1
|
Mao W, Zhou T, Zhang F, Qian M, Xie J, Li Z, Shu Y, Li Y, Xu H. Pan-cancer single-cell landscape of drug-metabolizing enzyme genes. Pharmacogenet Genomics 2024; 34:217-225. [PMID: 38814173 DOI: 10.1097/fpc.0000000000000538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
OBJECTIVE Varied expression of drug-metabolizing enzymes (DME) genes dictates the intensity and duration of drug response in cancer treatment. This study aimed to investigate the transcriptional profile of DMEs in tumor microenvironment (TME) at single-cell level and their impact on individual responses to anticancer therapy. METHODS Over 1.3 million cells from 481 normal/tumor samples across 9 solid cancer types were integrated to profile changes in the expression of DME genes. A ridge regression model based on the PRISM database was constructed to predict the influence of DME gene expression on drug sensitivity. RESULTS Distinct expression patterns of DME genes were revealed at single-cell resolution across different cancer types. Several DME genes were highly enriched in epithelial cells (e.g. GPX2, TST and CYP3A5 ) or different TME components (e.g. CYP4F3 in monocytes). Particularly, GPX2 and TST were differentially expressed in epithelial cells from tumor samples compared to those from normal samples. Utilizing the PRISM database, we found that elevated expression of GPX2, CYP3A5 and reduced expression of TST was linked to enhanced sensitivity of particular chemo-drugs (e.g. gemcitabine, daunorubicin, dasatinib, vincristine, paclitaxel and oxaliplatin). CONCLUSION Our findings underscore the varied expression pattern of DME genes in cancer cells and TME components, highlighting their potential as biomarkers for selecting appropriate chemotherapy agents.
Collapse
Affiliation(s)
- Wei Mao
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan
| | - Tao Zhou
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan
| | - Feng Zhang
- Center for Precision Medicine, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang
| | - Maoxiang Qian
- Institute of Pediatrics and Department of Hematology and Oncology, National Children's Medical Center, Children's Hospital of Fudan University, Shanghai
| | - Jianqiang Xie
- Department of Medicine and Surgery, Sichan Second Veterans Hospital
| | - Zhengyan Li
- Department of Radiology, West China Hospital, Sichuan University
| | - Yang Shu
- Gastric Cancer Center, West China Hospital, Sichuan University
| | - Yuan Li
- Institute of Digestive Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Heng Xu
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan
| |
Collapse
|
2
|
Zhou L, Wang X, Xia J, Zhang L, Xue L, Jia Q, Fu Z, Sun Z. Pharmacokinetic-pharmacodynamic modeling of the active components of Shenkang injection in rats with chronic renal failure and its protective effect on damaged renal cells. Biopharm Drug Dispos 2023; 44:406-419. [PMID: 37679901 DOI: 10.1002/bdd.2377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 07/14/2023] [Accepted: 08/20/2023] [Indexed: 09/09/2023]
Abstract
The study aimed to explore the pharmacokinetic and pharmacodynamic alterations of the active components of Shenkang injection (i.e. hydroxy saffron yellow pigment A [HSYA], tanshinol, rheum emodin, and astragaloside IV) in rats with chronic renal failure (CRF), and establish a pharmacokinetic-pharmacodynamic model (PK-PD model) in order to provide a scientific and theoretical basis for the rational clinical use of Shenkang injection. Sprague-Dawley (SD) rats were randomly divided into a normal group, model group, and Shenkang injection group. A rat model of CRF was induced by adenine gavage and then followed by drug administration via tail vein injection. Orbital blood was collected at different timepoints and the blood concentrations of the four active components were measured by UHPLC-Q-Orbitrap HRMS. Serum levels of creatinine (Scr), urea nitrogen (BUN), and uric acid (UA) were determined using an automatic biochemical analyzer. A PK-PD model was established, and DAS 3.2.6 software was used for model fitting as well as statistical analysis. TGF-β1 was utilized to induce normal rat kidney cells to construct a renal fibrosis model to investigate the protective effect of the pharmacological components on renal fibrosis. The pharmacokinetic analysis of hydroxy saffron yellow pigment A, tanshinol, rheum emodin, and astragaloside IV based on UHPLC-Q-Orbitrap