1
|
Peribañez-Dominguez S, Parra-Guillen Z, Troconiz IF. Development and Application of Physiologically-Based Pharmacokinetic Model to Predict Systemic and Organ Exposure of Colorectal Cancer Drugs. Pharmaceutics 2025; 17:57. [PMID: 39861705 PMCID: PMC11768185 DOI: 10.3390/pharmaceutics17010057] [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: 11/12/2024] [Revised: 12/23/2024] [Accepted: 12/31/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Colorectal cancer (CRC) holds the third and second position among cancers affecting men and women, respectively. Frequently, the first-line treatment for metastatic CRC consists of the intravenous administration of 5-fluorouracil and leucovorin in combination with oxaliplatin or irinotecan. Physiologically-based pharmacokinetic models (PBPK) aim to mechanistically incorporate body physiology and drug physicochemical attributes, enabling the description of both systemic and organ drug exposure based on the treatment specificities. This bottom-up approach represents an opportunity to personalize treatment and minimize the therapeutic risk/benefit ratio through the understanding of drug distribution within colorectal tissue. This project has the goal of characterizing the systemic and tissue exposure of four anti-cancer drugs in humans using a PBPK platform fed with data from the literature. METHODS A literature search was performed to collect clinical data on systemic concentration versus time profiles. Physicochemical features were obtained from the literature, as well as parameters associated with distribution, metabolism, and excretion. The PBPK models were built using PK-Sim®. RESULTS The data from 51 clinical studies were extracted and combined in one single dataset. The PBPK models successfully described the exposure vs. time profiles with respect to both, with both the typical tendency and dispersion shown by the data. The percentage of observations falling within the two-fold error bounds ranged between 94 and 100%. The colon/plasma AUCinf ratios were similar for 5-FU, oxaliplatin, and leucovorin, but it was significantly higher for irinotecan. CONCLUSIONS The PBPK models support tailored treatment approaches by linking in vitro studies to organ exposure. These models serve as the initial step towards incorporating a dedicated tumor compartment, which will further account for the variability in tumor microenvironment characteristics to improve therapeutic strategies.
Collapse
Affiliation(s)
- Sara Peribañez-Dominguez
- Department of Pharmaceutical Science, School of Pharmacy and Nutrition, University of Navarra, 31009 Pamplona, Spain; (Z.P.-G.); (I.F.T.)
- Navarra Institute for Health Research (IdiSNA), 31002 Pamplona, Spain
| | - Zinnia Parra-Guillen
- Department of Pharmaceutical Science, School of Pharmacy and Nutrition, University of Navarra, 31009 Pamplona, Spain; (Z.P.-G.); (I.F.T.)
- Navarra Institute for Health Research (IdiSNA), 31002 Pamplona, Spain
| | - Iñaki F. Troconiz
- Department of Pharmaceutical Science, School of Pharmacy and Nutrition, University of Navarra, 31009 Pamplona, Spain; (Z.P.-G.); (I.F.T.)
- Navarra Institute for Health Research (IdiSNA), 31002 Pamplona, Spain
- Navarra Institute of Data Science and Artificial Intelligence, DATAI, University of Navarra, 31009 Pamplona, Spain
| |
Collapse
|
2
|
Balinda HU, Kelly WJ, Kaklamani VG, Lathrop KI, Canola MM, Ghamasaee P, Sareddy GR, Michalek J, Gilbert AR, Surapaneni P, Tiziani S, Pandey R, Chiou J, Lodi A, Floyd JR, Brenner AJ. Sacituzumab Govitecan in patients with breast cancer brain metastases and recurrent glioblastoma: a phase 0 window-of-opportunity trial. Nat Commun 2024; 15:6707. [PMID: 39112464 PMCID: PMC11306739 DOI: 10.1038/s41467-024-50558-9] [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: 03/13/2024] [Accepted: 07/15/2024] [Indexed: 08/10/2024] Open
Abstract
Sacituzumab Govitecan (SG) is an antibody-drug conjugate that has demonstrated efficacy in patients with TROP-2 expressing epithelial cancers. In a xenograft model of intracranial breast cancer, SG inhibited tumor growth and increased mouse survival. We conducted a prospective window-of-opportunity trial (NCT03995706) at the University of Texas Health Science Center at San Antonio to examine the intra-tumoral concentrations and intracranial activity of SG in patients undergoing craniotomy for breast cancer with brain metastases (BCBM) or recurrent glioblastoma (rGBM). We enrolled 25 patients aged ≥18 years diagnosed with BCBM and rGBM to receive a single intravenous dose of SG at 10 mg/kg given one day before resection and continued on days 1 and 8 of 21-day cycles following recovery. The PFS was 8 months and 2 months for BCBM and rGBM cohorts, respectively. The OS was 35.2 months and 9.5 months, respectively. Grade≥3 AE included neutropenia (28%), hypokalemia (8%), seizure (8%), thromboembolic event (8%), urinary tract infection (8%) and muscle weakness of the lower limb (8%). In post-surgical tissue, the median total SN-38 was 249.8 ng/g for BCBM and 104.5 ng/g for rGBM, thus fulfilling the primary endpoint. Biomarker analysis suggests delivery of payload by direct release at target site and that hypoxic changes do not drive indirect release. Secondary endpoint of OS was 35.2 months for the BCBM cohort and 9.5 months for rGBM. Non-planned exploratory endpoint of ORR was 38% for BCBM and 29%, respectively. Exploratory endpoint of Trop-2 expression was observed in 100% of BCBM and 78% of rGBM tumors. In conclusion, SG was found to be well tolerated with adequate penetration into intracranial tumors and promising preliminary activity within the CNS. Trial Registration: Trial (NCT03995706) enrolled at Clinical Trials.gov as Neuro/Sacituzumab Govitecan/Breast Brain Metastasis/Glioblastoma/Ph 0: https://clinicaltrials.gov/study/NCT03995706?cond=NCT03995706 .
