1
|
Song F, Yi X, Zheng X, Zhang Z, Zhao L, Shen Y, Zhi Y, Liu T, Liu X, Xu T, Hu X, Zhang Y, Shou H, Huang P. Zebrafish patient-derived xenograft system for predicting carboplatin resistance and metastasis of ovarian cancer. Drug Resist Updat 2025; 78:101162. [PMID: 39571238 DOI: 10.1016/j.drup.2024.101162] [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: 09/26/2024] [Revised: 11/01/2024] [Accepted: 11/03/2024] [Indexed: 12/18/2024]
Abstract
AIMS Ovarian cancer (OC) remains a significant challenge in oncology due to high rates of drug resistance and disease relapse following standard treatment with surgery and platinum-based chemotherapy. Despite the widespread use of these treatments, no effective biomarkers currently exist to identify which patients will respond favorably to therapy. This study introduces a zebrafish patient-derived xenograft (PDX) system, capable of replicating both the carboplatin response and metastatic behavior observed in OC patients, within a rapid 3-day assay period. METHODS Two OC cell lines: carboplatin-sensitive (A2780) and resistant (OVCAR8) were used to assess differential responses to treatment in murine and zebrafish xenograft models. Tumor tissues from 16 OC patients were implanted into zebrafish embryos to test carboplatin responses and predict metastasis. Additionally, eight clinical OC samples were directly implanted into zebrafish embryos as part of a proof-of-concept demonstration. RESULTS The zebrafish xenografts accurately reflected the carboplatin sensitivity and resistance patterns seen in in vitro and murine models. The zebrafish PDX model demonstrated a 67 % success rate for implantation and a 100 % success rate for engraftment. Notably, the model effectively distinguished between metastatic and non-metastatic disease, with an area under the ROC curve (AUC) of 0.818. Furthermore, the zebrafish PDX model showed a high concordance with patient-specific responses to carboplatin. CONCLUSIONS This zebrafish PDX model offers a fast, accurate, and clinically relevant platform for evaluating carboplatin response and predicting metastasis in OC patients. It holds significant potential for advancing personalized medicine, allowing for more precise therapeutic outcome predictions and individualized treatment strategies.
Collapse
Affiliation(s)
- Feifeng Song
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China; Zhejiang Provincial Clinical Research Center for Malignant Tumor, Hangzhou 310014, China; Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Hangzhou 310014, China
| | - Xiaofen Yi
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China; Zhejiang Provincial Clinical Research Center for Malignant Tumor, Hangzhou 310014, China; Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Hangzhou 310014, China
| | - Xiaowei Zheng
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China; Zhejiang Provincial Clinical Research Center for Malignant Tumor, Hangzhou 310014, China; Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Hangzhou 310014, China
| | - Zhentao Zhang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China
| | - Linqian Zhao
- Center for Reproductive Medicine, Department of Gynecology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China
| | - Yan Shen
- Center for Reproductive Medicine, Department of Gynecology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China
| | - Ye Zhi
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China
| | - Ting Liu
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China; Zhejiang Provincial Clinical Research Center for Malignant Tumor, Hangzhou 310014, China; Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Hangzhou 310014, China
| | - Xiaozhen Liu
- General Surgery, Cancer Center, Department of Breast Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China
| | - Tong Xu
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China; Zhejiang Provincial Clinical Research Center for Malignant Tumor, Hangzhou 310014, China; Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Hangzhou 310014, China
| | - Xiaoping Hu
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China
| | - Yiwen Zhang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China; Zhejiang Provincial Clinical Research Center for Malignant Tumor, Hangzhou 310014, China; Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Hangzhou 310014, China.
| | - Huafeng Shou
- Center for Reproductive Medicine, Department of Gynecology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China.
| | - Ping Huang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, China; Zhejiang Provincial Clinical Research Center for Malignant Tumor, Hangzhou 310014, China; Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Hangzhou 310014, China.
| |
Collapse
|
2
|
Wang X, Zhu J, Li L, Zhao Q, Huang Y, Wen C, Chen D, Wu L. Utility of patient-derived xenografts to evaluate drug sensitivity and select optimal treatments for individual non-small-cell lung cancer patients. Mol Med 2024; 30:209. [PMID: 39528952 PMCID: PMC11556205 DOI: 10.1186/s10020-024-00934-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 09/11/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Patient-derived xenograft (PDX) is currently considered a preferred preclinical model to evaluate drug sensitivity, explore drug resistance mechanisms, and select individualized treatment regimens. METHODS Histopathological examination, immunohistochemistry and whole-exome sequencing confirmed similarity between our PDX tumors and primary tumors in terms of morphology and genetic characteristics. The drug reactivity of the PDX tumor was validated in vivo. The mechanisms of acquired resistance to Osimertinib PDX tumors were investigated by WES and WB. RESULTS We successfully established 13 NSCLC-PDXs derived from 62 patients, including eight adenocarcinomas, four squamous-cell carcinoma, and one large-cell neuroendocrine carcinoma. Histological subtype and clinical stage were significant factors affecting the successful PDXs establishment. The treatment responses to conventional chemotherapy in PDXs were entirely consistent with that of their corresponding patients. According to the genetic status of tumors, more appropriate targeted agents were selected in PDXs for their corresponding patients as alternative treatment options. In addition, a PDX model with acquired resistance to osimertinib was induced, and the overactivation of RAS mitogen-activated protein kinase (MAPK)-extracellular signal-regulated kinase (ERK) signaling pathway caused by the dual-specificity phosphatase 6 (DUSP6) M62I mutation was found to play a key role in the development of osimertinib resistance. Trametinib, a specific inhibitor of the MAPK-ERK pathway significantly slowed down the tumor growth in osimertinib-resistant PDX models, providing an alternative treatment in patients after osimertinib failure.
Collapse
Affiliation(s)
- Xiaoqing Wang
- Pharmacogenetics and Pharmacogenomics Laboratory, School of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Ju Zhu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Lingling Li
- Pharmacogenetics and Pharmacogenomics Laboratory, School of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Qilin Zhao
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yutang Huang
- Pharmacogenetics and Pharmacogenomics Laboratory, School of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Chunjie Wen
- Pharmacogenetics and Pharmacogenomics Laboratory, School of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Dan Chen
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Lanxiang Wu
- Pharmacogenetics and Pharmacogenomics Laboratory, School of Pharmacy, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
3
|
Abdelgalil AA, Alkahtani HM. Crizotinib: A comprehensive profile. PROFILES OF DRUG SUBSTANCES, EXCIPIENTS AND RELATED METHODOLOGY 2023; 48:39-69. [PMID: 37061275 DOI: 10.1016/bs.podrm.2022.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Crizotinib, approved in 2011, was the first approved inhibitor targeting anaplastic lymphoma kinase (ALK) It used for treatment of the patients with metastatic non-small cell lung cancer (NSCLC) that is anaplastic lymphoma kinase (ALK) positive. This chapter provides a complete review of crizotinib including nomenclature, physiochemical properties, methods of preparation, identification techniques and various qualitative and quantitative analytical techniques as well as pharmacology of crizotinib. In addition, the chapter also includes review of several methods for separation of crizotinib using chromatographic techniques.
Collapse
|
4
|
Honeywell RJ, Kathmann I, Giovannetti E, Tibaldi C, Smit EF, Rovithi MN, Verheul HM, Peters GJ. Epithelial Transfer of the Tyrosine Kinase Inhibitors Erlotinib, Gefitinib, Afatinib, Crizotinib, Sorafenib, Sunitinib, and Dasatinib: Implications for Clinical Resistance. Cancers (Basel) 2020; 12:3322. [PMID: 33182766 PMCID: PMC7696666 DOI: 10.3390/cancers12113322] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 11/07/2020] [Indexed: 12/19/2022] Open
Abstract
Background: tyrosine kinase inhibitors (TKIs) inhibit phosphorylation of signaling proteins. TKIs often show large variations in the clinic due to poor pharmacology, possibly leading to resistance. We compared gut absorption of inhibitors of epidermal growth factor receptor (erlotinib, gefitinib, and afatinib), ALK-cMET (crizotinib), PDGFR/BCR-Abl (dasatinib), and multikinase inhibitors (sunitinib and sorafenib). In clinical samples, we measured the disposition of each compound within various blood compartments. Methods: we used an optimized CaCo2 gut epithelial model to characterize 20 µM TKI absorption. The apical/basolateral transfer is considered to represent the gut/blood transfer. Drugs were measured using LC-MS/MS. Results: sorafenib and sunitinib showed the highest apical/basolateral transfer (Papp 14.1 and 7.7 × 10-6 cm/s, respectively), followed by dasatinib (3.4), afatinib (1.5), gefitinib (0.38), erlotinib (0.13), and crizotinib (n.d.). However, the net absorptions for dasatinib, afatinib, crizotinib, and erlotinib were highly negative (efflux ratios >5) or neutral/negative, sorafenib (0.86), gefitinib (1.0), and sunitinib (1.6). A high negative absorption may result in resistance because of a poor exposure of tissues to the drug. Accumulation of the TKIs at the end of the transfer period (A->B) was not detectable for erlotinib, very low for afatinib 0.45 pmol/μg protein), followed by gefitinib (0.79), dasatinib (1.1), sorafenib (1.65), and crizotinib (2.11), being highest for sunitinib (11.9). A similar pattern was found for accumulation of these drugs in other colon cell lines, WiDr and HT29. In clinical samples, drugs accumulated consistently in red blood cells; blood to plasma ratios were all > 3 (sorafenib) or over 30 for erlotinib. Conclusions: TKIs are consistently poorly absorbed, but accumulation in red blood cells seems to compensate for this.
Collapse
Affiliation(s)
- Richard J. Honeywell
- Department of Medical Oncology, Amsterdam UMC, VU University Medical Center, P.O. Box 7057, 1007 MB Amsterdam, The Netherlands; (R.J.H.); (I.K.); (E.G.); (M.N.R.)
- Department of Pharmacy, Amsterdam UMC, VU University Medical Center, P.O. Box 7057, 1007 MB Amsterdam, The Netherlands
| | - Ietje Kathmann
- Department of Medical Oncology, Amsterdam UMC, VU University Medical Center, P.O. Box 7057, 1007 MB Amsterdam, The Netherlands; (R.J.H.); (I.K.); (E.G.); (M.N.R.)
| | - Elisa Giovannetti
- Department of Medical Oncology, Amsterdam UMC, VU University Medical Center, P.O. Box 7057, 1007 MB Amsterdam, The Netherlands; (R.J.H.); (I.K.); (E.G.); (M.N.R.)
- Cancer Pharmacology Lab, AIRC Start-Up Unit, Fondazione Pisana per la Scienza, 56017 Pisa, Italy
| | - Carmelo Tibaldi
- Division of Oncology, Department of Oncology, S. Luca Hospital, 55100 Lucca, Italy;
| | - Egbert F. Smit
- Department of Thoracic Oncology, Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands;
| | - Maria N. Rovithi
- Department of Medical Oncology, Amsterdam UMC, VU University Medical Center, P.O. Box 7057, 1007 MB Amsterdam, The Netherlands; (R.J.H.); (I.K.); (E.G.); (M.N.R.)
| | - Henk M.W. Verheul
- Department of Medical Oncology, Radboud University Medical Center, Geert Grooteplein Zuid 8, 6525 GA Nijmegen, The Netherlands;
| | - Godefridus J. Peters
- Department of Medical Oncology, Amsterdam UMC, VU University Medical Center, P.O. Box 7057, 1007 MB Amsterdam, The Netherlands; (R.J.H.); (I.K.); (E.G.); (M.N.R.)
- Department of Biochemistry, Medical University of Gdansk, 80-211 Gdansk, Poland
| |
Collapse
|
5
|
Kath JE, Baranczak A. Target engagement approaches for pharmacological evaluation in animal models. Chem Commun (Camb) 2019; 55:9241-9250. [PMID: 31328738 DOI: 10.1039/c9cc02824b] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The field of chemical biology has introduced several approaches, typically using chemical probes, to measure the direct binding interaction of a small molecule with its biological target in cells. The use of these direct target engagement assays in pharmaceutical development can support mechanism of action hypothesis testing, rank ordering of compounds, and iterative improvements of chemical matter. This Feature Article highlights a newer application of these approaches: the quantification of target engagement in animal models to support late stage preclinical development and the nomination of a drug candidate to clinical trials. Broadly speaking, these efforts can be divided between compounds that covalently and reversibly interact with protein targets; recent examples for both categories are discussed for a range of targets, along with their limitations. New, promising technologies are also highlighted, in addition to the application of target engagement determination to new therapeutic modalities.
