinase inhibitor newly approved by FDA in 2017 to treat HER2-positive breast cancer, but the phase III trial of neratinib showed that 96% of the patients taking neratinib experienced diarrhea. So far very few mechanistic studies explore neratinib-induced gastrointestinal (GI) toxicity. Hereby, we performed toxicity studies in mice to characterize the potential mechanism underlying this adverse effect. C57BL/6 J mice were separated into three groups A, B, C. Group A received vehicle; group B was orally dosed with 100 mg/kg neratinib once daily for 18 days. Group C was dosed with 100 mg/kg neratinib for 12 days and switched to vehicle for 6 days. Intestine and liver were collected for further analysis. Human intestine-derived cells were treated with neratinib in vitro. Our results showed that 12 days treatment of neratinib caused persistent histological damage in mouse GI tract. Both gene expression and activity of Cyp3a11, the major enzyme metabolizing neratinib in mice was reduced in small intestine. The gene expression of proinflammatory cytokines increased throughout the GI tract. Such damages were not recovered after 6 days without neratinib treatment. In addition, in vitro data showed that neratinib was potent in killing human intestine-derived cell lines. Based on such findings, we hypothesized that neratinib downregulates intestinal CYP3A enzyme to cause excessive drug disposition, eventually leading to gut injury.

7.
Article in English | MEDLINE | ID: mdl-35077841

ABSTRACT

Depression is the second most common psychiatric disorder, affecting more than 340 million people of all ages worldwide. However, the mechanisms underlying the development of depression remain unclear, and existing antidepressants may cause clinical dependence and toxic side effects. Recently, emerging evidence from the fields of neuroscience, genetics, and genomics supports the modulatory role of long non-coding RNA (lncRNA) in depression. LncRNAs may mediate the pathogenesis of depression through multiple pathways, including regulating neurotransmitters and neurotrophic factors, affecting synaptic conduction, and regulating the ventriculo-olfactory neurogenic system. In addition, relying on genome-wide association study and molecular biological experiment, the possibility of lncRNA as a potential biomarker for the differential diagnosis of depression and other mental illnesses, including schizophrenia and anxiety disorders, is gradually being revealed. Thus, it is important to explore whether lncRNAs are potential therapeutic targets and diagnostic biomarkers for depression. Here, we summarize the genesis and function of lncRNAs and discuss the aberrant expression and functional roles of lncRNAs in the development, diagnosis, and therapy of depression, as well as the deficiencies and limitations of these studies. Moreover, we established a lncRNA-miRNA-mRNA-pathway-drug network of depression through bioinformatics analysis methods to deepen our understanding of the relationship between lncRNA and depression, promoting the clinical application of epigenetic research.


Subject(s)
Depression , RNA, Long Noncoding/genetics , Computational Biology , Depression/genetics , Depression/physiopathology , Epigenomics , Gene Expression Profiling , Gene Regulatory Networks , Genome-Wide Association Study , Humans , MicroRNAs/genetics , RNA, Messenger/genetics
8.
Food Funct ; 12(24): 12550-12564, 2021 Dec 13.
Article in English | MEDLINE | ID: mdl-34812830

ABSTRACT

The gut microbiome is known to be involved in depression development. Thus, phytochemicals changing gut microbiota may alleviate depression-like behaviors. Coniferyl ferulate (CF) is a long studied natural product and known to alleviate psychiatric disorders. However, its mechanism of action remains unclear. In this experimental study, oral administration of 50 mg kg-1 CF once daily attenuated weight loss and depression-like and anxiety-like behaviors induced by chronic unpredicted mild stress (CUMS) in mice. Four weeks of CF administration significantly ameliorated colonic inflammation, lowered the levels of IL-6, IL-1ß, and TNF-α, and restructured the gut microbiome, and microbial metabolism. Intestinal microbiota can impact the development and function of the brain via the microbiota-gut-brain axis. Therefore, oral administration of CF is a promising nutritional strategy to treat CUMS-induced depression via the regulation of microbiota and microbial metabolism.