Collapse
Affiliation(s)
- Henriette U Balinda
- Mays Cancer Center at UT Health San Antonio, 7979 Wurzbach Road, San Antonio, TX, 78229, USA
| | - William J Kelly
- Mays Cancer Center at UT Health San Antonio, 7979 Wurzbach Road, San Antonio, TX, 78229, USA
| | - Virginia G Kaklamani
- Mays Cancer Center at UT Health San Antonio, 7979 Wurzbach Road, San Antonio, TX, 78229, USA
| | - Kate I Lathrop
- Mays Cancer Center at UT Health San Antonio, 7979 Wurzbach Road, San Antonio, TX, 78229, USA
| | - Marcela Mazo Canola
- Mays Cancer Center at UT Health San Antonio, 7979 Wurzbach Road, San Antonio, TX, 78229, USA
| | - Pegah Ghamasaee
- Department of Neurosurgery, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA
| | - Gangadhara R Sareddy
- Mays Cancer Center at UT Health San Antonio, 7979 Wurzbach Road, San Antonio, TX, 78229, USA
- Department of Obstetrics & Gynecology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA
| | - Joel Michalek
- Department of Population Health Sciences Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, 8403 Floyd Curl Drive, San Antonio, TX, 78229, USA
| | - Andrea R Gilbert
- Department of Pathology and Laboratory Medicine, University of Texas Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA
| | - Prathibha Surapaneni
- START Center for Cancer Care, 155 E Sonterra Blvd STE. 200, San Antonio, TX, 78258, USA
| | - Stefano Tiziani
- Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, Austin, TX, 78723, USA
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX, 78712, USA
- Department of Oncology, Dell Medical School, Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX, 78723, USA
- Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, 78723, USA
| | - Renu Pandey
- Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, Austin, TX, 78723, USA
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Jennifer Chiou
- Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, Austin, TX, 78723, USA
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Alessia Lodi
- Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, Austin, TX, 78723, USA
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX, 78712, USA
| | - John R Floyd
- Department of Neurosurgery, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA
| | - Andrew J Brenner
- Mays Cancer Center at UT Health San Antonio, 7979 Wurzbach Road, San Antonio, TX, 78229, USA.
| |
Collapse
|
3
|
Zarnoosheh Farahani T, Nejadmoghaddam MR, Sari S, Ghahremanzadeh R, Minai-Tehrani A. Generation of anti-SN38 antibody for loading efficacy and therapeutic monitoring of SN38-containing therapeutics. Heliyon 2024; 10:e33232. [PMID: 39021912 PMCID: PMC11253049 DOI: 10.1016/j.heliyon.2024.e33232] [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] [Received: 11/09/2023] [Revised: 06/17/2024] [Accepted: 06/17/2024] [Indexed: 07/20/2024] Open
Abstract
SN38, one of the most potent anti-tumor analogues of the camptothecins (CPTs), has limitations in its direct formulation as an anticancer agent due to its super toxicity and poor solubility in water and pharmaceutically approved solvents. However, it has garnered significant scientific interest as a payload in conjugated nanomedicine platforms (e.g., SN-38lip, NK012, SNB-101, and ADCs) to enhance their effectiveness and safety. The development of these platforms necessitates a convenient quantitative determination of SN38 in preclinical and clinical studies, a need that our study directly addresses, offering a practical solution to a pressing problem in cancer research and drug development. This study details the meticulous process of generating poly and monoclonal antibodies (pAb and mAb) against SN38 and their application to measure the SN38 in naked and conjugated forms of SN38-conjugated ADCs. For this purpose, two haptens of SN38 were synthesized by introducing the glycine or 4-amino-4-oxobutanyol(glycine) moiety as a conjugation functional group of the SN38. IR, NMR and mass spectrometric techniques confirmed the chemical modifications of the haptens. The haptens were then conjugated to each bovine serum albumin (BSA) and keyhole limpet hemocyanin (KLH) protein. The SN38-KLH conjugates were meticulously examined for immunization and generation of pAb and mAb. The immunization efficiency, reactivity, binding affinity, specificity, and cross-reactivity of purified pAb and mAb against Irinotecan, a model for the emergence of an SN38 derivative in clinical settings, were evaluated using ELISA and western blotting (WB) techniques. Conjugation efficiency of the SN38 to the KLH was increased using 4-amino-4-oxobutanyol(glycine) moiety, as its immunization efficacy was more to generate pAb. Furthermore, only this hapten could immunized mice to generate mAb recognizing SN38 with nanomolar equilibrium affinity. Our recent findings strongly support the notion that the generated pAb employed in developing an ELISA effectively ascertains the presence of SN38 in SN38-conjugated ADC, with a test midpoint EC50 of 2.5 μg/mL. Our study's unique contribution to the field lies in the development of specific antibodies against SN38 for measuring it on ADC, a feat that has not been achieved before. These immunoassays can be readily applied to detect other SN38-conjugate therapeutic platforms, thereby enhancing their clinical knowledge translation. The affinity of both pAb and mAb also meets the acceptance criteria for quantifying SN38 in fluidic material, as well as in Therapeutic drug monitoring (TDM) studies, a crucial aspect of personalized medicine. The potential applications of the anti-SN38 antibodies extend to reducing SN38-induced systemic toxicity through an inverse targeting strategy, a novel approach that piques further interest in our findings.