Collapse
Affiliation(s)
- James E Kath
- Drug Discovery Science and Technology, AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064-6101, USA.
| | | |
Collapse
|
6
|
Zebrafish Xenograft Model of Human Lung Cancer for Evaluating Osimertinib Resistance. BIOMED RESEARCH INTERNATIONAL 2019; 2019:3129748. [PMID: 31346515 PMCID: PMC6620834 DOI: 10.1155/2019/3129748] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 06/11/2019] [Indexed: 12/24/2022]
Abstract
About half of NSCLC patients with EGFR mutation had secondary mutation T790M after treatment with a first-generation tyrosine kinase inhibitor (TKI), Gefitinib. The third-generation of EGFR-TKI Osimertinib is suitable for patients with EGFR mutation and T790M mutation. However, drug screening for NSCLC patients after the emergence of acquired resistance has become a difficult problem for clinicians. In this study, we established drug-resistant cell lines of Gefitinib and Osimertinib to evaluate cell proliferation in vitro. And we investigated the inhibitory effect of different drug concentration gradients on cancer cells. Zebrafish with high homology to human genes were selected as xenotransplantation models to compare the effects of different concentrations of Osimertinib on the proliferation and angiogenesis of zebrafish tumors after transplantation of different lung cancer cell lines. It was confirmed that Osimertinib could inhibit the proliferation of tumor cells with EGFR mutation and T790M resistance mutation in zebrafish, which was consistent with the clinical research conclusion.
Collapse
|
7
|
Gu Y, Sai Y, Wang J, Yu M, Wang G, Zhang L, Ren H, Fan S, Ren Y, Qing W, Su W. Preclinical pharmacokinetics, disposition, and translational pharmacokinetic/pharmacodynamic modeling of savolitinib, a novel selective cMet inhibitor. Eur J Pharm Sci 2019; 136:104938. [PMID: 31132401 DOI: 10.1016/j.ejps.2019.05.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 05/14/2019] [Accepted: 05/23/2019] [Indexed: 01/16/2023]
Abstract
Savolitinib is a novel small-molecule selective cMet inhibitor. This work characterized its pharmacokinetics in preclinical phase, established the preclinical relationships between PK, cMet modulation and anti-tumor efficacy. In vitro and in vivo animal studies were performed for PK characterization. Savolitinib showed good absorption, moderate tissue distribution, low to intermediate clearance, and low accumulation. Hepatic oxidative metabolism followed by urinary and biliary excretions was the major elimination pathway. Based on preclinical PK data, human PK profiles were predicted using empirical methods. Pharmacodynamic studies for evaluating cMet inhibition and anti-tumor efficacy were conducted in nude mice bearing Hs746t xenograft. PK/PD models were built to link the PD measurements to nude mouse PK. The established integrated preclinical PK/PD model contained a two-compartment non-linear PK model, a biomarker link model and a tumor growth transit model. The IC50 of cMet inhibition and the concentration achieving half of the maximal Hs746t tumor reduction by savolitinib were equal to 12.5 and 3.7 nM (free drug), respectively. Based on the predicted human PK data, as well as the established PK/PD model in nude mouse, the human PD (cMet inhibition) profiles were also simulated. This research supported clinical development of savolitinib. Understanding the preclinical PK/PD relationship of savolitinib provides translational insights into the cMet-targeted drug development.
Collapse
Affiliation(s)
- Yi Gu
- Hutchison MediPharma Limited, Building 4, 720 Cailun Road, Zhang-Jiang Hi-Tech Park, Shanghai 201203, China.
| | - Yang Sai
- Hutchison MediPharma Limited, Building 4, 720 Cailun Road, Zhang-Jiang Hi-Tech Park, Shanghai 201203, China.
| | - Jian Wang
- Hutchison MediPharma Limited, Building 4, 720 Cailun Road, Zhang-Jiang Hi-Tech Park, Shanghai 201203, China.
| | - Meijing Yu
- Hutchison MediPharma Limited, Building 4, 720 Cailun Road, Zhang-Jiang Hi-Tech Park, Shanghai 201203, China.
| | - Guanglin Wang
- Hutchison MediPharma Limited, Building 4, 720 Cailun Road, Zhang-Jiang Hi-Tech Park, Shanghai 201203, China.
| | - Li Zhang
- Hutchison MediPharma Limited, Building 4, 720 Cailun Road, Zhang-Jiang Hi-Tech Park, Shanghai 201203, China.
| | - Hongcan Ren
- Hutchison MediPharma Limited, Building 4, 720 Cailun Road, Zhang-Jiang Hi-Tech Park, Shanghai 201203, China.
| | - Shiming Fan
- Hutchison MediPharma Limited, Building 4, 720 Cailun Road, Zhang-Jiang Hi-Tech Park, Shanghai 201203, China.
| | - Yongxin Ren
- Hutchison MediPharma Limited, Building 4, 720 Cailun Road, Zhang-Jiang Hi-Tech Park, Shanghai 201203, China.
| | - Weiguo Qing
- Hutchison MediPharma Limited, Building 4, 720 Cailun Road, Zhang-Jiang Hi-Tech Park, Shanghai 201203, China.
| | - Weiguo Su
- Hutchison MediPharma Limited, Building 4, 720 Cailun Road, Zhang-Jiang Hi-Tech Park, Shanghai 201203, China.
| |
Collapse
|
8
|
Daryaee F, Tonge PJ. Pharmacokinetic-pharmacodynamic models that incorporate drug-target binding kinetics. Curr Opin Chem Biol 2019; 50:120-127. [PMID: 31030171 DOI: 10.1016/j.cbpa.2019.03.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/08/2019] [Accepted: 03/09/2019] [Indexed: 01/30/2023]
Abstract
Pharmacokinetic/pharmacodynamic (PK/PD) models predict the effect time course resulting from a drug dose. In this review, we summarize the development of mechanistic PK/PD models that explicitly integrate the kinetics of drug-target interactions into predictions of drug activity. Such mechanistic models are expected to have several advantages over approaches in which concentration and effect are linked using variations of the Hill equation, and where preclinical data are often used as a starting point for modeling drug activity. Instead, explicit use of the full kinetic scheme for drug binding enables time-dependent changes in target occupancy to be calculated using the kinetics of drug-target interactions and drug PK, providing a more precise picture of target engagement and drug action in the non-equilibrium environment of the human body. The mechanistic PK/PD models also generate target vulnerability functions that link target occupancy and effect, and inform on the sensitivity of a target to engagement by a drug. Key factors such as the rate of target turnover can also be integrated into the modeling which, together with target vulnerability, provide additional information on the PK profile required to achieve the desired pharmacological effect and on the utility of kinetic selectivity in developing drugs for specific targets.
Collapse
Affiliation(s)
- Fereidoon Daryaee
- Center for Advanced Study of Drug Action, Department of Chemistry, New York, USA
| | - Peter J Tonge
- Center for Advanced Study of Drug Action, Department of Chemistry, New York, USA; Department of Radiology, Stony Brook University, Stony Brook, New York, USA.
| |
Collapse
|
9
|
Wu J, Zhang J, Jiang M, Zhang T, Wang Y, Wang Z, Miao Y, Wang Z, Li W. Comparison between NOD/SCID mice and BALB/c mice for patient-derived tumor xenografts model of non-small-cell lung cancer. Cancer Manag Res 2018; 10:6695-6703. [PMID: 30584364 PMCID: PMC6289205 DOI: 10.2147/cmar.s181272] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background Patient-derived tumor xenografts (PDX) are considered as a more reliable experiment model for screening chemotherapeutic drugs. However, the tumorigenic rate differs depending on mouse strains, which generates the experimental variability. Materials and methods In this study, we built PDX models of human non-small-cell lung cancer (NSCLC) in NOD/SCID mice in comparison with BALB/c mice. Results The result showed that the tumorigenesis rate of NOD/SCID mice (46.2%, 18/39) was higher than that of BALB/c mice (17.39%, 4/23). Latent times of tumorigenesis of NOD/SCID mice (41±18 days) were shorter than these of BALB/c mice (53±17 days). Times of tumorigenesis of NOD/SCID mice (85±25 days) were shorter than that of BALB/c mice (104±14 days). In addition, squamous carcinoma tissues were more likely to form tumors than adenocarcinoma tissues in NOD/SCID mice (P=0.008) and BALB/c mice (P=0.09). Also tumors could retain patients’ tumor characteristics in NOD/SCID mice and BALB/c mice xenograft models. Conclusion It is worth mentioning that the result of the drug experiment in the PDX models was consistent with the effect of clinical chemotherapy. As a result, NOD/SCID mice have advantages in a higher rate of tumorigenesis, shorter latent times of tumorigenesis and times of tumorigenesis over BALB/c mice in PDX models. It can provide a more reliable model of drug screening.
Collapse
Affiliation(s)
- Jianbin Wu
- Department of Cellular and Molecular Biology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, People's Republic of China,
| | - Juntao Zhang
- Department of Cellular and Molecular Biology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, People's Republic of China,
| | - Mei Jiang
- Department of Cellular and Molecular Biology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, People's Republic of China,
| | - Tianhui Zhang
- Tumor Surgery, Beijing Chest Hospital, Capital Medical University, Beijing, People's Republic of China,
| | - Yue Wang
- Department of Cellular and Molecular Biology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, People's Republic of China,
| | - Ziyu Wang
- Department of Cellular and Molecular Biology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, People's Republic of China,
| | - Yaodong Miao
- Department of Cellular and Molecular Biology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, People's Republic of China,
| | - Zitong Wang
- Tumor Surgery, Beijing Chest Hospital, Capital Medical University, Beijing, People's Republic of China,
| | - Weiying Li
- Department of Cellular and Molecular Biology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, People's Republic of China,
| |
Collapse
|
10
|
Pierrillas PB, Henin E, Ogier J, Kraus-Berthier L, Chenel M, Bouzom F, Tod M. Tumor Growth Inhibition Modelling Based on Receptor Occupancy and Biomarker Activity of a New Bcl-2 Inhibitor in Mice. J Pharmacol Exp Ther 2018; 367:414-424. [PMID: 30228112 DOI: 10.1124/jpet.118.251694] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 09/10/2018] [Indexed: 03/08/2025] Open
Abstract
The Bcl-2 inhibitor S 55746 ((S)-N-(4-hydroxyphenyl)-3-(6-(3-(morpholinomethyl)-1,2,3,4-tetrahydroisoquinoline-2-carbonyl)benzo[d][1,3]dioxol-5-yl)-N-phenyl-5,6,7,8-tetrahydroindolizine-1-carboxamide) is able to restore apoptosis functions impaired by tumorigenesis in mice. Data from pharmacokinetic (PK), biomarker, and tumor growth studies in a xenograft mouse model were considered for population modeling. The aim of the modeling exercise was to link the kinetics of the drug to the biomarker and tumor-size time profiles to better understand its dose-effect relationship. The PK, caspase kinetics, and tumor dynamics were successfully characterized by the proposed pharmacokinetic-pharmacodynamic model. The nonlinear plasma PK was best described by a two-compartment disposition model with both saturable absorption and elimination. Caspase was activated above the effective drug-concentration threshold (CTHRE ), at which near-maximal activity was reached. Increasing the dose did not increase the activation but better sustained it. Tumor growth followed a biphasic pattern, with caspase having an all-or-none inhibiting effect, consistent with the bistability property of the caspase pathway. For tumor eradication, the CTHRE in plasma was 2876 ng ml-1, and the relative caspase activity threshold (CaspTHRE) was 46.5. There was a strong relationship between the time spent above these thresholds and tumor growth inhibition. Tumor growth was inhibited by 50% when CaspTHRE was exceeded 13.8% of the time and when CTHRE was exceeded 8.1% of the time per dosing. This semimechanistic approach, based on experimental mice data and in vitro parameters, provides an interesting tool to quantify or simulate antitumor effects and, eventually, to plan phase 1 studies.