HRMS was stable. The linear regression equations for the four active components were as follows: Y = 0.031X + 0.0091 (R2 = 0.9986) for hydroxy saffron yellow pigment A, Y = 0.0389X + 0.164 (R2 = 0.9979) for tanshinol, Y = 0.0257X + 0.0146 (R2 = 0.9973) for rheum emodin, and Y = 0.0763X + 0.0139 (R2 = 0.9993) for astragaloside IV, which indicated good linear relationships. The methodological investigation was stable, with the interday and intraday precision RSD <10%. Meanwhile, the recoveries ranged between 90% and 120%, in accordance with the requirements for in vivo analysis of drugs. Compared with the model group, the levels of Scr, BUN, and UA were significantly decreased after 20 min in the Shenkang injection group (p < 0.01). The PK-PD model showed that the four active components in the Shenkang injection group could fit well with the three effect measures (i.e. Scr, BUN, and UA), with the measured values similar to the predicted values. The cell model of renal fibrosis showed that the connective tissue growth factor and FN1 protein expression levels were significantly lower in the Shenkang injection group than those in the model group, and the cell fibrosis was improved. The established method for in vivo analysis of Shenkang injection was highly specific, with good separation of the components and simple operation. The total statistical moment could well integrate the pharmacokinetic parameters of the four active components. After treatment with Shenkang injection, all indexes in the administered group improved and showed significant inhibition of renal cell fibrosis in vitro. This study could provide scientific reference ideas for the clinical rational use of traditional Chinese medicine.
Collapse
Affiliation(s)
- Lin Zhou
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- School of Minerals Processing and Bioengineering, Central South University, Changsha, China
| | - Xiaohui Wang
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jinlan Xia
- School of Minerals Processing and Bioengineering, Central South University, Changsha, China
| | - Liyuan Zhang
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lianping Xue
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qingquan Jia
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhihui Fu
- Department of Chinese Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhi Sun
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
3
|
Perlik V, Kulasekaran A, Coutinho G, Votava M, Cardot JM. Relationship between Pharmacokinetic Profile and Clinical Efficacy Data of Three Different Forms of Locally Applied Flurbiprofen in the Mouth/Throat. Pharmaceutics 2023; 15:1863. [PMID: 37514048 PMCID: PMC10385846 DOI: 10.3390/pharmaceutics15071863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/16/2023] [Accepted: 06/16/2023] [Indexed: 07/30/2023] Open
Abstract
This study aimed to link pharmacokinetic (PK) data from different flurbiprofen preparations for the treatment of sore throat with published data to elucidate whether early efficacy is due to the local action of flurbiprofen or a systemic effect after absorption of the swallowed drug. Three comparative bioavailability studies conducted in healthy subjects provided data from flurbiprofen 8.75 mg formulations, including spray solution, spray gel, lozenges, and granules. A parallel interstudy comparison was made of PK parameters, including partial AUCs (pAUCs), using an ANOVA model with the calculation of 90% confidence intervals (CI) for the differences between least squares (LS) means for each of the test groups versus the respective reference groups. All three studies showed bioequivalence for the respective product comparisons. The interstudy comparison showed a slower rate of absorption for granules compared to spray solution (reference) based on Tmax, Cmax, and pAUCs for 1 h and 2 h. When AUC0.25h and AUC0.5h were considered, slower rates of absorption were also seen for lozenges and spray gel. The differences correlated with the reported time of onset of action, which is faster for the spray solution (20 min) compared to lozenges (26 min) and granules (30 min). These pAUCs provide useful data that allow for the discrimination between formulations. Moreover, the pAUC values represent <5% of the total AUC, suggesting that the early onset of pain relief is a response to immediate local absorption at the site of action rather than a systemic effect.