Collapse
Affiliation(s)
- Tahereh Zarnoosheh Farahani
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Nanobiotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | | | - Soyar Sari
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ramin Ghahremanzadeh
- Nanobiotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Arash Minai-Tehrani
- Nanobiotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| |
Collapse
|
4
|
Briki M, Murisier A, Guidi M, Seydoux C, Buclin T, Marzolini C, Girardin FR, Thoma Y, Carrara S, Choong E, Decosterd LA. Liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS) methods for the therapeutic drug monitoring of cytotoxic anticancer drugs: An update. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1236:124039. [PMID: 38490042 DOI: 10.1016/j.jchromb.2024.124039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/26/2024] [Accepted: 01/31/2024] [Indexed: 03/17/2024]
Abstract
In the era of precision medicine, there is increasing evidence that conventional cytotoxic agents may be suitable candidates for therapeutic drug monitoring (TDM)- guided drug dosage adjustments and patient's tailored personalization of non-selective chemotherapies. To that end, many liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS) assays have been developed for the quantification of conventional cytotoxic anticancer chemotherapies, that have been comprehensively and critically reviewed. The use of stable isotopically labelled internal standards (IS) of cytotoxic drugs was strikingly uncommon, accounting for only 48 % of the methods found, although their use could possible to suitably circumvent patients' samples matrix effects variability. Furthermore, this approach would increase the reliability of cytotoxic drug quantification in highly multi-mediated cancer patients with complex fluctuating pathophysiological and clinical conditions. LC-MS/MS assays can accommodate multiplexed analyses of cytotoxic drugs with optimal selectivity and specificity as well as short analytical times and, when using stable-isotopically labelled IS for quantification, provide concentrations measurements with a high degree of certainty. However, there are still organisational, pharmacological, and medical constraints to tackle before TDM of cytotoxic drugs can be more largely adopted in the clinics for contributing to our ever-lasting quest to improve cancer treatment outcomes.
Collapse
Affiliation(s)
- M Briki
- Laboratory of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; Service of Clinical Pharmacology, Department of Medicine, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; Bio/CMOS Interfaces Laboratory, École Polytechnique Fédérale de Lausanne-EPFL, 2002 Neuchâtel, Switzerland
| | - A Murisier
- Laboratory of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
| | - M Guidi
- Service of Clinical Pharmacology, Department of Medicine, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, University of Lausanne, 1206 Geneva, Switzerland; Centre for Research and Innovation in Clinical Pharmaceutical Sciences, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
| | - C Seydoux
- Internal Medicine Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
| | - T Buclin
- Service of Clinical Pharmacology, Department of Medicine, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
| | - C Marzolini
- Laboratory of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
| | - F R Girardin
- Laboratory of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; Service of Clinical Pharmacology, Department of Medicine, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
| | - Y Thoma
- School of Engineering and Management Vaud, HES-SO University of Applied Sciences and Arts Western Switzerland, 1401 Yverdon-les-Bains, Switzerland
| | - S Carrara
- Bio/CMOS Interfaces Laboratory, École Polytechnique Fédérale de Lausanne-EPFL, 2002 Neuchâtel, Switzerland
| | - E Choong
- Laboratory of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
| | - L A Decosterd
- Laboratory of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland.
| |
Collapse
|
5
|
Gasthuys E, van Ovost J, Vande Casteele S, Cosyns S, Ceelen W, Van Bocxlaer J, Vermeulen A. Development and validation of an UPLC-MS/MS method for the determination of irinotecan (CPT-11), SN-38 and SN-38 glucuronide in human plasma and peritoneal tumor tissue from patients with peritoneal carcinomatosis. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1233:123980. [PMID: 38215697 DOI: 10.1016/j.jchromb.2023.123980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/17/2023] [Accepted: 12/19/2023] [Indexed: 01/14/2024]
Abstract
Irinotecan (CPT-11), an antineoplastic drug, is used for the treatment of colorectal and pancreatic cancer due to its topoisomerase I inhibitory activity. CPT-11 is a prodrug which is converted to its active metabolite SN-38 by carboxylesterases. SN-38 is further metabolized to its inactive metabolite SN-38 glucuronide. When evaluating the pharmacokinetic properties of CPT-11 and its metabolites, it is important to accurately assess the concentrations in both plasma as well as tumor tissues. Therefore, the aim of the current study was to develop and validate a robust and sensitive ultra-high performance liquid chromatography-tandem mass spectrometry method to quantify the concentration of CPT-11 and its metabolites (SN-38 and SN-38 glucuronide) in human plasma and peritoneal tumor tissue. The sample preparation of plasma and tumor tissue consisted of protein precipitation and enzymatic digestion/liquid-liquid extraction, respectively. Chromatographic separation was achieved with an Acquity UPLC BEH C18 column combined with a VanGuard pre-column. The mobile phases consisted of water +0.1 % formic acid (mobile phase A) and acetonitrile +0.1 % formic acid (mobile phase B). Mass analysis was performed using a Xevo TQS tandem mass spectrometer in the positive electrospray ionization mode. Method validation was successfully performed by assessing linearity, precision and accuracy, lower limit of quantification, carry over, selectivity, matrix effect and stability according to the following guidelines: "Committee for Medicinal Products for Human use, Guideline on Bioanalytical Method Validation". A cross-validation of the developed method was performed in a pilot pharmacokinetic study, demonstrating the usefulness of the current method to quantify CPT-11 and its metabolites in the different matrices.