Collapse
Affiliation(s)
- Philippe B Pierrillas
- EMR 3738, Ciblage Thérapeutique en Oncologie, Faculté de Médecine et de Maïeutique Lyon-Sud Charles Mérieux, Université Claude Bernard Lyon 1, Oullins, France (P.P., E.H., M.T.); Pharmacie Hôpital de la Croix Rousse, Hospices Civils de Lyon, Lyon, France (M.T.); Centre de Pharmacocinétique et Métabolisme, Technologie Servier, Orléans, France (P.P., J.O., F.B.); Clinical Pharmacokinetics and Pharmacometrics Division, Servier, Paris, France (M.C.); and Institut de Recherches Internationales Servier, Oncology R&D Unit, Suresnes, France (L.K-B.)
| | - Emilie Henin
- EMR 3738, Ciblage Thérapeutique en Oncologie, Faculté de Médecine et de Maïeutique Lyon-Sud Charles Mérieux, Université Claude Bernard Lyon 1, Oullins, France (P.P., E.H., M.T.); Pharmacie Hôpital de la Croix Rousse, Hospices Civils de Lyon, Lyon, France (M.T.); Centre de Pharmacocinétique et Métabolisme, Technologie Servier, Orléans, France (P.P., J.O., F.B.); Clinical Pharmacokinetics and Pharmacometrics Division, Servier, Paris, France (M.C.); and Institut de Recherches Internationales Servier, Oncology R&D Unit, Suresnes, France (L.K-B.)
| | - Julien Ogier
- EMR 3738, Ciblage Thérapeutique en Oncologie, Faculté de Médecine et de Maïeutique Lyon-Sud Charles Mérieux, Université Claude Bernard Lyon 1, Oullins, France (P.P., E.H., M.T.); Pharmacie Hôpital de la Croix Rousse, Hospices Civils de Lyon, Lyon, France (M.T.); Centre de Pharmacocinétique et Métabolisme, Technologie Servier, Orléans, France (P.P., J.O., F.B.); Clinical Pharmacokinetics and Pharmacometrics Division, Servier, Paris, France (M.C.); and Institut de Recherches Internationales Servier, Oncology R&D Unit, Suresnes, France (L.K-B.)
| | - Laurence Kraus-Berthier
- EMR 3738, Ciblage Thérapeutique en Oncologie, Faculté de Médecine et de Maïeutique Lyon-Sud Charles Mérieux, Université Claude Bernard Lyon 1, Oullins, France (P.P., E.H., M.T.); Pharmacie Hôpital de la Croix Rousse, Hospices Civils de Lyon, Lyon, France (M.T.); Centre de Pharmacocinétique et Métabolisme, Technologie Servier, Orléans, France (P.P., J.O., F.B.); Clinical Pharmacokinetics and Pharmacometrics Division, Servier, Paris, France (M.C.); and Institut de Recherches Internationales Servier, Oncology R&D Unit, Suresnes, France (L.K-B.)
| | - Marylore Chenel
- EMR 3738, Ciblage Thérapeutique en Oncologie, Faculté de Médecine et de Maïeutique Lyon-Sud Charles Mérieux, Université Claude Bernard Lyon 1, Oullins, France (P.P., E.H., M.T.); Pharmacie Hôpital de la Croix Rousse, Hospices Civils de Lyon, Lyon, France (M.T.); Centre de Pharmacocinétique et Métabolisme, Technologie Servier, Orléans, France (P.P., J.O., F.B.); Clinical Pharmacokinetics and Pharmacometrics Division, Servier, Paris, France (M.C.); and Institut de Recherches Internationales Servier, Oncology R&D Unit, Suresnes, France (L.K-B.)
| | - François Bouzom
- EMR 3738, Ciblage Thérapeutique en Oncologie, Faculté de Médecine et de Maïeutique Lyon-Sud Charles Mérieux, Université Claude Bernard Lyon 1, Oullins, France (P.P., E.H., M.T.); Pharmacie Hôpital de la Croix Rousse, Hospices Civils de Lyon, Lyon, France (M.T.); Centre de Pharmacocinétique et Métabolisme, Technologie Servier, Orléans, France (P.P., J.O., F.B.); Clinical Pharmacokinetics and Pharmacometrics Division, Servier, Paris, France (M.C.); and Institut de Recherches Internationales Servier, Oncology R&D Unit, Suresnes, France (L.K-B.)
| | - Michel Tod
- EMR 3738, Ciblage Thérapeutique en Oncologie, Faculté de Médecine et de Maïeutique Lyon-Sud Charles Mérieux, Université Claude Bernard Lyon 1, Oullins, France (P.P., E.H., M.T.); Pharmacie Hôpital de la Croix Rousse, Hospices Civils de Lyon, Lyon, France (M.T.); Centre de Pharmacocinétique et Métabolisme, Technologie Servier, Orléans, France (P.P., J.O., F.B.); Clinical Pharmacokinetics and Pharmacometrics Division, Servier, Paris, France (M.C.); and Institut de Recherches Internationales Servier, Oncology R&D Unit, Suresnes, France (L.K-B.)
| |
Collapse
|
11
|
Morcos PN, Nueesch E, Jaminion F, Guerini E, Hsu JC, Bordogna W, Balas B, Mercier F. Exposure-response analysis of alectinib in crizotinib-resistant ALK-positive non-small cell lung cancer. Cancer Chemother Pharmacol 2018; 82:129-138. [PMID: 29748847 PMCID: PMC6010493 DOI: 10.1007/s00280-018-3597-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 05/01/2018] [Indexed: 12/22/2022]
Abstract
PURPOSE Alectinib is a selective and potent anaplastic lymphoma kinase (ALK) inhibitor that is active in the central nervous system (CNS). Alectinib demonstrated robust efficacy in a pooled analysis of two single-arm, open-label phase II studies (NP28673, NCT01801111; NP28761, NCT01871805) in crizotinib-resistant ALK-positive non-small-cell lung cancer (NSCLC): median overall survival (OS) 29.1 months (95% confidence interval [CI]: 21.3-39.0) for alectinib 600 mg twice daily (BID). We investigated exposure-response relationships from final pooled phase II OS and safety data to assess alectinib dose selection. METHODS A semi-parametric Cox proportional hazards model analyzed relationships between individual median observed steady-state trough concentrations (Ctrough,ss) for combined exposure of alectinib and its major metabolite (M4), baseline covariates (demographics and disease characteristics) and OS. Univariate logistic regression analysis analyzed relationships between Ctrough,ss and incidence of adverse events (AEs: serious and Grade ≥ 3). RESULTS Overall, 92% of patients (n = 207/225) had Ctrough,ss data and were included in the analysis. No statistically significant relationship was found between Ctrough,ss and OS following alectinib treatment. The only baseline covariates that statistically influenced OS were baseline tumor size and prior crizotinib treatment duration. Larger baseline tumor size and shorter prior crizotinib treatment were both associated with shorter OS. Logistic regression confirmed no significant relationship between Ctrough,ss and AEs. CONCLUSION Alectinib 600 mg BID provides systemic exposures at plateau of response for OS while maintaining a well-tolerated safety profile. This analysis confirms alectinib 600 mg BID as the recommended global dose for patients with crizotinib-resistant ALK-positive NSCLC.
Collapse
Affiliation(s)
| | | | | | | | - Joy C Hsu
- Roche Innovation Center New York, New York, NY, USA
| | | | | | | |
Collapse
|
12
|
Xu Y, Zhang F, Pan X, Wang G, Zhu L, Zhang J, Wen D, Lu S. Xenograft tumors derived from malignant pleural effusion of the patients with non-small-cell lung cancer as models to explore drug resistance. Cancer Commun (Lond) 2018; 38:19. [PMID: 29764505 PMCID: PMC5993147 DOI: 10.1186/s40880-018-0284-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 03/08/2018] [Indexed: 12/25/2022] Open
Abstract
Background Non-small cell lung cancer (NSCLC) patients with epidermal growth factor receptor (EGFR) mutations or anaplastic lymphoma kinase (ALK) fusions show dramatic responses to specific tyrosine kinase inhibitors (TKIs); however, after 10–12 months, secondary mutations arise that confer resistance. We generated a murine xenograft model using patient-derived NSCLC cells isolated from the pleural fluid of two patients with NSCLC to investigate the mechanisms of resistance against the ALK- and EGFR-targeted TKIs crizotinib and osimertinib, respectively. Methods Genotypes of patient biopsies and xenograft tumors were determined by whole exome sequencing (WES), and patients and xenograft-bearing mice received targeted treatment (crizotinib or osimertinib) accordingly. Xenograft mice were also treated for prolonged periods to identify whether the development of drug resistance and/or treatment responses were associated with tumor size. Finally, the pathology of patients biopsies and xenograft tumors were compared histologically. Results The histological characteristics and chemotherapy responses of xenograft tumors were similar to the actual patients. WES showed that the genotypes of the xenograft and patient tumors were similar (an echinoderm microtubule-associated protein-like 4-ALK (EML4–ALK) gene fusion (patient/xenograft: CTC15035EML4–ALK) and EGFR L858R and T790M mutations (patient/xenograft: CTC15063EGFR L858R, T790M)). After continuous crizotinib or osimertinib treatment, WES data suggested that acquired ALK E1210K mutation conferred crizotinib resistance in the CTC15035EML4–ALK xenograft, while decreased frequencies of EGFR L858R and T790M mutations plus the appearance of v-RAF murine sarcoma viral oncogene homolog B (BRAF) G7V mutations and phosphatidylinositol-4-phosphate 3-kinase catalytic subunit type 2 alpha (PIK3C2A) A86fs frame shift mutations led to osimertinib resistance in the CTC15063EGFR L858R, T790M xenografts. Conclusions We successfully developed a new method of generating drug resistance xenograft models from liquid biopsies using microfluidic technology, which might be a useful tool to investigate the mechanisms of drug resistance in NSCLC. Electronic supplementary material The online version of this article (10.1186/s40880-018-0284-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yunhua Xu
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 Huaihai West Road, Shanghai, 200030, China
| | - Feifei Zhang
- Shanghai LIDE Biotech Co., Ltd, 887 Zuchongzhi Rd, Pudong, Shanghai, 201203, China
| | - Xiaoqing Pan
- Shanghai LIDE Biotech Co., Ltd, 887 Zuchongzhi Rd, Pudong, Shanghai, 201203, China
| | - Guan Wang
- GenomiCare Biotechnology (Shanghai) Co., Ltd., Shanghai, 200233, China
| | - Lei Zhu
- Department of Pathology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Jie Zhang
- Department of Pathology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Danyi Wen
- Shanghai LIDE Biotech Co., Ltd, 887 Zuchongzhi Rd, Pudong, Shanghai, 201203, China.
| | - Shun Lu
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 Huaihai West Road, Shanghai, 200030, China.
| |
Collapse
|
13
|
Mathematical modeling identifies optimum lapatinib dosing schedules for the treatment of glioblastoma patients. PLoS Comput Biol 2018; 14:e1005924. [PMID: 29293494 PMCID: PMC5766249 DOI: 10.1371/journal.pcbi.1005924] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 01/12/2018] [Accepted: 12/12/2017] [Indexed: 12/15/2022] Open
Abstract
Human primary glioblastomas (GBM) often harbor mutations within the epidermal growth factor receptor (EGFR). Treatment of EGFR-mutant GBM cell lines with the EGFR/HER2 tyrosine kinase inhibitor lapatinib can effectively induce cell death in these models. However, EGFR inhibitors have shown little efficacy in the clinic, partly because of inappropriate dosing. Here, we developed a computational approach to model the in vitro cellular dynamics of the EGFR-mutant cell line SF268 in response to different lapatinib concentrations and dosing schedules. We then used this approach to identify an effective treatment strategy within the clinical toxicity limits of lapatinib, and developed a partial differential equation modeling approach to study the in vivo GBM treatment response by taking into account the heterogeneous and diffusive nature of the disease. Despite the inability of lapatinib to induce tumor regressions with a continuous daily schedule, our modeling approach consistently predicts that continuous dosing remains the best clinically feasible strategy for slowing down tumor growth and lowering overall tumor burden, compared to pulsatile schedules currently known to be tolerated, even when considering drug resistance, reduced lapatinib tumor concentrations due to the blood brain barrier, and the phenotypic switch from proliferative to migratory cell phenotypes that occurs in hypoxic microenvironments. Our mathematical modeling and statistical analysis platform provides a rational method for comparing treatment schedules in search for optimal dosing strategies for glioblastoma and other cancer types.