Collapse
Affiliation(s)
- Vit Perlik
- Institute of Pharmacology, First Faculty of Medicine, Charles University, Albertov 4, 12800 Prague, Czech Republic
| | | | - Graça Coutinho
- Reckitt Healthcare Limited, 103-105 Bath Road, Slough, Berkshire SL1 3UH, UK
| | - Martin Votava
- Cadore INV Scientific Consultant, Akademicka 1, 10800 Prague, Czech Republic
| | | |
Collapse
|
4
|
Liu J, Li X, Bai H, Yang X, Mu J, Yan R, Wang S. Traditional uses, phytochemistry, pharmacology, and pharmacokinetics of the root bark of Paeonia x suffruticosa andrews: A comprehensive review. JOURNAL OF ETHNOPHARMACOLOGY 2023; 308:116279. [PMID: 36822345 DOI: 10.1016/j.jep.2023.116279] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 02/11/2023] [Accepted: 02/12/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Moutan Cortex (MC), commonly known as "Mu dan pi", refers to the dried root bark of Paeonia x suffruticosa Andrews and is broadly used as a traditional herbal medication in China, Japan, and Korea. For thousands of years, it has been utilized to treat female genital, extravasated blood, cardiovascular, and stagnant blood disorders. AIM OF THE REVIEW The purpose of this review article was to summarize information on the traditional uses, phytochemistry, pharmacology and pharmacokinetics of MC, as well as to outline the further research directions for the development of new drugs and the associations between traditional uses and pharmacological effects. MATERIALS AND METHODS The information involved in the study was gathered from a variety of electronic resources, including PubMed, Web of Science, ScienceDirect, SciFinder, China Knowledge Resource Integrated Database, and Google Scholar. The date was from 1992 to 2022. RESULTS Approximately 163 chemical compounds have been extracted and identified from MC, including monoterpenes, monoterpene glycosides, triterpenes, phenolics, flavonoids, volatile oils, alkaloids, and others. In these categories, the monoterpene glycosides and phenols being the most common. A wide variety of pharmacological effects have been described for MC crude extracts and active molecules, such as antioxidant, anti-inflammatory, antibacterial and antiviral, antitumor, antidiabetic, organ protection, and neuroprotective activities, as well as treating cardiovascular diseases. Pharmacokinetics has been also used in the study of MC, including its crude extracts or chemical constituents, in order to explore the therapeutic mechanism, direct clinically appropriate application and provide new ideas for the exploitation of innovative medicines. CONCLUSION Modern pharmacological research has demonstrated that MC, as a significant therapeutic resource, has the ability to heal a wide range of diseases, particularly female genital and cardiovascular problems. These researches propose therapeutic ideas for the development of novel MC medicines. Furthermore, preclinical and clinical study have verified several observed pharmacological properties related with the traditional usages of MC.
Collapse
Affiliation(s)
- Jincai Liu
- Northwest University Faculty of Life and Health Science, Northwest University, 229 Taibai Road, Xi'an, Shaanxi, 710069, China
| | - Xiang Li
- Northwest University Faculty of Life and Health Science, Northwest University, 229 Taibai Road, Xi'an, Shaanxi, 710069, China
| | - Huixin Bai
- Northwest University Faculty of Life and Health Science, Northwest University, 229 Taibai Road, Xi'an, Shaanxi, 710069, China
| | - Xu Yang
- Northwest University Faculty of Life and Health Science, Northwest University, 229 Taibai Road, Xi'an, Shaanxi, 710069, China
| | - Jun Mu
- Northwest University Faculty of Life and Health Science, Northwest University, 229 Taibai Road, Xi'an, Shaanxi, 710069, China
| | - Ruonan Yan
- Northwest University Faculty of Life and Health Science, Northwest University, 229 Taibai Road, Xi'an, Shaanxi, 710069, China
| | - Siwang Wang
- Northwest University Faculty of Life and Health Science, Northwest University, 229 Taibai Road, Xi'an, Shaanxi, 710069, China.