Collapse
Affiliation(s)
- Elke Gasthuys
- Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| | - Judith van Ovost
- Department of Human Structure and Repair, Laboratory of Experimental Surgery Faculty of Medicine and Health Sciences, Ghent University, Corneel Heymanslaan 10, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Corneel Heymanslaan 10, 9000 Ghent, Belgium
| | - Sofie Vande Casteele
- Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Sarah Cosyns
- Department of Human Structure and Repair, Laboratory of Experimental Surgery Faculty of Medicine and Health Sciences, Ghent University, Corneel Heymanslaan 10, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Corneel Heymanslaan 10, 9000 Ghent, Belgium
| | - Wim Ceelen
- Department of Human Structure and Repair, Laboratory of Experimental Surgery Faculty of Medicine and Health Sciences, Ghent University, Corneel Heymanslaan 10, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Corneel Heymanslaan 10, 9000 Ghent, Belgium; Department of GI Surgery Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| | - Jan Van Bocxlaer
- Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - An Vermeulen
- Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| |
Collapse
|
6
|
Kong X, Xu Y, Gao P, Liu Y, Wang X, Zhao M, Jiang Y, Yang H, Cao Y, Ma L. Rapid detection of the irinotecan-related UGT1A1*28 polymorphism by asymmetric PCR melting curve analysis using one fluorescent probe. J Clin Lab Anal 2022; 36:e24578. [PMID: 35766440 PMCID: PMC9396174 DOI: 10.1002/jcla.24578] [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] [Received: 12/29/2021] [Revised: 06/09/2022] [Accepted: 06/18/2022] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Determination of UGT1A1 (TA)n polymorphism prior to irinotecan therapy is necessary to avoid severe adverse drug effects. Thus, accurate and reliable genotyping methods for (TA)n polymorphism are highly desired. Here, we present a new method for polymerase chain reaction (PCR) melting curve analysis using one fluorescent probe to discriminate the UGT1A1*1 [(TA)6 ] and *28 [(TA)7 ] genotypes. METHODS After protocol optimization, this technique was applied for genotyping of 64 patients (including 23 with UGT1A1*1/*1, 22 with *1/*28, and 19 with *28/*28) recruited between 2016 and 2021 in China-Japan Friendship Hospital. The accuracy of the method was evaluated by comparing the results with those of direct sequencing and fragment analysis. The intra- and inter-run precision of the melting temperatures (Tm s) were calculated to assess the reliability, and the limit of detection was examined to assess the sensitivity. RESULTS All genotypes were correctly identified with the new method, and its accuracy was higher than that of fragment analysis. The intra- and inter-run coefficients of variation for the Tm s were both ≤0.27%, with standard deviations ≤0.14°C. The limit of detection was 0.2 ng of input genomic DNA. CONCLUSION The developed PCR melting curve analysis using one fluorescent probe can provide accurate, reliable, rapid, simple, and low-cost detection of UGT1A1 (TA)n polymorphism, and its use can be easily generalized in clinical laboratories with a fluorescent PCR platform.
Collapse
Affiliation(s)
- Xiaomu Kong
- Department of Clinical Laboratory, China-Japan Friendship Hospital, Beijing, China
| | - Ye Xu
- Engineering Research Centre of Molecular Diagnostics, Ministry of Education, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Peng Gao
- Department of Clinical Laboratory, China-Japan Friendship Hospital, Beijing, China
| | - Yi Liu
- Department of Clinical Laboratory, China-Japan Friendship Hospital, Beijing, China
| | - Xuran Wang
- Engineering Research Centre of Molecular Diagnostics, Ministry of Education, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Meimei Zhao
- Department of Clinical Laboratory, China-Japan Friendship Hospital, Beijing, China
| | - Yongwei Jiang
- Department of Clinical Laboratory, China-Japan Friendship Hospital, Beijing, China
| | - Hui Yang
- Department of Clinical Laboratory, China-Japan Friendship Hospital, Beijing, China
| | - Yongtong Cao
- Department of Clinical Laboratory, China-Japan Friendship Hospital, Beijing, China
| | - Liang Ma
- Department of Clinical Laboratory, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
7
|
Sakai S, Kobuchi S, Ito Y, Sakaeda T. Assessment of drug-drug interaction and optimization in capecitabine and irinotecan combination regimen using a physiologically based pharmacokinetic model. J Pharm Sci 2021; 111:1522-1530. [PMID: 34965386 DOI: 10.1016/j.xphs.2021.12.021] [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: 08/26/2021] [Revised: 12/22/2021] [Accepted: 12/22/2021] [Indexed: 10/19/2022]
Abstract
Capecitabine and irinotecan (CPT-11) combination regimen (XELIRI) is used for colorectal cancer treatment. Capecitabine is metabolized to 5-fluorouracil (5-FU) by three enzymes, including carboxylesterase (CES). CES can also convert CPT-11 to 7-ethyl-10-hydroxycamptotecin (SN-38). CES is involved in the metabolic activation of both capecitabine and CPT-11, and it is possible that drug-drug interactions occur in XELIRI. Here, a physiologically based pharmacokinetic (PBPK) model was developed to evaluate drug-drug interactions. Capecitabine (180 mg/kg) and CPT-11 (180 mg/m2) were administered to rats, and blood (250 μL) was collected from the jugular vein nine times after administration. Metabolic enzyme activities and Ki values were calculated through in vitro experiments. The plasma concentration of 5-FU in XELIRI was significantly decreased compared to capecitabine monotherapy, and metabolism of capecitabine by CES was inhibited by CPT-11. A PBPK model was developed based on the in vivo and in vitro results. Furthermore, a PBPK model-based simulation was performed with the capecitabin dose ranging from 0 to 1000mol/kg in XELIRI, and it was found that an approximately 1.7-fold dosage of capecitabine was required in XELIRI for comparable 5-FU exposure with capecitabine monotherapy. PBPK model-based simulation will contribute to the optimization of colorectal cancer chemotherapy using XELIRI.