Subject(s)
Behavior, Animal/drug effects , Coumaric Acids/pharmacology , Depression/drug therapy , Depression/metabolism , Gastrointestinal Microbiome/drug effects , Administration, Oral , Animals , Coumaric Acids/administration & dosage , Coumaric Acids/metabolism , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL
9.
Toxicol Res (Camb) ; 10(3): 476-486, 2021 May.
Article in English | MEDLINE | ID: mdl-34141161

ABSTRACT

The epidermal growth factor receptor (EGFR) family is a class of receptor tyrosine kinase playing a central role in carcinogenesis and cancer progression. The members of this family, particularly EGFR and human epidermal growth factor receptor 2 (HER2), are the most extensively studied drug targets for malignancy. Today, numerous tyrosine kinase inhibitors targeting EGFR family have been developed to combat non-small-cell lung cancer and breast cancer. However, severe gastrointestinal (GI) toxicity leading to dose reduction and treatment discontinuation hampers the therapeutic outcome of EGFR inhibitors. Diarrhea is one of the most frequent GI side effects, especially when it comes to second-generation EGFR inhibitors. Enterocytes apoptosis and increased inflammation accompany with many oral EGFR inhibitors. Loperamide and budesonide are the first-line treatment to manage such adverse effects. However, current prophylaxis and management are all empirical interventions to relieve the symptom. They do not specifically target the toxicological mechanism of EGFR inhibitors. Hereby, those anti-diarrhea agents do not work well when used in cancer patients experiencing EGFR inhibitor-induced diarrhea. On the other hand, the toxicological mechanism of EGFR inhibitor-induced diarrhea is poorly understood. Thus, determining the mechanism behind such diarrhea is urgently in need for developing genuinely effective anti-diarrhea agents. This review aims to call attention to EGFR inhibitor-induced diarrhea, a highly occurring and devastating cancer drug toxicity.

10.
Front Pharmacol ; 11: 588508, 2020.
Article in English | MEDLINE | ID: mdl-33178026

ABSTRACT

Starting from December 2019, novel coronavirus disease 2019 (COVID-19) pandemic has caused tremendous economic loss and unprecedented health crisis across the globe. While the development of cure is at full speed, less attention and fewer effort have been spent on the prevention of this rapidly spreading respiratory infectious disease. Although so far, several vaccine candidates have advanced into clinical trials, limited data have been released regarding the vaccine efficacy and safety in human, not mention the long-term effectiveness of those vaccines remain as open question yet. Natural products and herbal medicines have been historically used for acute respiratory infection and generally show acceptable toxicity. The favorable stability for oral formulation and ease of scaling up manufacture make it ideal candidate for prophylactic. Hereby, we summarized the most recent advance in SARS-CoV-2 prevention including vaccine development as well as experimental prophylactics. Mainly, we reviewed the natural products showing inhibitory effect on human coronavirus, and discussed the herbal medicines lately used for COVID-19, especially focused on the herbal products already approved by regulatory agency with identifiable patent number. We demonstrated that to fill in the response gap between appropriate treatment and commercially available vaccine, repurposing natural products and herbal medicines as prophylactic will be a vigorous approach to stop or at least slow down SARS-CoV-2 transmission. In the interest of public health, this will lend health officials better control on the current pandemic.

11.
J Agric Food Chem ; 68(52): 15490-15500, 2020 Dec 30.
Article in English | MEDLINE | ID: mdl-33170671

ABSTRACT

As one of the empirical models of the chronic central inflammatory response, a spinal cord injury (SCI) deteriorates the neuronal survival and results in irreversible motor and sensory dysfunction below the injury area. Our previous studies have reported that maize bran feruloylated oligosaccharides (FOs) exert significant anti-inflammatory activities both in diabetes and colitis. However, no direct evidence of FOs alleviating central nervous inflammation was stated. This study aimed to investigate the therapeutic effect of FOs on SCI and its potential mechanism. Our results indicated that 4 weeks of FO administration effectively mitigated the inflammatory response via decreasing the number of microglia (labelled with Iba1), result in the expression of IL-1α, IL-2, IL-6, IL-18 and TNF-α downregulating, but the level of IL-10 and BDNF increases in the injured spinal cord. Moreover, FOs enhanced neuronal survival, ameliorated the scar cavities, and improved behaviors, including Basso mouse scale (BMS) scores and the gait of mice after SCI. Together, these results demonstrated that administration of FOs showed superior functional recovery effects in a SCI model. Also, FOs may modulate inflammatory activities by regulating the expression of proinflammatory factors, decreasing the production of inflammatory cells, and promoting functional recovery through the MAPK pathway following SCI.