Collapse
|
14
|
Wong H, Bohnert T, Damian-Iordache V, Gibson C, Hsu CP, Krishnatry AS, Liederer BM, Lin J, Lu Q, Mettetal JT, Mudra DR, Nijsen MJ, Schroeder P, Schuck E, Suryawanshi S, Trapa P, Tsai A, Wang H, Wu F. Translational pharmacokinetic-pharmacodynamic analysis in the pharmaceutical industry: an IQ Consortium PK-PD Discussion Group perspective. Drug Discov Today 2017; 22:1447-1459. [DOI: 10.1016/j.drudis.2017.04.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 04/03/2017] [Accepted: 04/25/2017] [Indexed: 02/06/2023]
|
15
|
Lucas CJ, Martin JH. Pharmacokinetic-Guided Dosing of New Oral Cancer Agents. J Clin Pharmacol 2017; 57 Suppl 10:S78-S98. [DOI: 10.1002/jcph.937] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 04/10/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Catherine J. Lucas
- Discipline of Clinical Pharmacology, School of Medicine and Public Health; University of Newcastle; New South Wales Australia
| | - Jennifer H. Martin
- Discipline of Clinical Pharmacology, School of Medicine and Public Health; University of Newcastle; New South Wales Australia
| |
Collapse
|
16
|
Carrara L, Lavezzi SM, Borella E, De Nicolao G, Magni P, Poggesi I. Current mathematical models for cancer drug discovery. Expert Opin Drug Discov 2017; 12:785-799. [PMID: 28595492 DOI: 10.1080/17460441.2017.1340271] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
INTRODUCTION Pharmacometric models represent the most comprehensive approaches for extracting, summarizing and integrating information obtained in the often sparse, limited, and less-than-optimally designed experiments performed in the early phases of oncology drug discovery. Whilst empirical methodologies may be enough for screening and ranking candidate drugs, modeling approaches are needed for optimizing and making economically viable the learn-confirm cycles within an oncology research program and anticipating the dose regimens to be investigated in the subsequent clinical development. Areas covered: Papers appearing in the literature of approximately the last decade reporting modeling approaches applicable to anticancer drug discovery have been listed and commented. Papers were selected based on the interest in the proposed methodology or in its application. Expert opinion: The number of modeling approaches used in the discovery of anticancer drugs is consistently increasing and new models are developed based on the current directions of research of new candidate drugs. These approaches have contributed to a better understanding of new oncological targets and have allowed for the exploitation of the relatively sparse information generated by preclinical experiments. In addition, they are used in translational approaches for guiding and supporting the choice of dosing regimens in early clinical development.
Collapse
Affiliation(s)
- Letizia Carrara
- a Dipartimento di Ingegneria Industriale e dell'Informazione , Università degli Studi di Pavia , Pavia , Italy
| | - Silvia Maria Lavezzi
- a Dipartimento di Ingegneria Industriale e dell'Informazione , Università degli Studi di Pavia , Pavia , Italy
| | - Elisa Borella
- a Dipartimento di Ingegneria Industriale e dell'Informazione , Università degli Studi di Pavia , Pavia , Italy
| | - Giuseppe De Nicolao
- a Dipartimento di Ingegneria Industriale e dell'Informazione , Università degli Studi di Pavia , Pavia , Italy
| | - Paolo Magni
- a Dipartimento di Ingegneria Industriale e dell'Informazione , Università degli Studi di Pavia , Pavia , Italy
| | - Italo Poggesi
- b Global Clinical Pharmacology , Janssen Research and Development , Cologno Monzese , Italy
| |
Collapse
|
17
|
Rossi A, Sacco PC, Santabarbara G, Sgambato A, Casaluce F, Palazzolo G, Maione P, Gridelli C. Developments in pharmacotherapy for treating metastatic non-small cell lung cancer. Expert Opin Pharmacother 2017; 18:151-163. [PMID: 28067062 DOI: 10.1080/14656566.2017.1280460] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Most patients with non-small cell lung (NSCLC), including squamous cell carcinoma, adenocarcinoma and large cell carcinoma, have advanced disease at diagnosis and systemic therapy is the standard-of-care. About 20% of Caucasian patients are affected by an oncogene-addicted advanced NSCLC for which correspondent inhibitors are available. Areas covered: The main state-of-the-art synthetic anticancer drugs in the groups of chemotherapeutics, epidermal growth factor receptor and anaplastic lymphoma kinase tyrosine kinase inhibitors for NSCLC treatment, are reviewed and discussed from phase III randomized practice-changing trials onwards. A structured search of bibliographic databases for peer-reviewed research literature and of main meetings using a focused review question was undertaken. Expert opinion: The survival of NSCLC patients is increasing, regardless of the presence or not of a specific target, due to the availability of new generation drugs. The continuous deep knowledge of the mechanisms of NSCLC development and the constant research into new drugs should lead to the discovery of new potential targets and the synthesis of corresponding inhibitors to improve the outcomes of each subgroup of patients in order to control the disease in a constantly growing percentage of patients.
Collapse
Affiliation(s)
- Antonio Rossi
- a Division of Medical Oncology , 'S.G. Moscati' Hospital , Avellino , Italy
| | | | | | - Assunta Sgambato
- a Division of Medical Oncology , 'S.G. Moscati' Hospital , Avellino , Italy
| | - Francesca Casaluce
- a Division of Medical Oncology , 'S.G. Moscati' Hospital , Avellino , Italy
| | | | - Paolo Maione
- a Division of Medical Oncology , 'S.G. Moscati' Hospital , Avellino , Italy
| | - Cesare Gridelli
- a Division of Medical Oncology , 'S.G. Moscati' Hospital , Avellino , Italy
| |
Collapse
|
18
|
Poon CC, Kelly JJ. Development of crizotinib, a rationally designed tyrosine kinase inhibitor for non-small cell lung cancer. Int J Cancer 2017; 140:1945-1954. [PMID: 27874172 DOI: 10.1002/ijc.30533] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 10/29/2016] [Accepted: 11/16/2016] [Indexed: 12/29/2022]
Abstract
Non-small cell lung cancer (NSCLC) is the number one cause of global mortality. Despite aggressive treatment, the prognosis is dismal. Patients with advanced NSCLC have a median survival of 4 months from the time of diagnosis. Fortunately, molecularly based approaches to drug discovery have yielded a tyrosine kinase inhibitor, crizotinib, which significantly prolongs median progression-free survival in a subset of patients. Although initial clinical trial results demonstrate crizotinib has a promising role to play in NSCLC treatment, development of resistance leaves much to be elucidated about how to effectively combat this deadly disease. In this review, we follow the discovery and development of crizotinib from bench to bedside and provide an example of successful bottom-up drug design. Then, we explore the clinical trial results that fast-tracked its eventual use as a frontline therapy for sensitive NSCLC patients and the development of resistance. Lastly, we discuss the potential for future uses of crizotinib both within and beyond NSCLC.
Collapse
Affiliation(s)
- Candice C Poon
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
| | - John J Kelly
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
19
|
Yamazaki S, Spilker ME, Vicini P. Translational modeling and simulation approaches for molecularly targeted small molecule anticancer agents from bench to bedside. Expert Opin Drug Metab Toxicol 2016; 12:253-65. [PMID: 26799750 DOI: 10.1517/17425255.2016.1141895] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Recent advances in molecular biology have enabled personalized cancer therapies with molecularly targeted agents (MTAs), which offer a promising future for cancer therapy. Dynamic modeling and simulation (M&S) is a powerful mathematical approach linking drug exposures to pharmacological responses, providing a quantitative assessment of in vivo drug potency. Accordingly, a growing emphasis is being placed upon M&S to quantitatively understand therapeutic exposure-response relationships of MTAs in nonclinical models. AREAS COVERED An overview of M&S approaches for MTAs in nonclinical models is presented with discussion about mechanistic extrapolation of antitumor efficacy from bench to bedside. Emphasis is placed upon recent advances in M&S approaches linking drug exposures, biomarker responses (e.g. target modulation) and pharmacological outcomes (e.g. antitumor efficacy). EXPERT OPINION For successful personalized cancer therapies with MTAs, it is critical to mechanistically and quantitatively understand their exposure-response relationships in nonclinical models, and to logically and properly apply such knowledge to the clinic. Particularly, M&S approaches to predict pharmacologically active concentrations of MTAs in patients based upon nonclinical data would be highly valuable in guiding the design and execution of clinical trials. Proactive approaches to understand their exposure-response relationships could substantially increase probability of achieving a positive proof-of-concept in the clinic.
Collapse
Affiliation(s)
- Shinji Yamazaki
- a Pharmacokinetics, Dynamics & Metabolism , Pfizer Worldwide Research & Development , San Diego , CA , USA
| | - Mary E Spilker
- a Pharmacokinetics, Dynamics & Metabolism , Pfizer Worldwide Research & Development , San Diego , CA , USA
| | - Paolo Vicini
- a Pharmacokinetics, Dynamics & Metabolism , Pfizer Worldwide Research & Development , San Diego , CA , USA
| |
Collapse
|
20
|
Ou SHI, Govindan R, Eaton KD, Otterson GA, Gutierrez ME, Mita AC, Argiris A, Brega NM, Usari T, Tan W, Ho SN, Robert F. Phase I Results from a Study of Crizotinib in Combination with Erlotinib in Patients with Advanced Nonsquamous Non-Small Cell Lung Cancer. J Thorac Oncol 2016; 12:145-151. [PMID: 27697581 DOI: 10.1016/j.jtho.2016.09.131] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 09/16/2016] [Accepted: 09/22/2016] [Indexed: 11/26/2022]
Abstract
INTRODUCTION This phase I trial was conducted to determine the safety, maximum tolerated dose (MTD)/recommended phase II dose, and efficacy of crizotinib plus erlotinib in patients with advanced NSCLC. METHODS Patients with NSCLC and an Eastern Cooperative Oncology Group performance status of 0 to 2 after failure of one or two prior chemotherapy regimens were eligible. Erlotinib, 100 mg, was given continuously once daily starting between day -14 and -7; crizotinib, 200 mg twice daily (dose level 1) or 150 mg twice daily (dose level -1), was added continuously beginning on day 1 of treatment cycle 1. Potential pharmacokinetic interactions between crizotinib and erlotinib were evaluated. RESULTS Twenty-seven patients received treatment; 26 received crizotinib plus erlotinib. Frequent adverse events were diarrhea, rash, decreased appetite, and fatigue. Dose-limiting toxicities were dehydration, diarrhea, dry eye, dysphagia, dyspepsia, esophagitis and vomiting. The MTD was crizotinib, 150 mg twice daily, with erlotinib, 100 mg once daily. Crizotinib increased the erlotinib area under the concentration-time curve 1.5-fold (dose level -1) and 1.8-fold (dose level 1). The plasma level of crizotinib appeared to be unaffected by coadministration of erlotinib. Two patients whose tumors harbored activating EGFR mutations achieved confirmed partial responses, one at each crizotinib dose level. CONCLUSIONS The MTD of the combination of crizotinib and erlotinib in patients with advanced NSCLC was crizotinib, 150 mg twice daily, with erlotinib, 100 mg once daily, which is less than the approved dose of either agent. The phase II portion of the study was not initiated.
Collapse
Affiliation(s)
- Sai-Hong Ignatius Ou
- Chao Family Comprehensive Cancer Center, University of California at Irvine Medical Center, Orange, California.
| | | | - Keith D Eaton
- Seattle Cancer Care Alliance and the University of Washington, Seattle, Washington
| | - Gregory A Otterson
- Ohio State University Comprehensive Cancer Center and the James Cancer Hospital, Columbus, Ohio
| | - Martin E Gutierrez
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, New Jersey
| | - Alain C Mita
- Cancer Therapy and Research Center, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Athanassios Argiris
- Cancer Therapy and Research Center, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | | | | | | | | | - Francisco Robert
- Birmingham Veterans Administration Hospital, Birmingham, Alabama; University of Alabama-Birmingham Comprehensive Cancer Center, Birmingham, Alabama
| |
Collapse
|
21
|
Wong H, Gould SE. Translational pharmacokinetic/pharmacodynamic analysis in cancer pharmacology: a tool to maximize the value of antitumor efficacy from tumor bearing mice. DRUG DISCOVERY TODAY. TECHNOLOGIES 2016; 21-22:51-56. [PMID: 27978988 DOI: 10.1016/j.ddtec.2016.08.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 08/17/2016] [Accepted: 08/18/2016] [Indexed: 06/06/2023]
Abstract
Translational pharmacokinetic/pharmacodynamic (PK/PD) analysis is becoming an increasingly important tool for the identification and selection of new anticancer agents. There are two important elements of effectively using PK/PD analysis to translate preclinical antitumor efficacy from tumor bearing mice (xenografts and allografts) to cancer patients. These two sometimes overlapping elements are termed translation 'WITHIN' and 'ACROSS' species. Translating 'WITHIN' species refers to the quantitative characterization of drug action and disease behavior within tumor bearing mice using PK/PD modeling in order to use this information to make predictions of drug response in humans. Translating 'ACROSS' species refers to use of PK/PD modeling to quantify species similarities and differences in drug response in order to understand the clinical relevance of preclinical efficacy data.