Collapse
Affiliation(s)
- Shuhei Sakai
- Department of Pharmacokinetics, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan
| | - Shinji Kobuchi
- Department of Pharmacokinetics, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan
| | - Yukako Ito
- Department of Pharmacokinetics, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan
| | - Toshiyuki Sakaeda
- Department of Pharmacokinetics, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan..
| |
Collapse
|
8
|
Aoullay Z, Van Wijk XMR, Yanhui M, Meddah B, Lynch KL, Cherrah Y, Wu AHB. Development of an LC-MS/MS Method for Measurement of Irinotecan and Its Major Metabolites in Plasma: Technical Considerations. Lab Med 2021; 53:47-52. [PMID: 34351422 DOI: 10.1093/labmed/lmab059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE Irinotecan (CPT-11) is an important drug used in the treatment of several solid tumor types. To minimize its toxicity, therapeutic drug monitoring of CPT-11 and its major metabolites (SN-38, SN-38-glucuronide [SN-38G], and APC) has been proposed. We aimed to develop a liquid chromatography-tandem mass spectrometry (LC-MS/MS) method for the quantification of CPT-11 and its major metabolites in plasma. METHODS Specimen preparation consisted of protein precipitation, evaporation, and reconstitution. Analyses were performed on a C18 column using reverse-phase gradient elution. Electrospray ionization and multiple reaction monitoring in positive mode were used for MS. The following heavy isotope-labeled internal standards were used: CPT-11 D10, SN-38 D3, SN-38G D3, and APC D3. RESULTS We found that CPT-11, SN-38G, and APC eluted at ~4.6 to 4.7 minutes, and SN-38 eluted at ~5.1 to 5.2 minutes. A second peak for SN-38 was detected at ~4.6 to 4.7 minutes. Given that the structure of SN-38 is found in CPT-11, SN-38G, and APC, and in the CPT-11 D10 used here, in-source fragmentation was the likely cause. In addition, we found that a low-level SN-38 impurity was present in CPT-11 D10 and to a lesser extent in SN-38 D3. CONCLUSION When developing methods for CPT-11 and its metabolites, it is important to consider the effects of in-source fragmentation and the choice of internal standards.
Collapse
Affiliation(s)
- Zineb Aoullay
- Laboratory of Pharmacology and Toxicology, Faculty of Medicine and Pharmacy of Rabat, University Mohamed V Rabat, Rabat, Morocco.,Department of Laboratory Medicine, University of California and Zuckerberg San Francisco General Hospital, San Francisco, CA, US
| | - Xander M R Van Wijk
- Department of Laboratory Medicine, University of California and Zuckerberg San Francisco General Hospital, San Francisco, CA, US.,Department of Pathology, University of Chicago, Chicago, IL, US
| | - Ma Yanhui
- Department of Laboratory Medicine, University of California and Zuckerberg San Francisco General Hospital, San Francisco, CA, US.,Department of Laboratory Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bouchra Meddah
- Laboratory of Pharmacology and Toxicology, Faculty of Medicine and Pharmacy of Rabat, University Mohamed V Rabat, Rabat, Morocco
| | - Kara L Lynch
- Department of Laboratory Medicine, University of California and Zuckerberg San Francisco General Hospital, San Francisco, CA, US
| | - Yahia Cherrah
- Laboratory of Pharmacology and Toxicology, Faculty of Medicine and Pharmacy of Rabat, University Mohamed V Rabat, Rabat, Morocco
| | - Alan H B Wu
- Department of Laboratory Medicine, University of California and Zuckerberg San Francisco General Hospital, San Francisco, CA, US
| |
Collapse
|
9
|
Tsotsou GE, Gkotzamani P, Petro V, Argyropoulou A, Karkalousos P. A simple, rapid and low-cost spectrophotometric method for irinotecan quantification in human plasma and in pharmaceutical dosage forms. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2021; 13:258-266. [PMID: 33367449 DOI: 10.1039/d0ay02201b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Irinotecan is an anticancer drug for which significant benefits from personalised dosing are expected. Quick procedures are therefore essential for monitoring irinotecan in treated patients. The objective of this work was to develop and validate a rapid and simple visible spectrophotometric method for quantitative determination of irinotecan in pharmaceutical dosage forms and to further investigate its usefulness for irinotecan analysis in plasma. Based on the shift of the irinotecan 355/368 nm-peak at very low pH (0.2) to 400 nm, we established a linear relationship between absorbance at 400 nm and irinotecan concentration in dilutions of an irinotecan solution for injection (R2 ≥ 0.999) and in plasma containing irinotecan (R2 ≥ 0.995). Background absorbance correction at 455 nm was essential to minimise background interference, solely in plasma samples. We fully validated the assay for quality control of the irinotecan solution in the injection dosage form: the standard curve was linear over the concentration range of 0.90 to at least 37.00 μg ml-1. The CV% on all quality control levels was determined to be ≤5.81% for repeatability and ≤6.62% for intermediate precision. Recovery was between 96.5% and 101.9%. Upon comparison with the LC/UV method, we demonstrated very good agreement and acceptable bias between the two methods (slope 0.973, y-intercept 0.0064). Similarly, the technical parameters of the assay in plasma satisfied international guidelines for method validation: the useful analytical range was determined to be between 0.93 and at least 10.00 μg ml-1. This is suitable for quantifying irinotecan in the plasma of treated patients, in the upper region of its therapeutic window, to decide whether dose adjustment is required. Repeatability and intermediate precision (CV%) were within 4.49% and 9.91%, respectively. Recovery was between 96.3% and 103.8%. There was a lack of significant interference by mild hemolysis or by icterus. Irinotecan extraction efficiency from plasma was within 77.9-68.5%. Our results indicated that the proposed method allows quantitative determination of irinotecan plasma levels with acceptable analytical characteristics. The advantages of the proposed method in both matrices, in terms of specificity, rapidity, simplicity, environmental impact and cost effectiveness, are discussed.