Subject(s)
Oligosaccharides/administration & dosage , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/immunology , Animals , Coumaric Acids/metabolism , Cytokines/genetics , Cytokines/immunology , Female , Humans , Mice , Motor Activity , Oligosaccharides/chemistry , Recovery of Function , Spinal Cord/drug effects , Spinal Cord/immunology , Spinal Cord/physiopathology , Spinal Cord Injuries/genetics , Spinal Cord Injuries/physiopathology
12.
Food Res Int ; 137: 109410, 2020 11.
Article in English | MEDLINE | ID: mdl-33233097

ABSTRACT

Gut microbiome has been proven to be involved in the development of type 2 diabetes (T2D). Additionally, increasing evidence showed that the composition of gut microbiome is highly associated with the outcome of T2D therapy. Previously we demonstrated that feruloylated oligosaccharides (FOs) and ferulic acid (FA) alleviated diabetic syndrome in rats, but the detailed mechanism has not been explored yet. In this study we strived to characterize how FOs and FA altered the gut microbiome and related metabolome in diabetic rats by using high-throughput sequencing of 16S rRNA and gas chromatography (GC). Our results showed that FOs reduced the abundance of Lactobacillus, Ruminococcus, Oscillibacter, and Desulfovibrio, but increased the abundance of Akkermansia, Phascolarctobacterium and Turicibacter. The structure of gut microbiome in FOs treated rats was similar with healthy rats rather than diabetic rats. Likewise, FA decreased the portion of Lactobacillus, Ruminococcus, but promoted the growth of Bacteroides, Blautia, Faecalibacterium, Parabacteroides and Phascolarctobacterium. Additionally, the short-chain fatty acids (SCFAs) and branched-chain fatty acids (BCFAs), the main bacterial lipid metabolites in gut mediating host glucose metabolism, was dramatically elevated along with FOs and FA treatment. Our findings indicated that FOs and FA attenuated diabetic syndrome in rats most likely by modulating the composition and metabolism of gut microbiome. The study gives new insight into the mechanism underlying the anti-diabetes effect of functional foods as well as facilitates the development of dietary supplements for diabetic patients.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Gastrointestinal Microbiome , Animals , Coumaric Acids , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Humans , Oligosaccharides/pharmacology , RNA, Ribosomal, 16S , Rats
13.
Expert Opin Drug Metab Toxicol ; 16(11): 1109-1124, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32841068

ABSTRACT

INTRODUCTION: Toxicity of chemotherapy drugs is the leading cause of poor therapeutic outcome in many cancer patients. Gastrointestinal (GI) toxicity and hepatotoxicity are among the most common side effects of current chemotherapies. Emerging studies indicate that many chemotherapy-induced toxicities are driven by drug metabolism, but very few reviews summarize the role of drug metabolism in chemotherapy-induced GI toxicity and hepatotoxicity. In this review, we highlighted the importance of drug metabolizing enzymes (DMEs) in chemotherapy toxicity. AREAS COVERED: Our review demonstrated that altered activity of DMEs play important role in chemotherapy-induced GI toxicity and hepatotoxicity. Besides direct changes in catalytic activities, the transcription of DMEs is also affected by inflammation, cell-signaling pathways, and/or by drugs in cancer patients due to the disease etiology. EXPERT OPINION: More studies should focus on how DMEs are altered during chemotherapy treatment, and how such changes affect the metabolism of chemotherapy drug itself. This mutual interaction between chemotherapies and DMEs can lead to excessive exposure of parent drug or toxic metabolites which ultimately cause GI adverse effect.


Subject(s)
Antineoplastic Agents/adverse effects , Chemical and Drug Induced Liver Injury/etiology , Gastrointestinal Diseases/chemically induced , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/metabolism , Chemical and Drug Induced Liver Injury/physiopathology , Gastrointestinal Diseases/physiopathology , Humans , Neoplasms/drug therapy , Signal Transduction/drug effects
14.
Int J Mol Sci ; 20(22)2019 Nov 16.
Article in English | MEDLINE | ID: mdl-31744051

ABSTRACT

Colorectal cancer (CRC) is the third most common cancer and has a high metastasis and reoccurrence rate. Long noncoding RNAs (lncRNAs) play an important role in CRC growth and metastasis. Recent studies revealed that lncRNAs participate in CRC progression by coordinating with microRNAs (miRNAs) and protein-coding mRNAs. LncRNAs function as competitive endogenous RNAs (ceRNAs) by competitively occupying the shared binding sequences of miRNAs, thus sequestering the miRNAs and changing the expression of their downstream target genes. Such ceRNA networks formed by lncRNA/miRNA/mRNA interactions have been found in a broad spectrum of biological processes in CRC, including liver metastasis, epithelial to mesenchymal transition (EMT), inflammation formation, and chemo-/radioresistance. In this review, we summarize typical paradigms of lncRNA-associated ceRNA networks, which are involved in the underlying molecular mechanisms of CRC initiation and progression. We comprehensively discuss the competitive crosstalk among RNA transcripts and the novel targets for CRC prognosis and therapy.