Collapse
Affiliation(s)
- Harvey Wong
- Faculty of Pharmaceutical Sciences, The University of British Columbia, 5505-2405 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada.
| | | |
Collapse
|
22
|
Watanabe Miyano S, Yamamoto Y, Kodama K, Miyajima Y, Mikamoto M, Nakagawa T, Kuramochi H, Funasaka S, Nagao S, Sugi NH, Okamoto K, Minoshima Y, Nakatani Y, Karoji Y, Ohashi I, Yamane Y, Okada T, Matsushima T, Matsui J, Iwata M, Uenaka T, Tsuruoka A. E7090, a Novel Selective Inhibitor of Fibroblast Growth Factor Receptors, Displays Potent Antitumor Activity and Prolongs Survival in Preclinical Models. Mol Cancer Ther 2016; 15:2630-2639. [DOI: 10.1158/1535-7163.mct-16-0261] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 08/04/2016] [Indexed: 11/16/2022]
|
23
|
Fujiwara Y, Hamada A, Mizugaki H, Aikawa H, Hata T, Horinouchi H, Kanda S, Goto Y, Itahashi K, Nokihara H, Yamamoto N, Ohe Y. Pharmacokinetic profiles of significant adverse events with crizotinib in Japanese patients with ABCB1 polymorphism. Cancer Sci 2016; 107:1117-23. [PMID: 27270784 PMCID: PMC4982581 DOI: 10.1111/cas.12983] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 05/24/2016] [Accepted: 06/06/2016] [Indexed: 01/11/2023] Open
Abstract
Crizotinib is a standard treatment for advanced ALK-positive non-small-cell lung cancer (NSCLC). We undertook this study to investigate the pharmacokinetics of crizotinib and clinical and pharmacogenomic factors that may increase the risk of adverse events (AEs). We defined clinically significant AEs as grade 4 hematological toxicity, grade ≥3 non-hematological toxicity, and any grade of interstitial lung disease. Eight subjects with ALK-positive NSCLC scheduled to receive crizotinib 250 mg twice daily were studied. Six patients were female and two were male, and most of the patients had low body weight with a median body weight of 46.8 kg (range, 42.4-61.0 kg). All patients developed AEs, five developing six clinically significant AEs. Six patients required dose reduction. In pharmacokinetic analysis, blood samples were obtained on days 1 and 15. The mean area under the plasma concentration-time curve from 0-12 h (AUC0-12 ) on day 15 was significantly increased in patients with clinically significant AEs (n = 5) compared with those without (n = 3) (P = 0.04). Genetic polymorphisms of ABCB1 were analyzed. One patient with the ABCB1 1236TT-2677TT-3435TT genotype was an outlier, with an AUC0-12 and peak concentrations on day 15 of 2.84× and 2.61× the mean, respectively, compared with those with other genotypes. Our results suggest that some Japanese NSCLC patients treated with crizotinib developed clinically significant toxicities that were related to altered pharmacokinetics parameters due to genotype and body weight factors.
Collapse
Affiliation(s)
- Yutaka Fujiwara
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan.,Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| | - Akinobu Hamada
- Division of Clinical Pharmacology and Translational Research, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Tokyo, Japan
| | - Hidenori Mizugaki
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan.,First Department of Medicine, Hokkaido University School of Medicine, Sapporo, Japan
| | - Hiroaki Aikawa
- Division of Clinical Pharmacology and Translational Research, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Tokyo, Japan
| | - Toshiyuki Hata
- Division of Clinical Pharmacology and Translational Research, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Tokyo, Japan
| | - Hidehito Horinouchi
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Shintaro Kanda
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yasushi Goto
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Kota Itahashi
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Hiroshi Nokihara
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Noboru Yamamoto
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan.,Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| | - Yuichiro Ohe
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
24
|
Zhang YC, Zhou Q, Wu YL. Efficacy of crizotinib in first-line treatment of adults with ALK-positive advanced NSCLC. Expert Opin Pharmacother 2016; 17:1693-701. [PMID: 27359268 DOI: 10.1080/14656566.2016.1208171] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION The treatment of advanced non-small cell lung cancer (NSCLC) has evolved from palliative cytotoxic chemotherapy to precise medicine based on genetic alternations over the last decade. Anaplastic lymphoma kinase (ALK) rearrangement characterizes a molecular subset of NSCLC with an impressive response to crizotinib. AREAS COVERED To analyze the efficacy of crizotinib in first-line treatment of adults with advanced ALK-positive NSCLC, updated data on development and recent advances of first-line crizotinib in this subset population are reviewed. EXPERT OPINION To date, crizotinib should be established as a standard of care in previously untreated advanced NSCLC with ALK-rearrangement. However, the efficacy of first-line crizotinib is limited by acquired resistance. Second generation ALK inhibitors have demonstrated clinical activity in both crizotinib-refractory and crizotinib naïve setting. How to maximize first-line benefit for advanced ALK-positive NSCLC remains challenging. Combinational strategy, advances in companion diagnostics and optimization of ALK inhibitors might contribute to improve outcome in this subset of patients in future.
Collapse
Affiliation(s)
- Yi-Chen Zhang
- a Graduate School , Southern Medical University , Guangzhou , PR China.,b Guangdong Lung Cancer Institute , Guangdong General Hospital and Guangdong Academy of Medical Sciences , Guangzhou , PR China
| | - Qing Zhou
- b Guangdong Lung Cancer Institute , Guangdong General Hospital and Guangdong Academy of Medical Sciences , Guangzhou , PR China
| | - Yi-Long Wu
- b Guangdong Lung Cancer Institute , Guangdong General Hospital and Guangdong Academy of Medical Sciences , Guangzhou , PR China
| |
Collapse
|
25
|
Discovery bioanalysis and in vivo pharmacology as an integrated process: a case study in oncology drug discovery. Bioanalysis 2016; 8:1481-98. [DOI: 10.4155/bio-2016-0062] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background: A bioanalytical team dedicated to in vivo pharmacology was set up to accelerate the selection and characterization of compounds to be evaluated in animal models in oncology. Results: A DBS-based serial microsampling procedure was optimized from sample collection to extraction to obtain a generic procedure. UHPLC–high-resolution mass spectrometer configuration allowed for fast quantitative and qualitative analysis. Using an optimized lead compound, we show how bioanalysis supported in vivo pharmacology by generating blood and tumor exposure, drug monitoring and PK/PD data. Conclusion: This process provided unique opportunities for the characterization of drug properties, selection and assessment of compounds in animal models and to support and expedite proof-of-concept studies in oncology.
Collapse
|
26
|
Sachs JR, Mayawala K, Gadamsetty S, Kang SP, de Alwis DP. Optimal Dosing for Targeted Therapies in Oncology: Drug Development Cases Leading by Example. Clin Cancer Res 2015; 22:1318-24. [DOI: 10.1158/1078-0432.ccr-15-1295] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 10/05/2015] [Indexed: 11/16/2022]
|
27
|
Qiu F, Gu Y, Wang T, Gao Y, Li X, Gao X, Cheng S. Quantification and pharmacokinetics of crizotinib in rats by liquid chromatography-tandem mass spectrometry. Biomed Chromatogr 2015; 30:962-8. [DOI: 10.1002/bmc.3636] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 10/06/2015] [Accepted: 10/12/2015] [Indexed: 12/12/2022]
Affiliation(s)
- Feng Qiu
- Beijing Key Laboratory of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine; Capital Medical University; Beijing 100069 China
| | - Yanan Gu
- Department of Biochemistry and Molecular Biology; Capital Medical University; Beijing 100069 China
- Beijing Key Laboratory of Cancer and Metastasis Research; Capital Medical University; Beijing 100069 China
| | - Tingting Wang
- Beijing Key Laboratory of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine; Capital Medical University; Beijing 100069 China
| | - Yingying Gao
- Beijing Key Laboratory of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine; Capital Medical University; Beijing 100069 China
| | - Xiao Li
- Beijing Key Laboratory of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine; Capital Medical University; Beijing 100069 China
| | - Xiangyu Gao
- Beijing Cancer Hospital and Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Surgery; Peking University Cancer Hospital and Institute; Beijing 100142 China
| | - Shan Cheng
- Department of Biochemistry and Molecular Biology; Capital Medical University; Beijing 100069 China
- Beijing Key Laboratory of Cancer and Metastasis Research; Capital Medical University; Beijing 100069 China
| |
Collapse
|
28
|
Loong HH, Mok K, Leung LKS, Mok TSK. Crizotinib in the management of advanced-stage non-small-cell lung cancer. Future Oncol 2015; 11:735-45. [PMID: 25757678 DOI: 10.2217/fon.14.314] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
ABSTRACT Rearrangement of ALK gene has been identified as exerting a potent transforming effect as driver oncogene in patients with non-small-cell lung cancer (NSCLC). Crizotinib is a small-molecule oral inhibitor of ALK, c-Met/HGF receptor and ROS1 receptor kinases. Its efficacy in ALK-rearranged NSCLC has been established. Crizotinib's effect on ROS1 receptor kinases and c-Met with relevance to NSCLC is also actively being explored. Resistance mechanisms such as secondary gatekeeper mutations in ALK gene and activation of other oncogenes have been identified to confer acquired resistance to crizotinib. This article reviews the pharmacological properties of crizotinib, preclinical and clinical results that led to its approval in ALK-positive NSCLC and current directions of clinical research in overcoming crizotinib resistance.
Collapse
Affiliation(s)
- Herbert H Loong
- Department of Clinical Oncology, State Key Laboratory in Oncology in South China, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | | | | | | |
Collapse
|
29
|
Sardu ML, Poggesi I, De Nicolao G. Biomarker- versus drug-driven tumor growth inhibition models: an equivalence analysis. J Pharmacokinet Pharmacodyn 2015. [PMID: 26209955 DOI: 10.1007/s10928-015-9427-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The mathematical modeling of tumor xenograft experiments following the dosing of antitumor drugs has received much attention in the last decade. Biomarker data can further provide useful insights on the pathological processes and be used for translational purposes in the early clinical development. Therefore, it is of particular interest the development of integrated pharmacokinetic-pharmacodynamic (PK-PD) models encompassing drug, biomarker and tumor-size data. This paper investigates the reciprocal consistency of three types of models: drug-to-tumor, such as established drug-driven tumor growth inhibition (TGI) models, drug-to-biomarker, e.g. indirect response models, and biomarker-to-tumor, e.g. the more recent biomarker-driven TGI models. In particular, this paper derives a mathematical relationship that guarantees the steady-state equivalence of the cascade of drug-to-biomarker and biomarker-to-tumor models with a drug-to-tumor TGI model. Using the Simeoni TGI model as a reference, conditions for steady-state equivalence are worked out and used to derive a new biomarker-driven model. Simulated and real data are used to show that in realistic cases the steady-state equivalence extends also to transient responses. The possibility of predicting the drug-to-tumor potency of a new candidate drug based only on biomarker response is discussed.
Collapse
Affiliation(s)
- Maria Luisa Sardu
- Dipartimento di Ingegneria Industriale e dell'Informazione, Università di Pavia, Via Ferrata 1, 27100, Pavia, Italy.
| | - Italo Poggesi
- Clinical Pharmacology & Pharmacometrics, Janssen Research & Development, 2340, Beerse, Belgium
| | - Giuseppe De Nicolao
- Dipartimento di Ingegneria Industriale e dell'Informazione, Università di Pavia, Via Ferrata 1, 27100, Pavia, Italy
| |
Collapse
|
30
|
Hamilton G, Rath B, Burghuber O. Pharmacokinetics of crizotinib in NSCLC patients. Expert Opin Drug Metab Toxicol 2015; 11:835-42. [PMID: 25732197 DOI: 10.1517/17425255.2015.1021685] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
INTRODUCTION For a subpopulation of NSCLC patients genetic rearrangement of the anaplastic lymphoma kinase (ALK) was found as driver mutation, which can be targeted by the selective inhibitor crizotinib. AREAS COVERED This article presents an overview of the clinical studies that provided the characterization of the pharmacokinetic parameters for the administration of crizotinib to cancer patients and the factors influencing the clinical profiles of this drug. EXPERT OPINION Crizotinib is administered orally as a capsule and clinical studies indicated 250 mg crizotinib BID continuously as the maximal tolerated dose in cancer patients. Bioavailability is ∼ 40% and pharmacokinetic parameters are influenced by food only to a minor degree. This dose of the drug corresponds to a significant inhibition of the mutated ALK, retards tumor growth and achieves clinical responses in the majority of patients. Crizotinib lactam is the single metabolite with minor inhibitory activity for the ALK fusion protein. Metabolization is executed mainly by CYP3A4/5 and is modulated by other drugs interacting with this cytochrome oxidase. Despite the one-fits-all approach in administration of crizotinib at a fixed dose the pharmacokinetic parameters indicate a stable steady state upon continuous administration, which achieves sufficient inhibition of the ALK drug target.