Collapse
Affiliation(s)
- Georgia Eleni Tsotsou
- Laboratory of Chemistry, Biochemistry and Cosmetology, Department of Biomedical Sciences, University of West Attica, Egaleo 122 43, Greece.
| | - Panagiota Gkotzamani
- Laboratory of Chemistry, Biochemistry and Cosmetology, Department of Biomedical Sciences, University of West Attica, Egaleo 122 43, Greece.
| | - Victoria Petro
- Laboratory of Chemistry, Biochemistry and Cosmetology, Department of Biomedical Sciences, University of West Attica, Egaleo 122 43, Greece.
| | - Ariadne Argyropoulou
- Laboratory of Chemistry, Biochemistry and Cosmetology, Department of Biomedical Sciences, University of West Attica, Egaleo 122 43, Greece.
| | - Petros Karkalousos
- Laboratory of Chemistry, Biochemistry and Cosmetology, Department of Biomedical Sciences, University of West Attica, Egaleo 122 43, Greece.
| |
Collapse
|
10
|
Zhao K, Guo T, Sun X, Xiong T, Ren X, Wu L, Yang R, Sun H, Shi S, Zhang J. Mechanism and optimization of supramolecular complexation-enhanced fluorescence spectroscopy for the determination of SN-38 in plasma and cells. LUMINESCENCE 2020; 36:531-542. [PMID: 33125824 DOI: 10.1002/bio.3973] [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: 02/17/2020] [Revised: 10/11/2020] [Accepted: 10/22/2020] [Indexed: 11/06/2022]
Abstract
Quantitative detection of two different forms of SN-38 in biological samples is, currently, cumbersome and difficult. A revisit to the mechanism of supramolecular complexation-enhanced fluorescence spectroscopy helps to optimize the determination of SN-38 in plasma and the cellular pharmacokinetics in A549 cells based on the supramolecular complexation. Firstly, the inclusion mechanism dominated by thermodynamic constants was determined by measuring kinetic/thermodynamic parameters (kon , koff , ΔG, ΔH, ΔS). On this basis, the best effect of fluorescence sensitization was optimized through screening the interaction conditions (cyclodextrin species and concentrations, drug levels, temperature, pH of the buffer, and reaction time). Furthermore, the proportional relationship between the concentration of the inclusion complex and the fluorescence intensity was confirmed. Finally, a highly sensitive, selective spectrofluorimetric method was established and validated for quantitative analysis of the lactone and carboxylate molecular states of SN-38 plasma levels in rats and cell membrane transfer kinetics in A549 cell lines. The limits of detection for the lactone and carboxylate forms in plasma were found to be 0.44 ng·ml-1 and 0.28 ng·ml-1 , respectively. Precision and accuracy met the requirements of biological samples analysis. The proposed detection method provided a reference for elucidating the biodistribution of SN-38.