| |
Collapse
|
5
|
Giaretta A, Petrucci G, Rocca B, Toffolo GM. Physiologically based modelling of the antiplatelet effect of aspirin: A tool to characterize drug responsiveness and inform precision dosing. PLoS One 2022; 17:e0268905. [PMID: 35976924 PMCID: PMC9385056 DOI: 10.1371/journal.pone.0268905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 05/11/2022] [Indexed: 11/18/2022] Open
Abstract
A computational approach involving mathematical modeling and in silico experiments was used to characterize the determinants of extent and duration of platelet cyclooxygenase (COX)-1 inhibition by aspirin and design precision dosing in patients with accelerated platelet turnover or reduced drug bioavailability. To this purpose, a recently developed physiologically-based pharmacokinetics (PK) and pharmacodynamics (PD) model of low-dose aspirin in regenerating platelets and megakaryocytes, was used to predict the main features and determinants of platelet COX-1 inhibition. The response to different aspirin regimens in healthy subjects and in pathological conditions associated with alterations in aspirin PK (i.e., severely obese subjects) or PD (i.e., essential thrombocytemya patients), were simulated. A model sensitivity analysis was performed to identify the main processes influencing COX-1 dynamics. In silico experiments and sensitivity analyses indicated a major role for megakaryocytes and platelet turnover in determining the extent and duration of COX-1 inhibition by once-daily, low-dose aspirin. They also showed the superiority of reducing the dosing interval vs increasing the once-daily dose in conditions of increased platelet turnover, while suggested specific dose adjustments in conditions of possible reduction in drug bioavailability. In conclusion, the consistency of our model-based findings with experimental data from studies in healthy subjects and patients with essential thrombocythemia supports the potential of our approach for describing the determinants of platelet inhibition by aspirin and informing precision dosing which may guide personalized antithrombotic therapy in different patient populations, especially in those under-represented in clinical trials or in those associated with poor feasibility.
Collapse
Affiliation(s)
- Alberto Giaretta
- Department of Information Engineering, University of Padova, Padova, Italy
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
- * E-mail: ,
| | - Giovanna Petrucci
- Department of Pharmacology, Catholic University School of Medicine, Rome, Italy
| | - Bianca Rocca
- Department of Pharmacology, Catholic University School of Medicine, Rome, Italy
| | | |
Collapse
|
6
|
Li Z, Li Z, Wang B, Liu J. Influence of release rate, dose and co-administration on pharmacokinetics, pharmacodynamics and PK-PD relationship of tanshinone IIA and tanshinol. Eur J Pharm Sci 2022; 168:106042. [PMID: 34656775 DOI: 10.1016/j.ejps.2021.106042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 11/28/2022]
Abstract
The present study aims to investigate the influence of release rate, dose and co-administration on pharmacokinetics (PK) and pharmacodynamics (PD) of tanshinone IIA (TA) and tanshinol (TS), and reveal the changes in their PK-PD relationships. Sustained and immediate release pellets of TS and TA were prepared respectively, and oral administrated to angina model rabbits according to the experimental design. The administration dose of TS was 50, 35 or 20 mg/kg and that of TA was 30 mg/kg. Then, plasma concentrations of TS and TA were measured to evaluate the pharmacokinetics. Pharmacodynamic biomarkers including cardiac troponin (cTn-I), creatine kinase (CK-MB), superoxide dismutase (SOD) and nitric oxide (NO) were measured to evaluated the effects of cardioprotection, amelioration of oxidative stress and vasorelaxation of TS and TA. Parameters such as maximum plasma concentration (Cmax), maximum effect (Emax), time to Cmax or Emax (TCmax or TEmax), areas under the plasma concentration or effect curves (AUC0-∞ or AUEC) and pharmacodynamic efficiency (EFF) were calculated based on