Collapse
Affiliation(s)
- Gerhard Hamilton
- Ludwig Boltzmann Cluster of Translational Oncology , 1090 Vienna , Austria +43 1 40400 66270 ; +43 1 40400 66270 ;
| | | | | |
Collapse
|
31
|
Lee D, Sung ES, Ahn JH, An S, Huh J, You WK. Development of antibody-based c-Met inhibitors for targeted cancer therapy. Immunotargets Ther 2015; 4:35-44. [PMID: 27471710 PMCID: PMC4918247 DOI: 10.2147/itt.s37409] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Signaling pathways mediated by receptor tyrosine kinases (RTKs) and their ligands play important roles in the development and progression of human cancers, which makes RTK-mediated signaling pathways promising therapeutic targets in the treatment of cancer. Compared with small-molecule compounds, antibody-based therapeutics can more specifically recognize and bind to ligands and RTKs. Several antibody inhibitors of RTK-mediated signaling pathways, such as human epidermal growth factor receptor 2, vascular endothelial growth factor, epidermal growth factor receptor or vascular endothelial growth factor receptor 2, have been developed and are widely used to treat cancer patients. However, since the therapeutic options are still limited in terms of therapeutic efficacy and types of cancers that can be treated, efforts are being made to identify and evaluate novel RTK-mediated signaling pathways as targets for more efficacious cancer treatment. The hepatocyte growth factor/c-Met signaling pathway has come into the spotlight as a promising target for development of potent cancer therapeutic agents. Multiple antibody-based therapeutics targeting hepatocyte growth factor or c-Met are currently in preclinical or clinical development. This review focuses on the development of inhibitors of the hepatocyte growth factor/c-Met signaling pathway for cancer treatment, including critical issues in clinical development and future perspectives for antibody-based therapeutics.
Collapse
Affiliation(s)
- Dongheon Lee
- Hanwha Chemical R&D Center, Biologics Business Unit, Daejeon, Republic of Korea
| | - Eun-Sil Sung
- Hanwha Chemical R&D Center, Biologics Business Unit, Daejeon, Republic of Korea
| | - Jin-Hyung Ahn
- Hanwha Chemical R&D Center, Biologics Business Unit, Daejeon, Republic of Korea
| | - Sungwon An
- Hanwha Chemical R&D Center, Biologics Business Unit, Daejeon, Republic of Korea
| | - Jiwon Huh
- Hanwha Chemical R&D Center, Biologics Business Unit, Daejeon, Republic of Korea
| | - Weon-Kyoo You
- Hanwha Chemical R&D Center, Biologics Business Unit, Daejeon, Republic of Korea
| |
Collapse
|
32
|
Abstract
Despite the enormously important and gratifying advances in cancer treatment outcomes for children with cancer, cancer remains the biggest cause of death from disease in children. Because the etiology and biology of cancers that occur in children differ dramatically from those that occur in adults, the immediate extrapolation of efficacy and safety of new cancer drugs to childhood cancer indications is not possible. We discuss factors that will play key roles in guiding pediatric oncologists as they select lines of research to pursue in their quest for more effective treatments for children with cancer.
Collapse
|
33
|
Schuck E, Bohnert T, Chakravarty A, Damian-Iordache V, Gibson C, Hsu CP, Heimbach T, Krishnatry AS, Liederer BM, Lin J, Maurer T, Mettetal JT, Mudra DR, Nijsen MJ, Raybon J, Schroeder P, Schuck V, Suryawanshi S, Su Y, Trapa P, Tsai A, Vakilynejad M, Wang S, Wong H. Preclinical pharmacokinetic/pharmacodynamic modeling and simulation in the pharmaceutical industry: an IQ consortium survey examining the current landscape. AAPS JOURNAL 2015; 17:462-73. [PMID: 25630504 DOI: 10.1208/s12248-014-9716-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 12/26/2014] [Indexed: 11/30/2022]
Abstract
The application of modeling and simulation techniques is increasingly common in preclinical stages of the drug discovery and development process. A survey focusing on preclinical pharmacokinetic/pharmacodynamics (PK/PD) analysis was conducted across pharmaceutical companies that are members of the International Consortium for Quality and Innovation in Pharmaceutical Development. Based on survey responses, ~68% of companies use preclinical PK/PD analysis in all therapeutic areas indicating its broad application. An important goal of preclinical PK/PD analysis in all pharmaceutical companies is for the selection/optimization of doses and/or dose regimens, including prediction of human efficacious doses. Oncology was the therapeutic area with the most PK/PD analysis support and where it showed the most impact. Consistent use of more complex systems pharmacology models and hybrid physiologically based pharmacokinetic models with PK/PD components was less common compared to traditional PK/PD models. Preclinical PK/PD analysis is increasingly being included in regulatory submissions with ~73% of companies including these data to some degree. Most companies (~86%) have seen impact of preclinical PK/PD analyses in drug development. Finally, ~59% of pharmaceutical companies have plans to expand their PK/PD modeling groups over the next 2 years indicating continued growth. The growth of preclinical PK/PD modeling groups in pharmaceutical industry is necessary to establish required resources and skills to further expand use of preclinical PK/PD modeling in a meaningful and impactful manner.
Collapse
Affiliation(s)
- Edgar Schuck
- Modeling and Simulation, Eisai Inc., 155 Tice Blvd, Woodcliff Lake, NJ, 07677, USA,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Mould DR, Walz AC, Lave T, Gibbs JP, Frame B. Developing Exposure/Response Models for Anticancer Drug Treatment: Special Considerations. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2015. [PMID: 26225225 PMCID: PMC4369756 DOI: 10.1002/psp4.16] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Anticancer agents often have a narrow therapeutic index (TI), requiring precise dosing to ensure sufficient exposure for clinical activity while minimizing toxicity. These agents frequently have complex pharmacology, and combination therapy may cause schedule-specific effects and interactions. We review anticancer drug development, showing how integration of modeling and simulation throughout development can inform anticancer dose selection, potentially improving the late-phase success rate. This article has a companion article in Clinical Pharmacology & Therapeutics with practical examples.
Collapse
Affiliation(s)
- D R Mould
- Projections Research Phoenixville, Pennsylvania, USA
| | - A-C Walz
- Roche Pharma Research and Early Development, Modeling & Simulation, Pharmaceutical Sciences, Roche Innovation Center Basel F. Hoffmann-La Roche, Basel, Switzerland
| | - T Lave
- Roche Pharma Research and Early Development, Modeling & Simulation, Pharmaceutical Sciences, Roche Innovation Center Basel F. Hoffmann-La Roche, Basel, Switzerland
| | - J P Gibbs
- Amgen Thousand Oaks, California, USA
| | - B Frame
- Projections Research Phoenixville, Pennsylvania, USA
| |
Collapse
|
35
|
Yamazaki S, Lam JL, Zou HY, Wang H, Smeal T, Vicini P. Mechanistic understanding of translational pharmacokinetic-pharmacodynamic relationships in nonclinical tumor models: a case study of orally available novel inhibitors of anaplastic lymphoma kinase. Drug Metab Dispos 2015; 43:54-62. [PMID: 25349124 DOI: 10.1124/dmd.114.061143] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The orally available novel small molecules PF06463922 [(10R)-7-amino-12-fluoro-2,10,16-trimethyl-15-oxo-10,15,16,17-tetrahydro-2H-8,4-(metheno)pyrazolo[4,3-h][2,5,11]benzoxadiazacyclotetradecine-3-carbonitrile] and PF06471402 [(10R)-7-amino-12-fluoro-2,10,16-trimethyl-15-oxo-10,15,16,17-tetrahydro-2H-8,4-(azeno)pyrazolo[4,3-h][2,5,11]benzoxadiazacyclo-tetradecine-3-carbonitrile] are second-generation anaplastic lymphoma kinase (ALK) inhibitors targeted to both naïve and resistant patients with non-small cell lung cancer (NSCLC) to the first-generation ALK inhibitor crizotinib. The objectives of the present study were to characterize and compare the pharmacokinetic-pharmacodynamic (PKPD) relationships of PF06463922 and PF06471402 for target modulation in tumor and antitumor efficacy in athymic mice implanted with H3122 NSCLC cells expressing a crizotinib-resistant echinoderm microtubule-associated protein-like 4 (EML4)-ALK mutation, EML4-ALK(L1196M). Furthermore, the PKPD relationships for these ALK inhibitors were evaluated and compared between oral administration and subcutaneous constant infusion (i.e., between different pharmacokinetic [PK] profiles). Oral and subcutaneous PK profiles of these ALK inhibitors were adequately described by a one-compartment PK model. An indirect response model extended with a modulator fit the time courses of PF06463922- and PF06471402-mediated target modulation (i.e., ALK phosphorylation) with an estimated unbound EC50,in vivo of 36 and 20 nM, respectively, for oral administration, and 100 and 69 nM, respectively, for subcutaneous infusion. A drug-disease model based on the turnover concept fit tumor growth curves inhibited by PF06463922 and PF06471402 with estimated unbound tumor stasis concentrations of 51 and 27 nM, respectively, for oral administration, and 116 and 70 nM, respectively, for subcutaneous infusion. Thus, the EC50,in vivo to EC60,in vivo estimates for ALK inhibition corresponded to the concentrations required tumor stasis in all cases, suggesting that the pharmacodynamic relationships of target modulation to antitumor efficacy were consistent among the ALK inhibitors, even when the PK profiles with different administration routes were considerably different.
Collapse
Affiliation(s)
- Shinji Yamazaki
- Pharmacokinetics, Dynamics and Metabolism (S.Y., J.L.L., P.V.) and Oncology Research Unit (H.Y.Z., H.W., T.S.), Pfizer Worldwide Research & Development, San Diego, California
| | - Justine L Lam
- Pharmacokinetics, Dynamics and Metabolism (S.Y., J.L.L., P.V.) and Oncology Research Unit (H.Y.Z., H.W., T.S.), Pfizer Worldwide Research & Development, San Diego, California
| | - Helen Y Zou
- Pharmacokinetics, Dynamics and Metabolism (S.Y., J.L.L., P.V.) and Oncology Research Unit (H.Y.Z., H.W., T.S.), Pfizer Worldwide Research & Development, San Diego, California
| | - Hui Wang
- Pharmacokinetics, Dynamics and Metabolism (S.Y., J.L.L., P.V.) and Oncology Research Unit (H.Y.Z., H.W., T.S.), Pfizer Worldwide Research & Development, San Diego, California
| | - Tod Smeal
- Pharmacokinetics, Dynamics and Metabolism (S.Y., J.L.L., P.V.) and Oncology Research Unit (H.Y.Z., H.W., T.S.), Pfizer Worldwide Research & Development, San Diego, California
| | - Paolo Vicini
- Pharmacokinetics, Dynamics and Metabolism (S.Y., J.L.L., P.V.) and Oncology Research Unit (H.Y.Z., H.W., T.S.), Pfizer Worldwide Research & Development, San Diego, California
| |
Collapse
|
36
|
Ilie M, Nunes M, Blot L, Hofman V, Long-Mira E, Butori C, Selva E, Merino-Trigo A, Vénissac N, Mouroux J, Vrignaud P, Hofman P. Setting up a wide panel of patient-derived tumor xenografts of non-small cell lung cancer by improving the preanalytical steps. Cancer Med 2014; 4:201-11. [PMID: 25470237 PMCID: PMC4329004 DOI: 10.1002/cam4.357] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 08/26/2014] [Accepted: 08/31/2014] [Indexed: 12/30/2022] Open
Abstract
With the ongoing need to improve therapy for non–small cell lung cancer (NSCLC) there has been increasing interest in developing reliable preclinical models to test novel therapeutics. Patient-derived tumor xenografts (PDX) are considered to be interesting candidates. However, the establishment of such model systems requires highly specialized research facilities and introduces logistic challenges. We aimed to establish an extensive well-characterized panel of NSCLC xenograft models in the context of a long-distance research network after careful control of the preanalytical steps. One hundred fresh surgically resected NSCLC specimens were shipped in survival medium at room temperature from a hospital-integrated biobank to animal facilities. Within 24 h post-surgery, tumor fragments were subcutaneously xenografted into immunodeficient mice. PDX characterization was performed by histopathological, immunohistochemical, aCGH and next-generation sequencing approaches. For this model system, the tumor take rate was 35%, with higher rates for squamous carcinoma (60%) than for adenocarcinoma (13%). Patients for whom PDX tumors were obtained had a significantly shorter disease-free survival (DFS) compared to patients for whom no PDX tumors (P = 0.039) were obtained. We established a large panel of PDX NSCLC models with a high frequency of mutations (29%) in EGFR, KRAS, NRAS, MEK1, BRAF, PTEN, and PI3KCA genes and with gene amplification (20%) of c-MET and FGFR1. This new patient-derived NSCLC xenograft collection, established regardless of the considerable time required and the distance between the clinic and the animal facilities, recapitulated the histopathology and molecular diversity of NSCLC and provides stable and reliable preclinical models for human lung cancer research.