Collapse
Affiliation(s)
- Kena Zhao
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China.,Center for Drug Delivery System, Shanghai Institute of Materia Medica, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Tao Guo
- Center for Drug Delivery System, Shanghai Institute of Materia Medica, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xian Sun
- Center for Drug Delivery System, Shanghai Institute of Materia Medica, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Ting Xiong
- Center for Drug Delivery System, Shanghai Institute of Materia Medica, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai, 201203, China.,Key Laboratory of Modern Chinese Medicine Preparations, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, China
| | - Xiaohong Ren
- Center for Drug Delivery System, Shanghai Institute of Materia Medica, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Li Wu
- Center for Drug Delivery System, Shanghai Institute of Materia Medica, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Rui Yang
- Institute for Control of Pharmaceutical Excipient and Packaging Material, National Institutes for Food and Drug Control, Beijing, 100050, China.,NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, National Institutes for Food and Drug Control, Beijing, 102600, China
| | - Huimin Sun
- Institute for Control of Pharmaceutical Excipient and Packaging Material, National Institutes for Food and Drug Control, Beijing, 100050, China.,NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, National Institutes for Food and Drug Control, Beijing, 102600, China
| | - Senlin Shi
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Jiwen Zhang
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China.,Center for Drug Delivery System, Shanghai Institute of Materia Medica, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai, 201203, China.,Key Laboratory of Modern Chinese Medicine Preparations, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, China.,NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, National Institutes for Food and Drug Control, Beijing, 102600, China
| |
Collapse
|
11
|
Pandey R, Gruslova A, Chiou J, Brenner AJ, Tiziani S. Stable Isotope Dilution LC-HRMS Assay To Determine Free SN-38, Total SN-38, and SN-38G in a Tumor Xenograft Model after Intravenous Administration of Antibody-Drug Conjugate (Sacituzumab Govitecan). Anal Chem 2020; 92:1260-1267. [PMID: 31765123 DOI: 10.1021/acs.analchem.9b04419] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Antibody-drug conjugates (ADCs) have gained significant interest over the past few years due to their targeted delivery, higher efficacy, decreased toxicity and improved therapeutic index over conventional anticancer therapies. Sacituzumab govitecan (SG) is an ADC composed of a Trop-2-targeted antibody conjugated to the cytotoxic payload SN-38. SG is currently being evaluated in clinical trials of several solid cancers. In this nonclinical study, we have developed a highly sensitive and selective approach to measure free and total SN-38 and its glucuronidation metabolite (SN-38G) using stable isotope dilution (SID) ultrahigh-performance liquid chromatography-high resolution mass spectrometry (UHPLC-HRMS). An efficient and fast hydrolysis procedure (2 h at 100 °C) was established to release SN-38, conjugated to the antibody by carbonate linkage. The assay involves the extraction of free SN-38, SN-38G by protein precipitation, and subsequent acid hydrolysis of the protein layer to release antibody-bound SN-38. The developed UHPLC-HRMS method resulted in good linearity (r2 ≥ 0.997), accuracy (RE ≤ ± 9.1%), precision (CVs ≤ 7.7%), and extraction recoveries (85.6-109.3%). The validated method was applied in the plasma and tumor of mice bearing human brain (U251) and breast (MDA-MB-468) tumor xenografts treated with a single dose (0.5 mg) of SG for 6 h. Results revealed the presence of trace level of SN-38G and free SN-38 in plasma, which suggests an improved therapeutic index of SG. The established method makes a significant contribution to the assessment of SG in different cancers.
Collapse
|
12
|
Effects of inflammation on irinotecan pharmacokinetics and development of a best-fit PK model. Chem Biol Interact 2020; 316:108933. [DOI: 10.1016/j.cbi.2019.108933] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 11/29/2019] [Accepted: 12/19/2019] [Indexed: 01/11/2023]
|
13
|
Tarasiuk A, Fichna J. Gut microbiota: what is its place in pharmacology? Expert Rev Clin Pharmacol 2019; 12:921-930. [DOI: 10.1080/17512433.2019.1670058] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Aleksandra Tarasiuk
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
14
|
Qin Y, Kang A, Zhou G, Wang H, Wei W, Cao Y, Chen Y, Wang J, Shi Y, Tang Y, Jiang J. Carboxylesterase and UDP-glucuronosyltransferases mediated metabolism of irinotecan: In vitro
and in vivo
insights from quantitative ultra-performance liquid chromatography-mass spectrometry analysis. Biomed Chromatogr 2018; 32:e4320. [DOI: 10.1002/bmc.4320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 06/04/2018] [Accepted: 06/09/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Yifeng Qin
- College of Pharmacy and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization; Shaanxi University of Chinese Medicine; Xi'an Shaanxi Province China
- School of Traditional Chinese Pharmacy; China Pharmaceutical University; Nanjing Jiangsu Province China
| | - An Kang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae; Nanjing University of Chinese Medicine; Nanjing Jiangsu Province China
| | - Guisheng Zhou
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae; Nanjing University of Chinese Medicine; Nanjing Jiangsu Province China
| | - Huan Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae; Nanjing University of Chinese Medicine; Nanjing Jiangsu Province China
| | - Wei Wei
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae; Nanjing University of Chinese Medicine; Nanjing Jiangsu Province China
| | - Yujie Cao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae; Nanjing University of Chinese Medicine; Nanjing Jiangsu Province China
| | - Yanyan Chen
- College of Pharmacy and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization; Shaanxi University of Chinese Medicine; Xi'an Shaanxi Province China
| | - Jing Wang
- College of Pharmacy and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization; Shaanxi University of Chinese Medicine; Xi'an Shaanxi Province China
| | - Yajun Shi
- College of Pharmacy and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization; Shaanxi University of Chinese Medicine; Xi'an Shaanxi Province China
| | - Yuping Tang