non-compartmental analysis. Beside, PK-PD relationship/hysteresis was evaluated. The TEmax was less sensitive than TCmax to changes in release rate. The Emax, AUEC and EFF showed increasing trend as the decrease of release rate even that the AUC0-∞ showed no significant difference. In addition, slow drug release decreased the magnitude of hysteresis of TS and TA. The sensitivities of Emax and AUEC of four biomarkers to changes in dose were varied and relatively lower than those of Cmax and AUC0-∞. The EFF decreased and the magnitude of hysteresis increased for high dose. The Cmax and AUC0-∞ of TS and TA showed little difference after co-administration. The Emax and AUEC of four biomarkers increased for immediate release pellets (P < 0.05 or P < 0.01) and generally decreased for sustained release pellets (P < 0.05 or P < 0.01) after co-administration. In addition, the magnitudes of hysteresis of four biomarkers decreased for immediate release pellets and generally increased for sustained release pellets after co-administration. In summary, the dissociated and unstable PK-PD relationship should be considered during optimization of dosage forms and regimens to make sure the rationality, safety and efficacy. These findings could also provide some valuable information for the development and clinical therapy of other drugs.
Collapse
Affiliation(s)
- Zhenghua Li
- Department of Pharmaceutics, China Pharmaceutical University, No.24 Tongjiaxiang, Nanjing 210009, China
| | - Ziyi Li
- Department of Pharmaceutics, China Pharmaceutical University, No.24 Tongjiaxiang, Nanjing 210009, China
| | - Bingwei Wang
- Department of Pharmaceutics, China Pharmaceutical University, No.24 Tongjiaxiang, Nanjing 210009, China
| | - Jianping Liu
- Department of Pharmaceutics, China Pharmaceutical University, No.24 Tongjiaxiang, Nanjing 210009, China.
| |
Collapse
|
7
|
Fu J, Zhang H, Liu S, Wu J, Zhang Y, Gao Y, Song F, Qin Y, Hu X, Liu Z. An integrated strategy using LC-MS/MS combined with in vivo microdialysis for the simultaneous determination of lignans of Schisandra chinensis (Turcz.) Baill. Fructus and endogenous neurotransmitters: application in pharmacokinetic and pharmacodynamic studies. Food Funct 2021; 12:8932-8945. [PMID: 34606559 DOI: 10.1039/d1fo01682b] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Schisandra chinensis (Turcz.) Baill Fructus (SCF) is the ripe fruit of Schisandra chinensis (Turcz.) Baill, and is often used as a neuroprotective drink. Modern pharmacological studies have shown that lignans are the main bioactive components responsible for neuroprotection and have potential in the treatment of Alzheimer's disease (AD). However, the mechanism of action of SCF in the treatment of AD from the pharmacokinetics-pharmacodynamics (PK-PD) perspective remains not well established. The purpose of this study is to investigate and compare the pharmacokinetic differences of lignans in normal and AD rats, as well as to investigate their effects on neurotransmitters and their role in the treatment of AD. To achieve this goal, an integrated strategy using LC-MS/MS combined with in vivo microdialysis for the simultaneous determination of lignans of SCF and endogenous neurotransmitters has been developed and validated. The results show that the pharmacokinetic behaviors of ten lignans in the AD group were significantly different from those in the normal group. The AD group had better absorption and slower elimination than the normal group. In addition, the pharmacodynamic results of the Morris water maze (MWM) test, biochemical tests, histopathological examination, as well as immunohistochemistry analysis showed that lignans could improve the learning and memory of AD rats. The oral administration of SCF could restore the levels of the neurotransmitter parameters; seven neurotransmitters showed clockwise or counterclockwise changes with the four lignans in the hippocampal region. Taken together, the PK and PD studies based on in vivo microdialysis sampling might offer novel insights into the mechanisms of action of SCF against AD.