Collapse
Affiliation(s)
- Marius Ilie
- Laboratory of Clinical and Experimental Pathology, Louis Pasteur Hospital, Nice, France; IRCAN Team 3, Inserm U1081/UMR CNRS 7284, Faculty of Medicine of Nice, University of Nice Sophia Antipolis, Nice, France; Faculty of Medicine, University of Nice Sophia-Antipolis, Nice, France; Hospital Related Biobank BB-0033-00025, Louis Pasteur Hospital, Nice, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
|
38
|
Yamazaki S, Lam JL, Zou HY, Wang H, Smeal T, Vicini P. Translational pharmacokinetic-pharmacodynamic modeling for an orally available novel inhibitor of anaplastic lymphoma kinase and c-Ros oncogene 1. J Pharmacol Exp Ther 2014; 351:67-76. [PMID: 25073473 DOI: 10.1124/jpet.114.217141] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
An orally available macrocyclic small molecule, PF06463922 [(10R)-7-amino-12-fluoro-2,10,16-trimethyl-15-oxo-10,15,16,17-tetrahydro-2H-8,4-(metheno)pyrazolo[4,3-h][2,5,11]benzoxadiazacyclotetradecine-3-carbonitrile], is a selective inhibitor of anaplastic lymphoma kinase (ALK) and c-Ros oncogene 1 (ROS1). The objectives of the present study were to characterize the pharmacokinetic-pharmacodynamic relationships of PF06463922 between its systemic exposures, pharmacodynamic biomarker (target modulation), and pharmacologic response (antitumor efficacy) in athymic mice implanted with H3122 non-small cell lung carcinomas expressing echinoderm microtubule-associated protein-like 4 (EML4)-ALK mutation (EML4-ALK(L1196M)) and with NIH3T3 cells expressing CD74-ROS1. In these nonclinical tumor models, PF06463922 was orally administered to animals with EML4-ALK(L1196M) and CD74-ROS1 at twice daily doses of 0.3-20 and 0.01-3 mg/kg per dose, respectively. Plasma concentration-time profiles of PF06463922 were adequately described by a one-compartment pharmacokinetic model. Using the model-simulated plasma concentrations, a pharmacodynamic indirect response model with a modulator sufficiently fit the time courses of target modulation (i.e., ALK phosphorylation) in tumors of EML4-ALK(L1196M)-driven models with EC50,in vivo of 36 nM free. A drug-disease model based on an indirect response model reasonably fit individual tumor growth curves in both EML4-ALK(L1196M)- and CD74-ROS1-driven models with the estimated tumor stasis concentrations of 51 and 6.2 nM free, respectively. Thus, the EC60,in vivo (52 nM free) for ALK inhibition roughly corresponded to the tumor stasis concentration in an EML4-ALK(L1196M)-driven model, suggesting that 60% ALK inhibition would be required for tumor stasis. Accordingly, we proposed that the EC60,in vivo for ALK inhibition corresponding to the tumor stasis could be considered a minimum target efficacious concentration of PF06463922 for cancer patients in a phase I trial.
Collapse
Affiliation(s)
- Shinji Yamazaki
- Pharmacokinetics, Dynamics and Metabolism (S.Y., J.L.L., P.V.) and Oncology Research Unit (H.Y.Z., H.W., T.S.), Pfizer Worldwide Research & Development, San Diego, California
| | - Justine L Lam
- Pharmacokinetics, Dynamics and Metabolism (S.Y., J.L.L., P.V.) and Oncology Research Unit (H.Y.Z., H.W., T.S.), Pfizer Worldwide Research & Development, San Diego, California
| | - Helen Y Zou
- Pharmacokinetics, Dynamics and Metabolism (S.Y., J.L.L., P.V.) and Oncology Research Unit (H.Y.Z., H.W., T.S.), Pfizer Worldwide Research & Development, San Diego, California
| | - Hui Wang
- Pharmacokinetics, Dynamics and Metabolism (S.Y., J.L.L., P.V.) and Oncology Research Unit (H.Y.Z., H.W., T.S.), Pfizer Worldwide Research & Development, San Diego, California
| | - Tod Smeal
- Pharmacokinetics, Dynamics and Metabolism (S.Y., J.L.L., P.V.) and Oncology Research Unit (H.Y.Z., H.W., T.S.), Pfizer Worldwide Research & Development, San Diego, California
| | - Paolo Vicini
- Pharmacokinetics, Dynamics and Metabolism (S.Y., J.L.L., P.V.) and Oncology Research Unit (H.Y.Z., H.W., T.S.), Pfizer Worldwide Research & Development, San Diego, California
| |
Collapse
|
39
|
Crizotinib: a review of its use in the treatment of anaplastic lymphoma kinase-positive, advanced non-small cell lung cancer. Drugs 2014; 73:2031-51. [PMID: 24288180 DOI: 10.1007/s40265-013-0142-z] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Crizotinib (Xalkori(®)) is an orally active, small molecule inhibitor of multiple receptor tyrosine kinases, including anaplastic lymphoma kinase (ALK), c-Met/hepatocyte growth factor receptor and c-ros oncogene 1. In the EU, crizotinib has been conditionally approved for the treatment of adults with previously treated, ALK-positive, advanced non-small cell lung cancer (NSCLC). This approval has been based on objective response rate and tolerability data from two ongoing phase I/II studies (PROFILE 1001 and PROFILE 1005); these results have been substantiated and extended by findings from an ongoing phase III study (PROFILE 1007) in patients with ALK-positive, advanced NSCLC who had received one prior platinum-based regimen. Those treated with crizotinib experienced significant improvements in progression-free survival, objective response rate, lung cancer symptoms and global quality of life, as compared with those treated with standard second-line chemotherapy (pemetrexed or docetaxel). The relative survival benefit with crizotinib is unclear, however, as the data are still immature and likely to be confounded by the high cross-over rate among chemotherapy recipients. Crizotinib treatment was generally well tolerated in the three PROFILE studies, with liver transaminase elevations and neutropenia being the most common grade 3 or 4 adverse events. Crizotinib is the standard of care in terms of the treatment of patients with ALK-positive, advanced NSCLC; while the current EU approval is for second (or subsequent)-line use only, the first-line use of the drug is being evaluated in ongoing phase III studies. Key issues relating to the use of crizotinib in clinical practice include identifying the small subset of eligible patients, the almost inevitable development of resistance and the high cost of treatment.
Collapse
|
40
|
Johnson TR, Tan W, Goulet L, Smith EB, Yamazaki S, Walker GS, O’Gorman MT, Bedarida G, Zou HY, Christensen JG, Nguyen LN, Shen Z, Dalvie D, Bello A, Smith BJ. Metabolism, excretion and pharmacokinetics of [14C]crizotinib following oral administration to healthy subjects. Xenobiotica 2014; 45:45-59. [DOI: 10.3109/00498254.2014.941964] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
41
|
Roberts MS, Turner DC, Broniscer A, Stewart CF. Determination of crizotinib in human and mouse plasma by liquid chromatography electrospray ionization-tandem mass spectrometry (LC-ESI-MS/MS). J Chromatogr B Analyt Technol Biomed Life Sci 2014; 960:151-7. [PMID: 24811158 DOI: 10.1016/j.jchromb.2014.04.035] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 04/21/2014] [Accepted: 04/22/2014] [Indexed: 11/19/2022]
Abstract
An LC-ESI-MS/MS method using high-throughput solid-phase extraction (SPE) was developed and validated to measure crizotinib in human and mouse plasma to support ongoing clinical and preclinical pharmacokinetic studies. Chromatographic separation of mouse or human plasma extracts was performed on a Supelco Discovery c18 column (50 mm × 2.1mm, 5.0 μ) with gradient elution using a combination of acidified aqueous and methanol (MeOH) mobile phases. The mass-to-charge transition monitored for detection and quantitation of crizotinib was m/z 450.2>260.2 while the stable label internal standard (ISTD) was monitored at m/z 457.2>267.3. The validation studies demonstrated that the assay is both precise and accurate with %CV<9% and accuracies within 8% of nominal target concentration across all concentrations tested for both the human and mouse plasma matrices. Sample volumes required for analysis were 50 and 25 μL for human plasma and mouse plasma, respectively. Calibration curves were linear over a range of 5-5,000 ng/mL for human plasma and 2-2,000 ng/mL for mouse plasma. The use of a 96-well plate format enabled rapid extraction as well as compatibility with automated workflows. The method was successfully applied to analyze crizotinib concentrations in plasma samples collected from children enrolled on a phase I pediatric study investigating the use of crizotinib for treatment of pediatric brain tumors.
Collapse
Affiliation(s)
- Michael S Roberts
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - David C Turner
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Alberto Broniscer
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Pediatrics, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
| | - Clinton F Stewart
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
42
|
Clinical pharmacokinetics of tyrosine kinase inhibitors: implications for therapeutic drug monitoring. Ther Drug Monit 2014; 35:562-87. [PMID: 24052062 DOI: 10.1097/ftd.0b013e318292b931] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The treatment of many malignancies has been improved in recent years by the introduction of molecular targeted therapies. These drugs interact preferentially with specific targets that are mutated and/or overexpressed in malignant cells. A group of such targets are the tyrosine kinases, against which a number of inhibitors (tyrosine kinase inhibitors, TKIs) have been developed. Imatinib, a TKI with targets that include the breakpoint cluster region-Abelson (bcr-abl) fusion protein kinase and mast/stem cell growth factor receptor kinase (c-Kit), was the first clinically successful drug of this type and revolutionized the treatment and prognosis of chronic myeloid leukemia and gastrointestinal stromal tumors. This success paved the way for the development of other TKIs for the treatment of a range of hematological malignancies and solid tumors. To date, 14 TKIs have been approved for clinical use and many more are under investigation. All these agents are given orally and are substrates of a range of drug transporters and metabolizing enzymes. In addition, some TKIs are capable of inhibiting their own transporters and metabolizing enzymes, making their disposition and metabolism at steady-state unpredictable. A given dose can therefore give rise to markedly different plasma concentrations in different patients, favoring the selection of resistant clones in the case of subtherapeutic exposure, and increasing the risk of toxicity if dosage is excessive. The aim of this review was to summarize current knowledge of the clinical pharmacokinetics and known adverse effects of the TKIs that are available for clinical use and to provide practical guidance on the implications of these data in patient management, in particular with respect to therapeutic drug monitoring.
Collapse
|
43
|
Practical Guidelines for Therapeutic Drug Monitoring of Anticancer Tyrosine Kinase Inhibitors: Focus on the Pharmacokinetic Targets. Clin Pharmacokinet 2014; 53:305-25. [DOI: 10.1007/s40262-014-0137-2] [Citation(s) in RCA: 137] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
44
|
Maroun CR, Rowlands T. The Met receptor tyrosine kinase: a key player in oncogenesis and drug resistance. Pharmacol Ther 2013; 142:316-38. [PMID: 24384534 DOI: 10.1016/j.pharmthera.2013.12.014] [Citation(s) in RCA: 164] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 12/12/2013] [Indexed: 12/14/2022]
Abstract
The Met receptor tyrosine kinase (RTK) is an attractive oncology therapeutic target. Met and its ligand, HGF, play a central role in signaling pathways that are exploited during the oncogenic process, including regulation of cell proliferation, invasion, angiogenesis, and cancer stem cell regulation. Elevated Met and HGF as well as numerous Met genetic alterations have been reported in human cancers and correlate with poor outcome. Alterations of pathways that regulate Met, such as the ubiquitin ligase c-Cbl are also likely to activate Met in the oncogenic setting. Moreover, interactive crosstalk between Met and other receptors such as EGFR, HER2 and VEGFR, underlies a key role for Met in resistance to other RTK-targeted therapies. A large body of preclinical and clinical data exists that supports the use of either antibodies or small molecule inhibitors that target Met or HGF as oncology therapeutics. The prognostic potential of Met expression has been suggested from studies in numerous cancers including lung, renal, liver, head and neck, stomach, and breast. Clinical trials using Met inhibitors indicate that the level of Met expression is a determinant of trial outcome, a finding that is actively under investigation in multiple clinical scenarios. Research in Met prognostics and predictors of drug response is now shifting toward more sophisticated methodologies suitable for development as validated and effective biomarkers that can be partnered with therapeutics to improve patient survival.