- College of Pharmacy and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization; Shaanxi University of Chinese Medicine; Xi'an Shaanxi Province China
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae; Nanjing University of Chinese Medicine; Nanjing Jiangsu Province China
| | - Jianqin Jiang
- School of Traditional Chinese Pharmacy; China Pharmaceutical University; Nanjing Jiangsu Province China
| |
Collapse
|
15
|
Gao S, Tao Z, Zhou J, Wang Z, Yun Y, Li M, Zhang F, Chen W, Miao Y. One-Step Solid Extraction for Simultaneous Determination of Eleven Commonly Used Anticancer Drugs and One Active Metabolite in Human Plasma by HPLC-MS/MS. JOURNAL OF ANALYTICAL METHODS IN CHEMISTRY 2018; 2018:7967694. [PMID: 30046507 PMCID: PMC6036832 DOI: 10.1155/2018/7967694] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 05/07/2018] [Indexed: 06/08/2023]
Abstract
Therapeutic drug monitoring for anticancer drugs could timely reflect in vivo drug exposure, and it was a powerful tool for adjusting and maintaining drug concentration into a reasonable range, so that an enhanced efficacy and declined adverse reactions could be achieved. A liquid chromatography-tandem mass spectrometry method had been developed and fully validated for simultaneous determination of paclitaxel, docetaxel, vinblastine, vinorelbine, pemetrexed, carboplatin, etoposide, cyclophosphamide, ifosfamide, gemcitabine, irinotecan, and SN-38 (an active metabolite of irinotecan) in human plasma from cancer patients after intravenous drip of chemotherapy drugs. One-step solid-phase extraction was successfully applied using an Ostro sample preparation 96-well plate for plasma samples pretreated with acetonitrile containing 0.1% formic acid. Chromatographic separation was achieved on an Atlantis T3-C18 column (2.1 × 100 mm, 3.0 μm) with gradient elution using a mobile phase consisting of acetonitrile and 10 mM ammonium acetate plus 0.1% formic acid in water, and the flow rate was 0.25 mL/min. The Agilent G6410A triple quadrupole liquid chromatography-mass spectrometry system was operated under the multiple reaction monitoring mode with an electrospray ionization in the positive mode. Linear range was 25.0-2500.0 ng for paclitaxel, 10.0-1000.0 ng for docetaxel and SN-38, 100.0-10000.0 ng for vinorelbine and pemetrexed, 10.0-10000.0 ng for vinblastine and irinotecan, 1.0-1000.0 ng for cyclophosphamide and ifosfamide, 50.0-5000.0 ng for carboplatin, etoposide, and gemcitabine. Linearity coefficients of correlation were >0.99 for all analytes. The intraday and interday accuracy and precision of the method were within ±15.0% and less than 15%. The mean recovery and matrix effect as well as stability of all the analytes ranged from 56.2% to 98.9% and 85.2% to 101.3% as well as within ±15.0%. This robust and efficient method was successfully applied to implement therapeutic drug monitoring for cancer patients in clinical application.
Collapse
Affiliation(s)
- Shouhong Gao
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Zhengbo Tao
- Department of Orthopaedics, First Affiliated Hospital, China Medical University, 155 Nan Jing Bei Street, Shenyang, Liaoning 110001, China
| | - Jingya Zhou
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Zhipeng Wang
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Yunlei Yun
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Mingming Li
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Feng Zhang
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Wansheng Chen
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Yejun Miao
- Department of Psychiatry, Ankang Hospital, Ningbo, Zhejiang 315000, China
| |
Collapse
|
16
|
Atasilp C, Chansriwong P, Sirachainan E, Reungwetwattana T, Puangpetch A, Prommas S, Sirilerttrakul S, Rerkarmnuaychoke B, Wongwaisayawan S, Sukasem C. Determination of irinotecan, SN-38 and SN-38 glucuronide using HPLC/MS/MS: Application in a clinical pharmacokinetic and personalized medicine in colorectal cancer patients. J Clin Lab Anal 2017; 32. [PMID: 28393405 DOI: 10.1002/jcla.22217] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 02/27/2017] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Irinotecan (CPT-11) is chemotherapy used mainly in the metastatic colorectal cancer. The purpose of this study was to develop and validate the LC-MS/MS for the simultaneous determination of CPT-11, SN-38, and SN-38G. METHODS A 100 μL of plasma was prepared after protein precipitation and analyzed on a C18 column using 0.1% acetic acid in water and 0.1% acetic acid in acetonitrile as mobile phases. The mass spectrometer worked with multiple reaction monitoring (MRM) in positive scan mode. The standard curves were linear on a concentration range of 5-10 000 ng/mL for CPT-11, 5-1000 ng/mL for SN-38, and 8-1000 ng/mL for SN-38G. RESULTS In this assay, the intra and interday precision consisted of ≤9.11% and ≤11.29% for CPT-11, ≤8.70% and 8.31% for SN-38, and ≤9.90 and 9.64% for SN-38G. CONCLUSION This method was successfully used to quantify CPT-11, SN-38, and SN-38G and applied to a pharmacokinetic study.
Collapse
Affiliation(s)
- Chalirmporn Atasilp
- Division of Pharmacogenomics and Personalized Medicine, Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.,Laboratory for Pharmacogenomics, Clinical Pathology, Somdetch Phra Debharatana Medical Centre, Ramathibodi Hospital, Bangkok, Thailand
| | - Pichai Chansriwong
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Ekapob Sirachainan
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Thanyanan Reungwetwattana
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Apichaya Puangpetch
- Division of Pharmacogenomics and Personalized Medicine, Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.,Laboratory for Pharmacogenomics, Clinical Pathology, Somdetch Phra Debharatana Medical Centre, Ramathibodi Hospital, Bangkok, Thailand
| | - Santirhat Prommas
- Division of Pharmacogenomics and Personalized Medicine, Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.,Laboratory for Pharmacogenomics, Clinical Pathology, Somdetch Phra Debharatana Medical Centre, Ramathibodi Hospital, Bangkok, Thailand
| | - Suwannee Sirilerttrakul
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Budsaba Rerkarmnuaychoke
- Division of Human Genetics Laboratory, Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Sansanee Wongwaisayawan
- Division of Anatomical Pathology, Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Chonlaphat Sukasem
- Division of Pharmacogenomics and Personalized Medicine, Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.,Laboratory for Pharmacogenomics, Clinical Pathology, Somdetch Phra Debharatana Medical Centre, Ramathibodi Hospital, Bangkok, Thailand
| |
Collapse
|