Collapse
Affiliation(s)
- Jun Fu
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China. .,National Center of Mass Spectrometry in Changchun & Jilin Provincial Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Hongxu Zhang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China. .,National Center of Mass Spectrometry in Changchun & Jilin Provincial Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Shu Liu
- National Center of Mass Spectrometry in Changchun & Jilin Provincial Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Jiajie Wu
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China. .,National Center of Mass Spectrometry in Changchun & Jilin Provincial Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Yuying Zhang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China. .,National Center of Mass Spectrometry in Changchun & Jilin Provincial Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Yang Gao
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China. .,National Center of Mass Spectrometry in Changchun & Jilin Provincial Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Fengrui Song
- National Center of Mass Spectrometry in Changchun & Jilin Provincial Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Yuhua Qin
- Hainan Tropical Marine University, Sanya, 572022, China
| | - Xiuli Hu
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China.
| | - Zhongying Liu
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China.
| |
Collapse
|
8
|
Villasana K, Quintero W, Montero Y, Pino C, Uzcategui O, Torres G, Prada M, Pozo L, Bauta W, Jimenez W. Effect of an ionic antineoplastic agent Cytoreg on blood chemistry in a Wistar rat model. Med Gas Res 2021; 12:18-23. [PMID: 34472498 PMCID: PMC8447950 DOI: 10.4103/2045-9912.324592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Cytoreg is an ionic therapeutic agent comprising a mixture of hydrochloric, sulfuric, phosphoric, hydrofluoric, oxalic, and citric acids. In diluted form, it has demonstrated efficacy against human cancers in vitro and in vivo. Although Cytoreg is well tolerated in mice, rats, rabbits, and dogs by oral and intravenous administration, its mechanism of action is not documented. The acidic nature of Cytoreg could potentially disrupt the pH and levels of ions and dissolved gases in the blood. Here, we report the effects of the intravenous administration of Cytoreg on the arterial pH, oxygen and carbon dioxide pressures, and bicarbonate, sodium, potassium, and chloride concentrations. Our results demonstrate that Cytoreg does not disturb the normal blood pH, ion levels, or carbon dioxide content, but increases oxygen levels in rats. These data are consistent with the excellent tolerability of intravenous Cytoreg observed in rabbits, and dogs. The study was approved by the Bioethics Committee of the University of the Andes, Venezuela (CEBIOULA) (approval No. 125) on November 3, 2019.
Collapse
Affiliation(s)
- Katiusca Villasana
- Biochemistry Laboratories, Department of Pathology, University of the Andes, Mérida, Venezuela
| | - William Quintero
- Department of Biology, Faculty of Science, University of the Andes, Mérida, Venezuela
| | | | - Cristian Pino
- Department of Technical and Experimental Surgery, Faculty of Medicine, University of the Andes, Mérida, Venezuela
| | - Oscar Uzcategui
- Department of Technical and Experimental Surgery, Faculty of Medicine, University of the Andes, Mérida, Venezuela
| | - Geizon Torres
- Vitalis, Clinical Laboratory, Smart Health Laboratory, Merida, Venezuela
| | - Mariangel Prada
- Vitalis, Clinical Laboratory, Smart Health Laboratory, Merida, Venezuela