Collapse
Affiliation(s)
- Christiane R Maroun
- Mirati Therapeutics, 7150 Frederick-Banting, Suite 200, Montreal, Quebec H4S 2A1, Canada.
| | - Tracey Rowlands
- Mirati Therapeutics, 7150 Frederick-Banting, Suite 200, Montreal, Quebec H4S 2A1, Canada
| |
Collapse
|
45
|
Wong H, Gould SE, Budha N, Darbonne WC, Kadel EE, La H, Alicke B, Halladay JS, Erickson R, Portera C, Tolcher AW, Infante JR, Mamounas M, Flygare JA, Hop CECA, Fairbrother WJ. Learning and confirming with preclinical studies: modeling and simulation in the discovery of GDC-0917, an inhibitor of apoptosis proteins antagonist. Drug Metab Dispos 2013; 41:2104-13. [PMID: 24041744 DOI: 10.1124/dmd.113.053926] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The application of modeling and simulation techniques is increasingly common in the preclinical stages of the drug development process. GDC-0917 [(S)-1-((S)-2-cyclohexyl-2-((S)-2-(methylamino)propanamido)acetyl)-N-(2-(oxazol-2-yl)-4-phenylthiazol-5-yl)pyrrolidine-2-carboxamide] is a potent second-generation antagonist of inhibitor of apoptosis (IAP) proteins that is being developed for the treatment of various cancers. GDC-0917 has low to moderate clearance in the mouse (12.0 ml/min/kg), rat (27.0 ml/min/kg), and dog (15.3 ml/min/kg), and high clearance in the monkey (67.6 ml/min/kg). Accordingly, oral bioavailability was lowest in monkeys compared with other species. Based on our experience with a prototype molecule with similar structure, in vitro-in vivo extrapolation was used to predict a moderate clearance (11.5 ml/min/kg) in humans. The predicted human volume of distribution was estimated using simple allometry at 6.69 l/kg. Translational pharmacokinetic-pharmacodynamic (PK-PD) analysis using results from MDA-MB-231-X1.1 breast cancer xenograft studies and predicted human pharmacokinetics suggests that ED50 and ED90 targets can be achieved in humans using acceptable doses (72 mg and 660 mg, respectively) and under an acceptable time frame. The relationship between GDC-0917 concentrations and pharmacodynamic response (cIAP1 degradation) was characterized using an in vitro peripheral blood mononuclear cell immunoassay. Simulations of human GDC-0917 plasma concentration-time profile and cIAP1 degradation at the 5-mg starting dose in the phase 1 clinical trial agreed well with observations. This work shows the importance of leveraging information from prototype molecules and illustrates how modeling and simulation can be used to add value to preclinical studies in the early stages of the drug development process.
Collapse
Affiliation(s)
- Harvey Wong
- Departments of Drug Metabolism and Pharmacokinetics (H.W., H.L., J.S.H., C.E.C.A.H.), Translational Oncology (S.E.G., B.A.), Clinical Pharmacokinetics (N.B.), Oncology Biomarkers (W.C.D., E.E.K.), Exploratory Clinical Development (C.P., M.M.), Safety Assessment (R.E.), Medicinal Chemistry (J.A.F.), and Early Discovery Biochemistry (W.J.F.), Genentech, Inc., South San Francisco, California; South Texas Accelerated Research Therapeutics, LLC, San Antonio, Texas (A.W.T.); and Sarah Cannon Research Institute/Tennessee Oncology, Nashville, Tennessee (J.R.I.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Warner K, Crispatzu G, Al-Ghaili N, Weit N, Florou V, You MJ, Newrzela S, Herling M. Models for mature T-cell lymphomas--a critical appraisal of experimental systems and their contribution to current T-cell tumorigenic concepts. Crit Rev Oncol Hematol 2013; 88:680-95. [PMID: 23972664 DOI: 10.1016/j.critrevonc.2013.07.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 07/18/2013] [Accepted: 07/18/2013] [Indexed: 02/03/2023] Open
Abstract
Mature T-cell lymphomas/leukemias (MTCL) have been understudied lymphoid neoplasms that currently receive growing attention. Our historically rudimentary molecular understanding and dissatisfactory interventional success in this complex and for the most part poor-prognostic group of tumors is only slightly improving. A major limiting aspect in further progress in these rare neoplasms is the lack of suitable model systems that would substantially facilitate pathogenic studies and pre-clinical drug evaluations. Such representations of MTCL have thus far not been systematically appraised. We therefore provide an overview on existing models and point out their particular advantages and limitations in the context of the specific scientific questions. After addressing issues of species-specific differences and classifications, we summarize data on MTCL cell lines of human as well as murine origin, on murine strain predispositions to MTCL, on available models of genetically engineered mice, and on transplant systems. From an in-silico meta-analysis of available primary data of gene expression profiles on human MTCL we cross-reference genes reported to transform T-cells in mice and reflect on their general vs entity-restricted relevance and on target-promoter influences. Overall, we identify the urgent need for new models of higher fidelity to human MTCL with respect to their increasingly recognized diversity and to predictions of drug response.
Collapse
Affiliation(s)
- Kathrin Warner
- Laboratory of lymphocyte signaling and oncoproteome, CECAD, Cologne University, Cologne, Germany; Senckenberg Institute of Pathology, Goethe-University, Frankfurt/M., Germany
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Cañadas I, Rojo F, Taus Á, Arpí O, Arumí-Uría M, Pijuan L, Menéndez S, Zazo S, Dómine M, Salido M, Mojal S, García de Herreros A, Rovira A, Albanell J, Arriola E. Targeting Epithelial-to-Mesenchymal Transition with Met Inhibitors Reverts Chemoresistance in Small Cell Lung Cancer. Clin Cancer Res 2013; 20:938-50. [DOI: 10.1158/1078-0432.ccr-13-1330] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
48
|
Yamazaki S. Translational pharmacokinetic-pharmacodynamic modeling from nonclinical to clinical development: a case study of anticancer drug, crizotinib. AAPS JOURNAL 2012; 15:354-66. [PMID: 23250669 DOI: 10.1208/s12248-012-9436-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2012] [Accepted: 11/01/2012] [Indexed: 02/07/2023]
Abstract
Attrition risk related to efficacy is still a major reason why new chemical entities fail in clinical trials despite recently increased understanding of translational pharmacology. Pharmacokinetic-pharmacodynamic (PKPD) analysis is key to translating in vivo drug potency from nonclinical models to patients by providing a quantitative assessment of in vivo drug potency with mechanistic insight of drug action. The pharmaceutical industry is clearly moving toward more mechanistic and quantitative PKPD modeling to have a deeper understanding of translational pharmacology. This paper summarizes an anticancer drug case study describing the translational PKPD modeling of crizotinib, an orally available, potent small molecule inhibitor of multiple tyrosine kinases including anaplastic lymphoma kinase (ALK) and mesenchymal-epithelial transition factor (MET), from nonclinical to clinical development. Overall, the PKPD relationships among crizotinib systemic exposure, ALK or MET inhibition, and tumor growth inhibition (TGI) in human tumor xenograft models were well characterized in a quantitative manner using mathematical modeling: the results suggest that 50% ALK inhibition is required for >50% TGI whereas >90% MET inhibition is required for >50% TGI. Furthermore, >75% ALK inhibition and >95% MET inhibition in patient tumors were projected by PKPD modeling during the clinically recommended dosing regimen, twice daily doses of crizotinib 250 mg (500 mg/day). These simulation results of crizotinib-mediated ALK and MET inhibition appeared consistent with the currently reported clinical responses. In summary, the present paper presents an anticancer drug example to demonstrate that quantitative PKPD modeling can be used for predictive translational pharmacology from nonclinical to clinical development.
Collapse
Affiliation(s)
- Shinji Yamazaki
- Pharmacokinetics, Dynamics and Metabolism, La Jolla Laboratories, Pfizer Worldwide Research & Development, 10777 Science Center Drive, San Diego, CA 92121, USA.
| |
Collapse
|
49
|
Tumati V, Kumar S, Yu L, Chen B, Choy H, Saha D. Effect of PF-02341066 and radiation on non-small cell lung cancer cells. Oncol Rep 2012; 29:1094-100. [PMID: 23254764 PMCID: PMC3981113 DOI: 10.3892/or.2012.2198] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Accepted: 11/05/2012] [Indexed: 01/10/2023] Open
Abstract
Recently, a fusion protein of echinoderm microtubule associated protein like-4 (EML4) and anaplastic lymphoma kinase (ALK) has been found in non-small cell lung cancer (NSCLC) patients. In addition, endogenous expression of phosphorylated c-Met was found to be increased in many invasive NSCLC cases. PF-02341066 (crizotinib) is a novel dual c-Met and EML4-ALK inhibitor, and preclinical studies have shown that treatment with ALK inhibitors leads to drastic tumor regression in xenograft models. A phase I trial of PF-02341066 yielded a 53% response rate and a disease control rate of 79%. We evaluated crizotinib as a potential radiation-sensitizing agent in multiple established NSCLC cell lines with varying expression levels of c-Met and EML4-ALK. The combined effect of ionizing radiation (IR) and PF-02341066 was determined by the surviving cell fraction, cell cycle distribution, apoptosis, DNA double-strand break repair in 5 NSCLC cell lines (A549, H460, H3122, H2228 and H1993) and in in vivo xenograft studies. Treatment of NSCLC cells with either PF-02341066 alone or PF-02341066 + IR did not significantly alter cellular radiosensitivity, DNA repair kinetics and cell cycle distribution; no significant enhancement of tumor growth delay was noted in response to the combined treatment of PF-02341066 + IR. EML4-ALK and c-Met inhibition leads to activation of parallel pathways that converge on Akt signaling which abrogates any radiation-sensitizing effect. Although PF-02341066 is an effective therapy able to suppress tumor growth in tumors that exhibit positivity for either EML4-ALK or c-Met, it did not affect the intrinsic radiation response of tumor cell lines. In the present study, we demonstrated that PF-02341066 did not enhance radiation sensitivity in a panel of NSCLC cell lines.
Collapse
Affiliation(s)
- Vasu Tumati
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | | | | | | | |
Collapse
|
50
|
Chun SG, Choe KS, Iyengar P, Yordy JS, Timmerman RD. Isolated central nervous system progression on Crizotinib: an Achilles heel of non-small cell lung cancer with EML4-ALK translocation? Cancer Biol Ther 2012; 13:1376-83. [PMID: 22986231 DOI: 10.4161/cbt.22255] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Advanced non-small lung cancer (NSCLC) remains almost uniformly lethal with marginal long-term survival despite efforts to target specific oncogenic addiction pathways that may drive these tumors with small molecularly targeted agents and biologics. The EML4-ALK fusion gene encodes a chimeric tyrosine kinase that activates the Ras signaling pathway, and this fusion protein is found in approximately 5% of NSCLC. Targeting EML4-ALK with Crizotinib in this subset of NSCLC has documented therapeutic efficacy, but the vast majority of patients eventually develop recurrent disease that is often refractory to further treatments. We present the clinicopathologic features of three patients with metastatic NSCLC harboring the EML4-ALK translocation that developed isolated central nervous system (CNS) metastases in the presence of good disease control elsewhere in the body. These cases suggest a differential response of NSCLC to Crizotinib in the brain in comparison to other sites of disease, and are consistent with a previous report of poor CNS penetration of Crizotinib. Results of ongoing clinical trials will clarify whether the CNS is a major sanctuary site for EML4-ALK positive NSCLC being treated with Crizotinib. While understanding molecular mechanisms of resistance is critical to overcome therapeutic resistance, understanding physiologic mechanisms of resistance through analyzing anatomic patterns of failure may be equally crucial to improve long-term survival for patients with EML4-ALK translocation positive NSCLC.
Collapse
Affiliation(s)
- Stephen G Chun
- Department of Radiation Oncology, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | | | | | | | | |
Collapse
|