Subject(s)
Biomarkers, Tumor/genetics , Colorectal Neoplasms/diagnosis , RNA, Long Noncoding/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Regulatory Networks , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/secondary , MicroRNAs/metabolism , RNA, Long Noncoding/genetics , RNA, Messenger/metabolism
15.
Food Res Int ; 122: 443-449, 2019 08.
Article in English | MEDLINE | ID: mdl-31229098

ABSTRACT

Various Maillard reaction systems were established to investigate the effect of maize bran feruloylated oligosaccharides (FOs) on the formation of both endogenous and exogenous advanced glycation end-products (AGEs) under different pH values. The formation of AGEs and four kinds of dicarbonyl compounds (glyoxal, methylglyoxal, 3-deoxyglucosulose, 2,3-butanedione) were determined by fluorescence spectrophotometry and HPLC, respectively. Results showed that maize bran FOs effectively inhibited the production of fluorescent AGEs. Moreover, increase in pH, temperature and concentration of FOs in reaction systems elevated the inhibition effects of feruloylated oligosaccharides, fluorescent AGEs and dicarbonyl compounds. Mainly because of the ester bond of FOs at high pH and/or temperature systems are readily to be hydrolyzed, resulting in the release of ferulic acid.


Subject(s)
Amino Acids/metabolism , Dietary Fiber/analysis , Glucose/metabolism , Glycation End Products, Advanced/metabolism , Oligosaccharides/analysis , Serum Albumin, Bovine/metabolism , Zea mays/chemistry , Coumaric Acids/metabolism , Diacetyl/metabolism , Glycation End Products, Advanced/antagonists & inhibitors , Glyoxal/metabolism , Hydrogen-Ion Concentration , Hydrolysis , Oligosaccharides/pharmacology , Pyruvaldehyde/metabolism , Temperature
16.
Ann Pharmacother ; 53(6): 612-620, 2019 06.
Article in English | MEDLINE | ID: mdl-30607980

ABSTRACT

OBJECTIVE: To review the chemistry, pharmacology, pharmacokinetics, safety, and efficacy of neratinib in human epidermal growth factor receptor (HER2)+ breast cancer (BC). DATA SOURCES: A PubMed search was performed using the term neratinib between September 12, 2018, and November 21, 2018. References of published articles and reviews were also assessed for additional information. STUDY SELECTION AND DATA EXTRACTION: English-language preclinical and clinical studies on the chemistry, pharmacology, pharmacokinetics, safety, and efficacy of neratinib were evaluated. DATA SYNTHESIS: Neratinib, an irreversible inhibitor of HER1, HER2, and HER4, is Food and Drug Administration approved for the extended adjuvant treatment of stage I-III HER2+ BC to follow trastuzumab-based therapy. A phase III study has demonstrated statistically significant improvement in 5-year disease-free survival rate (90.2 vs 87.7; hazard ratio = 0.73, 95% CI = 0.57-0.92, P = 0.0083). Its most common adverse effect is diarrhea, observed in more than 90% of patients. The incidence of grade 3/4 diarrhea (~40%) is reduced by half with loperamide prophylaxis, which is recommended for the first 8 weeks of neratinib therapy. Other common adverse reactions are nausea and fatigue. The patients need to be monitored for liver function tests and drug interactions with acid-reducing agents, CYP3A4 inhibitors/inducers, and P-glycoprotein substrates with narrow therapeutic window. Relevance to Patient Care and Clinical Practice: American Society of Clinical Oncology and National Comprehensive Cancer Network clinical guidelines suggest the use of neratinib for extended adjuvant therapy following 1-year trastuzumab in stage I to III HER2+ BC. Diarrhea remains a clinically significant but manageable adverse event. CONCLUSION: Neratinib significantly improves treatment outcomes and has manageable toxicity in stage I to III HER2+ BC patients.


Subject(s)
Breast Neoplasms/drug therapy , Quinolines/therapeutic use , Adult , Female , Humans , Middle Aged , Quinolines/pharmacology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...