| | - Lewis Pozo
- Cytorex de Venezuela SA, Maracaibo, Venezuela
| | | | | |
Collapse
|
9
|
Ludbrook G, Li F, Sleigh J, Liang Y. Assessments of Onset and Duration of Drug Effects and Pharmacokinetics by Dose Level of HSK3486, a New Sedative-Hypnotic Agent, in Healthy Female/Male Subjects: A Phase I Multiarm Randomized Controlled Clinical Trial [Retracted]. Anesth Analg 2021:00000539-990000000-00051. [PMID: 33464758 DOI: 10.1213/ane.0000000000005343] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Guy Ludbrook
- From the PARC Clinical Research, Royal Adelaide Hospital, Adelaide, South Australia, Australia
- Department of Anaesthesia, University of Adelaide, Adelaide, South Australia, Australia
| | - Fangqiong Li
- Department of Research and Development, Haisco Pharmaceutical Group Co, Ltd, Chengdu, China
| | - Jamie Sleigh
- Department of Anaesthesia, Waikato Clinical Campus, University of Auckland, Hamilton, New Zealand
| | - Yong Liang
- Department of Research and Development, Haisco Pharmaceutical Group Co, Ltd, Chengdu, China
| |
Collapse
|
10
|
Loisios-Konstantinidis I, Dressman J. Physiologically Based Pharmacokinetic/Pharmacodynamic Modeling to Support Waivers of In Vivo Clinical Studies: Current Status, Challenges, and Opportunities. Mol Pharm 2020; 18:1-17. [PMID: 33320002 DOI: 10.1021/acs.molpharmaceut.0c00903] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Physiologically based pharmacokinetic/pharmacodynamic (PBPK/PD) modeling has been extensively applied to quantitatively translate in vitro data, predict the in vivo performance, and ultimately support waivers of in vivo clinical studies. In the area of biopharmaceutics and within the context of model-informed drug discovery and development (MID3), there is a rapidly growing interest in applying verified and validated mechanistic PBPK models to waive in vivo clinical studies. However, the regulatory acceptance of PBPK analyses for biopharmaceutics and oral drug absorption applications, which is also referred to variously as "PBPK absorption modeling" [Zhang et al. CPT: Pharmacometrics Syst. Pharmacol. 2017, 6, 492], "physiologically based absorption modeling", or "physiologically based biopharmaceutics modeling" (PBBM), remains rather low [Kesisoglou et al. J. Pharm. Sci. 2016, 105, 2723] [Heimbach et al. AAPS J. 2019, 21, 29]. Despite considerable progress in the understanding of gastrointestinal (GI) physiology, in vitro biopharmaceutic and in silico tools, PBPK models for oral absorption often suffer from an incomplete understanding of the physiology, overparameterization, and insufficient model validation and/or platform verification, all of which can represent limitations to their translatability and predictive performance. The complex interactions of drug substances and (bioenabling) formulations with the highly dynamic and heterogeneous environment of the GI tract in different age, ethnic, and genetic groups as well as disease states have not been yet fully elucidated, and they deserve further research. Along with advancements in the understanding of GI physiology and refinement of current or development of fully mechanistic in silico tools, we strongly believe that harmonization, interdisciplinary interaction, and enhancement of the translational link between in vitro, in silico, and in vivo will determine the future of PBBM. This Perspective provides an overview of the current status of PBBM, reflects on challenges and knowledge gaps, and discusses future opportunities around PBPK/PD models for oral absorption of small and large molecules to waive in vivo clinical studies.
Collapse
Affiliation(s)
| | - Jennifer Dressman
- Institute of Pharmaceutical Technology, Goethe University, Frankfurt am Main 60438, Germany.,Fraunhofer Institute of Translational Pharmacology and Medicine (ITMP), Carl-von-Noorden Platz 9, Frankfurt am Main 60438, Germany
| |
Collapse
|
11
|
Pan S, Zhou J, Zhou S, Huang Z, Meng J. Pharmacokinetic-pharmacodynamic modeling for Moutan Cortex/Moutan Cortex charcoal and the contributions of the chemical component using support vector regression with particle swarm optimization. RSC Adv 2020; 10:24454-24462. [PMID: 35516193 PMCID: PMC9055091 DOI: 10.1039/d0ra04111d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/27/2020] [Accepted: 06/18/2020] [Indexed: 11/21/2022] Open
Abstract
Moutan Cortex (MC) and Moutan Cortex charcoal (MCC) are two kinds of Chinese medicinal materials widely used in traditional Chinese medicine (TCM) with opposite drug efficacy. And the contributions of the chemical component to the drug efficacy are still not clear. In our study, a support vector regression (SVR) model with particle swarm optimization (PSO) has been developed for simultaneously characterizing the pharmacokinetics (PK) and pharmacodynamics (PD) of MC/MCC. Then the contributions of the chemical component to the drug efficacy of MC/MCC are calculated by the weight analysis of SVR. The experimental results show that the effective substances found by the PSO-SVR model in MC and MCC are consistent with TCM theory. And the PSO-SVR model is a better model for PK-PD compared with the back-propagation neural network (BPNN). In conclusion, the PSO-SVR is a valuable tool that linked PK and PD profiles of MC/MCC with multiple components and identified the contributions of multiple therapeutic materials to the drug efficacy.
Collapse
Affiliation(s)
- Sixing Pan
- College of Public Health, Guangdong Pharmaceutical University Guangzhou 510310 China +86 020 39352095
| | - Jianan Zhou
- College of Public Health, Guangdong Pharmaceutical University Guangzhou 510310 China +86 020 39352095
| | - Sujuan Zhou
- College of Medical Information Engineering, Guangdong Pharmaceutical University Guangzhou 510006 China +86 020 39352095
| | - Zhangpeng Huang
- College of Medical Information Engineering, Guangdong Pharmaceutical University Guangzhou 510006 China +86 020 39352095
- Medicinal Information & Real World Engineering Technology Center, Guangdong Pharmaceutical University Guangzhou 510006 China
| | - Jiang Meng
- College of Traditional Chinese Medicine, Guangdong Pharmaceutical University Guangzhou 510006 China +86 020 39352169
| |
Collapse
|
12
|
Maharao N, Antontsev V, Wright M, Varshney J. Entering the era of computationally driven drug development. Drug Metab Rev 2020; 52:283-298. [PMID: 32083960 DOI: 10.1080/03602532.2020.1726944] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Historically, failure rates in drug development are high; increased sophistication and investment throughout the process has shifted the reasons for attrition, but the overall success rates have remained stubbornly and consistently low. Only 8% of new entities entering clinical testing gain regulatory approval, indicating that significant obstacles still exist for efficient therapeutic development. The continued high failure rate can be partially attributed to the inability to link drug exposure with the magnitude of observed safety and efficacy-related pharmacodynamic (PD) responses; frequently, this is a result of nonclinical models exhibiting poor prediction of human outcomes across a wide range of disease conditions, resulting in faulty evaluation of drug toxicology and efficacy. However, the increasing quality and standardization of experimental methods in preclinical stages of testing has created valuable data sets within companies that can be leveraged to further improve the efficiency and accuracy of preclinical prediction for both pharmacokinetics (PK) and PD. Models of Quantitative structure-activity relationships (QSAR), physiologically based pharmacokinetics (PBPK), and PK/PD relationships have also improved efficiency. Founded on a core understanding of biochemistry and physiological interactions of xenobiotics, these in silico methods have the potential to increase the probability of compound success in clinical trials. Integration of traditional computational methods with machine-learning approaches and existing internal pharma databases stands to make a fundamental impact on the speed and accuracy of predictions during the process of drug development and approval.
Collapse
|
13
|
Loisios-Konstantinidis I, Cristofoletti R, Fotaki N, Turner DB, Dressman J. Establishing virtual bioequivalence and clinically relevant specifications using in vitro biorelevant dissolution testing and physiologically-based population pharmacokinetic modeling. case example: Naproxen. Eur J Pharm Sci 2020; 143:105170. [DOI: 10.1016/j.ejps.2019.105170] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 11/25/2019] [Accepted: 11/26/2019] [Indexed: 01/19/2023]
|